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
|
Lucas-Herald AK, Mitchell RT. Testicular Sertoli Cell Hormones in Differences in Sex Development. Front Endocrinol (Lausanne) 2022; 13:919670. [PMID: 35909548 PMCID: PMC9329667 DOI: 10.3389/fendo.2022.919670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
The Sertoli cells of the testes play an essential role during gonadal development, in addition to supporting subsequent germ cell survival and spermatogenesis. Anti-Müllerian hormone (AMH) is a member of the TGF-β superfamily, which is secreted by immature Sertoli cells from the 8th week of fetal gestation. lnhibin B is a glycoprotein, which is produced by the Sertoli cells from early in fetal development. In people with a Difference or Disorder of Sex Development (DSD), these hormones may be useful to determine the presence of testicular tissue and potential for spermatogenesis. However, fetal Sertoli cell development and function is often dysregulated in DSD conditions and altered production of Sertoli cell hormones may be detected throughout the life course in these individuals. As such this review will consider the role of AMH and inhibin B in individuals with DSD.
Collapse
Affiliation(s)
- Angela K. Lucas-Herald
- Developmental Endocrinology Research Group, University of Glasgow, Glasgow, United Kingdom
| | - Rod T. Mitchell
- MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Department of Paediatric Endocrinology, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
| |
Collapse
|
3
|
Ademi H, Djari C, Mayère C, Neirijnck Y, Sararols P, Rands CM, Stévant I, Conne B, Nef S. Deciphering the origins and fates of steroidogenic lineages in the mouse testis. Cell Rep 2022; 39:110935. [PMID: 35705036 DOI: 10.1016/j.celrep.2022.110935] [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: 07/28/2020] [Revised: 03/22/2022] [Accepted: 05/19/2022] [Indexed: 11/03/2022] Open
Abstract
Leydig cells (LCs) are the major androgen-producing cells in the testis. They arise from steroidogenic progenitors (SPs), whose origins, maintenance, and differentiation dynamics remain largely unknown. Single-cell transcriptomics reveal that the mouse steroidogenic lineage is specified as early as embryonic day 12.5 (E12.5) and has a dual mesonephric and coelomic origin. SPs specifically express the Wnt5a gene and evolve rapidly. At E12.5 and E13.5, they give rise first to an intermediate population of pre-LCs, and finally to fetal LCs. At E16.5, SPs possess the characteristics of the dormant progenitors at the origin of adult LCs and are also transcriptionally closely related to peritubular myoid cells (PMCs). In agreement with our in silico analysis, in vivo lineage tracing indicates that Wnt5a-expressing cells are bona fide progenitors of PMCs as well as fetal and adult LCs, contributing to most of the LCs present in the fetal and adult testis.
Collapse
Affiliation(s)
- Herta Ademi
- 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
| | - Cyril Djari
- 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
| | - 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, 1211 Geneva, Switzerland
| | - Yasmine Neirijnck
- 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
| | - Pauline Sararols
- 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
| | - Chris M Rands
- 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; Swiss Institute of Bioinformatics, 1211 Geneva, Switzerland
| | - 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
| | - Béatrice Conne
- Department of Genetic Medicine and Development, 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
|
4
|
Lin L, Xu Q, Chen Q, Chen H, Ying Y, Li Z, Zhang S, Ma F, Yu Y, Ge RS. Triadimefon increases fetal Leydig cell proliferation but inhibits its differentiation of male fetuses after gestational exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 228:112942. [PMID: 34737156 DOI: 10.1016/j.ecoenv.2021.112942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 06/13/2023]
Abstract
Triadimefon is a broad-spectrum fungicide widely applied in the agriculture. It is believed to be an endocrine disruptor. Whether triadimefon can inhibit the development of fetal Leydig cells and the underlying mechanisms are unknown. Thirty-two female pregnant Sprague-Dawley rats were randomly assigned into four groups and were dosed via gavage of triadimefon (0, 25, 50, and 100 mg/kg/day) for 9 days from gestational day (GD) 12-20. Triadimefon significantly reduced serum testosterone level in male fetuses at 100 mg/kg. The double immunofluorescence staining of proliferating cell nuclear antigen (PCNA) and cytochrome P450 cholesterol side-chain cleavage (a biomarker for fetal Leydig cells) was used to measure PCNA-labeling in fetal Leydig cells. It markedly increased fetal Leydig cell number primarily via increasing single cell population and elevated the PCNA-labeling of fetal Leydig cells in male fetuses at 100 mg/kg while it induced abnormal aggregation of fetal Leydig cells. The expression levels of fetal Leydig cell genes, Lhcgr, Scarb1, Star, Cyp11a1, Hsd3b1, Cyp17a1, Hsd17b3, Insl3 and Nr5a1, were determined to explore its effects on fetal Leydig cell development. We found that triadimefon markedly down-regulated the expression of Leydig cell genes, Hsd17b3, Insl3, and Nr5a1 as low as 25 mg/kg and Scarb1 and Cyp11a1 at 100 mg/kg. It did not affect Sertoli cell number but markedly down-regulated the expression of Sertoli cell gene Amh at 50 and 100 mg/kg. Triadimefon significantly down-regulated the expression of antioxidant genes Sod1, Gpx1, and Cat at 25-100 mg/kg, suggesting that it can induce oxidative stress in fetal testis, and it reduced the phosphorylation of ERK1/2 and AKT2 at 100 mg/kg, indicating that it can inhibit the development of fetal Leydig cells. In conclusion, gestational exposure to triadimefon inhibits the development of fetal Leydig cells in male fetuses by inhibiting its differentiation.
Collapse
Affiliation(s)
- Liben Lin
- Department of Pathology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Qiang Xu
- Department of Pathology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Quanxu Chen
- Department of Pathology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Haiqiong Chen
- Department of Pediatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Yingfen Ying
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Zengqiang Li
- Department of Anesthesiology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Song Zhang
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Feifei Ma
- Department of Anesthesiology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Yige Yu
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Ren-Shan Ge
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Anesthesiology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
5
|
Jorgensen A, Svingen T, Miles H, Chetty T, Stukenborg JB, Mitchell RT. Environmental Impacts on Male Reproductive Development: Lessons from Experimental Models. Horm Res Paediatr 2021; 96:190-206. [PMID: 34607330 DOI: 10.1159/000519964] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/11/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Male reproductive development in mammals can be divided into a gonadal formation phase followed by a hormone-driven differentiation phase. Failure of these processes may result in Differences in Sex Development (DSD), which may include abnormalities of the male reproductive tract, including cryptorchidism, hypospadias, infertility, and testicular germ cell cancer (TGCC). These disorders are also considered to be part of a testicular dysgenesis syndrome (TDS) in males. Whilst DSDs are considered to result primarily from genetic abnormalities, the development of TDS disorders is frequently associated with environmental factors. SUMMARY In this review, we will discuss the development of the male reproductive system in relation to DSD and TDS. We will also describe the experimental systems, including studies involving animals and human tissues or cells that can be used to investigate the role of environmental factors in inducing male reproductive disorders. We will discuss recent studies investigating the impact of environmental chemicals (e.g., phthalates and bisphenols), lifestyle factors (e.g., smoking) and pharmaceuticals (e.g., analgesics) on foetal testis development. Finally, we will describe the evidence, involving experimental and epidemiologic approaches, for a role of environmental factors in the development of specific male reproductive disorders, including cryptorchidism, hypospadias, and TGCC. KEY MESSAGES Environmental exposures can impact the development and function of the male reproductive system in humans. Epidemiology studies and experimental approaches using human tissues are important to translate findings from animal studies and account for species differences in response to environmental exposures.
Collapse
Affiliation(s)
- Anne Jorgensen
- Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Terje Svingen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Harriet Miles
- Royal Hospital for Children and Young People, Edinburgh, UK
| | - Tarini Chetty
- Royal Hospital for Children and Young People, Edinburgh, UK
| | - Jan-Bernd Stukenborg
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Rod T Mitchell
- Royal Hospital for Children and Young People, Edinburgh, UK
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
6
|
Johansson HKL, Taxvig C, Olsen GPM, Svingen T. Effects of the Hedgehog Signaling Inhibitor Itraconazole on Developing Rat Ovaries. Toxicol Sci 2021; 182:60-69. [PMID: 33905526 PMCID: PMC8285011 DOI: 10.1093/toxsci/kfab048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Early ovary development is considered to be largely hormone independent; yet, there are associations between fetal exposure to endocrine disrupting chemicals and reproductive disorders in women. This can potentially be explained by perturbations to establishment of ovarian endocrine function rather than interference with an already established hormone system. In this study we explore if Hedgehog (HH) signaling, a central pathway for correct ovary development, can be disrupted by exposure to HH-disrupting chemicals, using the antifungal itraconazole as model compound. In the mouse Leydig cell line TM3, used as a proxy for ovarian theca cells, itraconazole exposure had a suppressing effect on genes downstream of HH signaling, such as Gli1. Exposing explanted rat ovaries (gestational day 22 or postnatal day 3) to 30 µM itraconazole for 72 h induced significant suppression of genes in the HH signaling pathway with altered Ihh, Gli1, Ptch1, and Smo expression similar to those previously observed in Ihh/Dhh knock-out mice. Exposing rat dams to 50 mg/kg bw/day in the perinatal period did not induce observable changes in the offspring's ovaries. Overall, our results suggest that HH signal disruptors may affect ovary development with potential long-term consequences for female reproductive health. However, potent HH inhibitors would likely cause severe teratogenic effects at doses lower than those causing ovarian dysgenesis, so the concern with respect to reproductive disorder is for the presence of HH disruptors at low concentration in combination with other ovary or endocrine disrupting compounds.
Collapse
Affiliation(s)
- Hanna Katarina Lilith Johansson
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Camilla Taxvig
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Gustav Peder Mohr Olsen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Terje Svingen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| |
Collapse
|
7
|
Kothandapani A, Larsen MC, Lee J, Jorgensen JS, Jefcoate CR. Distinctive functioning of STARD1 in the fetal Leydig cells compared to adult Leydig and adrenal cells. Impact of Hedgehog signaling via the primary cilium. Mol Cell Endocrinol 2021; 531:111265. [PMID: 33864885 DOI: 10.1016/j.mce.2021.111265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 10/21/2022]
Abstract
STARD1 stimulates cholesterol transfer to mitochondrial CYP11A1 for conversion to pregnenolone. A cholesterol-binding START domain is guided by an N-terminal domain in a cell selective manner. Fetal and adult Leydig cells (FLC, ALC) show distinct Stard1 regulation. sm- FISH microscopy, which resolves individual molecules of Stard1 mRNA, shows uniformly high basal expression in each FLC. In ALC, in vivo, and cultured MA-10 cells, basal Stard1 expression is minimal. PKA activates loci asynchronously, with delayed splicing/export of 3.5 kb mRNA to mitochondria. After 60 min, ALC transition to an integrated mRNA delivery to mitochondria that is seen in FLC. Sertoli cells cooperate in Stard1 stimulation in FLC by delivering DHH to the primary cilium. There PTCH, SMO and cholesterol cooperate to release GLI3 to activate the Stard1 locus, probably by directing histone changes. ALC lack cilia. PKA then primes locus activation. FLC and ALC share similar SIK/CRTC/CREB regulation characterized for adrenal cells.
Collapse
Affiliation(s)
- Anbarasi Kothandapani
- Department of Comparative Biosciences, University of Wisconsin School of Veterinary Medicine, Madison, WI, 53706, USA
| | - Michele Campaigne Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Jinwoo Lee
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Joan S Jorgensen
- Department of Comparative Biosciences, University of Wisconsin School of Veterinary Medicine, Madison, WI, 53706, USA
| | - Colin R Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA.
| |
Collapse
|
8
|
Shen YC, Shami AN, Moritz L, Larose H, Manske GL, Ma Q, Zheng X, Sukhwani M, Czerwinski M, Sultan C, Chen H, Gurczynski SJ, Spence JR, Orwig KE, Tallquist M, Li JZ, Hammoud SS. TCF21 + mesenchymal cells contribute to testis somatic cell development, homeostasis, and regeneration in mice. Nat Commun 2021; 12:3876. [PMID: 34162856 PMCID: PMC8222243 DOI: 10.1038/s41467-021-24130-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/04/2021] [Indexed: 12/14/2022] Open
Abstract
Testicular development and function rely on interactions between somatic cells and the germline, but similar to other organs, regenerative capacity declines in aging and disease. Whether the adult testis maintains a reserve progenitor population remains uncertain. Here, we characterize a recently identified mouse testis interstitial population expressing the transcription factor Tcf21. We found that TCF21lin cells are bipotential somatic progenitors present in fetal testis and ovary, maintain adult testis homeostasis during aging, and act as potential reserve somatic progenitors following injury. In vitro, TCF21lin cells are multipotent mesenchymal progenitors which form multiple somatic lineages including Leydig and myoid cells. Additionally, TCF21+ cells resemble resident fibroblast populations reported in other organs having roles in tissue homeostasis, fibrosis, and regeneration. Our findings reveal that the testis, like other organs, maintains multipotent mesenchymal progenitors that can be potentially leveraged in development of future therapies for hypoandrogenism and/or infertility.
Collapse
Affiliation(s)
- Yu-Chi Shen
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | - Lindsay Moritz
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Hailey Larose
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Gabriel L Manske
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Qianyi Ma
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Xianing Zheng
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Meena Sukhwani
- Department of Obstetrics, Gynecology and Reproductive Sciences, Integrative Systems Biology Graduate Program, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael Czerwinski
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Caleb Sultan
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Haolin Chen
- Biochemistry and Molecular Biology, Bloomberg School of Public Health, John Hopkins, USA
| | | | - Jason R Spence
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, Integrative Systems Biology Graduate Program, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michelle Tallquist
- University of Hawaii, Center for Cardiovascular Research, Honolulu, HI, USA
| | - Jun Z Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Saher Sue Hammoud
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA.
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.
- Department of Urology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
9
|
Enhanced Negative Regulation of the DHH Signaling Pathway as a Potential Mechanism of Ascrotal Testes in Laurasiatherians. Evol Biol 2021. [DOI: 10.1007/s11692-021-09542-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
10
|
Qi Q, Dong Z, Zhang N, Wang L, Shao C, Xu W. Cloning, expression and functional analysis of the desert hedgehog (dhh) gene in Chinese tongue sole (Cynoglossus semilaevis). Gene Expr Patterns 2020; 39:119163. [PMID: 33359643 DOI: 10.1016/j.gep.2020.119163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/02/2020] [Accepted: 12/18/2020] [Indexed: 12/24/2022]
Abstract
Desert hedgehog (dhh) is a gene that is crucial for spermatogenesis and Leydig cell differentiation, but little is known regarding its influence on gonadal differentiation and development in fish. To understand its function, we cloned and characterized the dhh gene from Cynoglossus semilaevis (csdhh). The full length csdhh cDNA was 2473 bp, including a 1386 bp open reading frame (ORF), a 475 bp 5'-UTR, and a 612 bp 3'-UTR, encoding a predicted protein of 461 amino acid residues. Phylogenetic analysis showed that the putative protein belongs to the hedgehog (HH) family, and contains typical HH-N and HH-C domains. Amino acid sequence analysis revealed that CsDhh shares many features with Dhh analogues in other teleost species. Real-time quantitative PCR showed that csdhh was detected in eight different tissues in male and female tongue sole. During early embryonic development, the relative expression of the csdhh was significantly higher in the neural stage than in other embryonic developmental stages (P < 0.05). csdhh was detected at 20 days after hatching (dah) and at the critical period of male gonadal differentiation (80-95 dah), the relative expression of the csdhh was significantly higher in the male gonads than the female gonads. In 5, 8, and 12 month old gonads, the relative expression of the csdhh was significantly higher in male and pseudo-male than in female fish. The in situ hybridization (ISH) results showed that the hybridization signal was strongly expressed in primary and secondary spermatocytes, spermatids, and sertoli cells of the 1-year-old fish testis, with only weak signal expression in the corresponding ovarian tissue. These results suggest that csdhh is highly conserved in evolution and plays an important role in spermatogenesis in males and pseudo-males.
Collapse
Affiliation(s)
- Qian Qi
- Henan University of Science and Technology, Luoyang, 471000, China; Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Science (CAFS), Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology Qingdao, 266071, China
| | - Zhongdian Dong
- Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Ning Zhang
- Guangdong South China Sea Key Laboratory of Aquaculture for Aquatic Economic Animals, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Liang Wang
- Yantai Marine Economic Research Institute, Yantai, 264003, China
| | - Changwei Shao
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Science (CAFS), Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology Qingdao, 266071, China
| | - Wenteng Xu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Science (CAFS), Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology Qingdao, 266071, China.
| |
Collapse
|
11
|
Gawrychowska A, Iżycka-Świeszewska E, Lipska-Ziętkiewicz BS, Kuleszo D, Bautembach-Minkowska J, Łosin M, Stefanowicz J. Dysgerminoma with a Somatic Exon 17 KIT Mutation and SHH Pathway Activation in a Girl with Turner Syndrome. Diagnostics (Basel) 2020; 10:diagnostics10121067. [PMID: 33321690 PMCID: PMC7763800 DOI: 10.3390/diagnostics10121067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/29/2020] [Accepted: 12/04/2020] [Indexed: 11/23/2022] Open
Abstract
This article reports a case of a 7-year-old girl with Turner syndrome, treated with growth hormone (GH), who developed ovarian dysgerminoma. The patient karyotype was mosaic for chromosome Xq deletion: 46,X,del(X)(q22)/45,X. No Y chromosome sequences were present. Molecular studies revealed the presence of a driving mutation in exon 17 of the KIT gene in the neoplastic tissue, as well as Sonic-hedgehog (SHH) pathway activation at the protein level. The patient responded well to chemotherapy and remained in complete remission. This is the first case of dysgerminoma in a Turner syndrome patient with such oncogenic pathway.
Collapse
Affiliation(s)
- Ada Gawrychowska
- Department of Paediatrics, Haematology and Oncology, Clinical University Centre, 7 Debinki Street, 80-952 Gdansk, Poland;
| | - Ewa Iżycka-Świeszewska
- Department of Pathology and Neuropathology, Faculty of Health Sciences, Medical University of Gdansk, 3a Maria Sklodowska-Curie Street, 80-210 Gdansk, Poland;
| | - Beata S. Lipska-Ziętkiewicz
- Centre for Rare Diseases, Medical University of Gdansk, 7 Debinki Street, 80-952 Gdansk, Poland;
- Clinical Genetics Unit, Department of Biology and Medical Genetics, Faculty of Medicine, Medical University of Gdansk, 1 Debinki Street, 80-211 Gdansk, Poland
| | - Dominika Kuleszo
- Department of Biology and Medical Genetics, Faculty of Medicine, Medical University of Gdansk, 1 Debinki Street, 80-211 Gdansk, Poland;
| | - Joanna Bautembach-Minkowska
- Department of Paediatrics, Diabetology and Endocrinology, Clinical University Centre, 7 Debinki Street, 80-952 Gdansk, Poland;
| | - Marcin Łosin
- Department of Surgery and Urology for Children and Adolescents, Faculty of Medicine, Medical University of Gdansk, 1-6 Nowe Ogrody Street, 80-803 Gdansk, Poland;
| | - Joanna Stefanowicz
- Department of Paediatrics, Haematology and Oncology, Clinical University Centre, 7 Debinki Street, 80-952 Gdansk, Poland;
- Department of Paediatrics, Haematology and Oncology, Faculty of Medicine, Medical University of Gdansk, 7 Debinki Street, 80-210 Gdansk, Poland
- Faculty of Health Sciences, Medical University of Gdansk, 3a Maria Sklodowska-Curie Street, 80-210 Gdansk, Poland
- Correspondence: ; Tel.: +48-58-349-2808
| |
Collapse
|
12
|
Disorders of Sex Development-Novel Regulators, Impacts on Fertility, and Options for Fertility Preservation. Int J Mol Sci 2020; 21:ijms21072282. [PMID: 32224856 PMCID: PMC7178030 DOI: 10.3390/ijms21072282] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/09/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022] Open
Abstract
Disorders (or differences) of sex development (DSD) are a heterogeneous group of congenital conditions with variations in chromosomal, gonadal, or anatomical sex. Impaired gonadal development is central to the pathogenesis of the majority of DSDs and therefore a clear understanding of gonadal development is essential to comprehend the impacts of these disorders on the individual, including impacts on future fertility. Gonadal development was traditionally considered to involve a primary 'male' pathway leading to testicular development as a result of expression of a small number of key testis-determining genes. However, it is increasingly recognized that there are several gene networks involved in the development of the bipotential gonad towards either a testicular or ovarian fate. This includes genes that act antagonistically to regulate gonadal development. This review will highlight some of the novel regulators of gonadal development and how the identification of these has enhanced understanding of gonadal development and the pathogenesis of DSD. We will also describe the impact of DSDs on fertility and options for fertility preservation in this context.
Collapse
|
13
|
Mäkelä JA, Koskenniemi JJ, Virtanen HE, Toppari J. Testis Development. Endocr Rev 2019; 40:857-905. [PMID: 30590466 DOI: 10.1210/er.2018-00140] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/17/2018] [Indexed: 12/28/2022]
Abstract
Production of sperm and androgens is the main function of the testis. This depends on normal development of both testicular somatic cells and germ cells. A genetic program initiated from the Y chromosome gene sex-determining region Y (SRY) directs somatic cell specification to Sertoli cells that orchestrate further development. They first guide fetal germ cell differentiation toward spermatogenic destiny and then take care of the full service to spermatogenic cells during spermatogenesis. The number of Sertoli cells sets the limits of sperm production. Leydig cells secrete androgens that determine masculine development. Testis development does not depend on germ cells; that is, testicular somatic cells also develop in the absence of germ cells, and the testis can produce testosterone normally to induce full masculinization in these men. In contrast, spermatogenic cell development is totally dependent on somatic cells. We herein review germ cell differentiation from primordial germ cells to spermatogonia and development of the supporting somatic cells. Testicular descent to scrota is necessary for normal spermatogenesis, and cryptorchidism is the most common male birth defect. This is a mild form of a disorder of sex differentiation. Multiple genetic reasons for more severe forms of disorders of sex differentiation have been revealed during the last decades, and these are described along with the description of molecular regulation of testis development.
Collapse
Affiliation(s)
- Juho-Antti Mäkelä
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jaakko J Koskenniemi
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Helena E Virtanen
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland.,Department of Pediatrics, Turku University Hospital, Turku, Finland
| |
Collapse
|
14
|
Shimizu N, Matsuda M. Identification of a Novel Zebrafish Mutant Line that Develops Testicular Germ Cell Tumors. Zebrafish 2018; 16:15-28. [PMID: 30300574 DOI: 10.1089/zeb.2018.1604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Testicular tumors are the most common solid malignant tumors in men 20-35 years of age. Although most of testicular tumors are curable, current treatments still fail in 15%-20% of patients. However, insufficient understanding of the molecular basis and lack of animal models limit development of more effective treatments. This study reports the identification of a novel zebrafish mutant line, ns1402, which develops testicular germ cell tumors (TGCTs). While both male and female ns1402 mutants were fertile at young age, male ns1402 mutants became infertile as early as 9 months of age. This infertility was associated with progressive loss of mature sperm. Failure of spermatogenesis was, at least in part, explained by progressive loss of mature Leydig cells, a source of testosterone that is essential for spermatogenesis. Interestingly, TGCTs in ns1402 mutants contained a large number of Sertoli cells and gene expression profiles of Sertoli cells were altered before loss of mature Leydig cells. This suggests that changes in Sertoli cell properties happened first, followed by loss of mature Leydig cells and failure of spermatogenesis. Taken together, this study emphasizes the importance of cell-cell interactions and cell signaling in the testis for spermatogenesis and tissue homeostasis.
Collapse
Affiliation(s)
- Nobuyuki Shimizu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Miho Matsuda
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey
| |
Collapse
|
15
|
Rotgers E, Jørgensen A, Yao HHC. At the Crossroads of Fate-Somatic Cell Lineage Specification in the Fetal Gonad. Endocr Rev 2018; 39:739-759. [PMID: 29771299 PMCID: PMC6173476 DOI: 10.1210/er.2018-00010] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/09/2018] [Indexed: 01/07/2023]
Abstract
The reproductive endocrine systems are vastly different between males and females. This sexual dimorphism of the endocrine milieu originates from sex-specific differentiation of the somatic cells in the gonads during fetal life. Most gonadal somatic cells arise from the adrenogonadal primordium. After separation of the adrenal and gonadal primordia, the gonadal somatic cells initiate sex-specific differentiation during gonadal sex determination with the specification of the supporting cell lineages: Sertoli cells in the testis vs granulosa cells in the ovary. The supporting cell lineages then facilitate the differentiation of the steroidogenic cell lineages, Leydig cells in the testis and theca cells in the ovary. Proper differentiation of these cell types defines the somatic cell environment that is essential for germ cell development, hormone production, and establishment of the reproductive tracts. Impairment of lineage specification and function of gonadal somatic cells can lead to disorders of sexual development (DSDs) in humans. Human DSDs and processes for gonadal development have been successfully modeled using genetically modified mouse models. In this review, we focus on the fate decision processes from the initial stage of formation of the adrenogonadal primordium in the embryo to the maintenance of the somatic cell identities in the gonads when they become fully differentiated in adulthood.
Collapse
Affiliation(s)
- Emmi Rotgers
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Durham, North Carolina
| | - Anne Jørgensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,International Research and Research Training Center in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen, Denmark
| | - Humphrey Hung-Chang Yao
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Durham, North Carolina
| |
Collapse
|
16
|
Liu C, Rodriguez KF, Brown PR, Yao HHC. Reproductive, Physiological, and Molecular Outcomes in Female Mice Deficient in Dhh and Ihh. Endocrinology 2018; 159:2563-2575. [PMID: 29788357 PMCID: PMC6287595 DOI: 10.1210/en.2018-00095] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/11/2018] [Indexed: 02/05/2023]
Abstract
Ovarian development requires coordinate communications among oocytes, granulosa cells, and theca cells. Two Hedgehog (Hh) pathway ligands, Desert hedgehog (Dhh) and Indian hedgehog (Ihh), are produced by the granulosa cells and work together to regulate theca cell specification and development. Mice lacking both Dhh and Ihh had loss of normal ovarian function, which raised the question of which biological actions are specifically controlled by each ligand during folliculogenesis. By comparing the reproductive fitness, hormonal profiles, and ovarian transcriptomes among control, Dhh single-knockout (KO), Ihh KO, and Dhh/Ihh double-knockout (DKO) mice, we examined the specific roles of Dhh and Ihh in these processes. Dhh/Ihh DKO female mice were infertile because of a lack of theca cells and their steroid product androgen. Although Dhh and Ihh KO mice were fertile with normal folliculogenesis, they had decreased androgen production and alterations in their ovarian transcriptomes. Absence of Ihh led to aberrant steroidogenesis and elevated inflammation responses, which were not found in Dhh KO mouse ovaries, implicating that IHH has a greater impact than DHH on the activation of the Hh signaling pathway in the ovary. Our findings provide insight into not only how the Hh pathway influences folliculogenesis but also the distinct and overlapping roles of Dhh and Ihh in supporting ovarian development.
Collapse
Affiliation(s)
- Chang Liu
- Reproductive and Developmental Biology Group, National Institute of
Environmental Health Sciences, Durham, North Carolina
| | - Karina F Rodriguez
- Reproductive and Developmental Biology Group, National Institute of
Environmental Health Sciences, Durham, North Carolina
| | - Paula R Brown
- Reproductive and Developmental Biology Group, National Institute of
Environmental Health Sciences, Durham, North Carolina
| | - Humphrey H-C Yao
- Reproductive and Developmental Biology Group, National Institute of
Environmental Health Sciences, Durham, North Carolina
- Correspondence: Humphrey H.-C. Yao, PhD, Reproductive Developmental Biology Laboratory, National
Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, Mail Drop C4-10,
Research Triangle Park, North Carolina 27709. E-mail:
| |
Collapse
|
17
|
Richards JS, Ren YA, Candelaria N, Adams JE, Rajkovic A. Ovarian Follicular Theca Cell Recruitment, Differentiation, and Impact on Fertility: 2017 Update. Endocr Rev 2018; 39:1-20. [PMID: 29028960 PMCID: PMC5807095 DOI: 10.1210/er.2017-00164] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/12/2017] [Indexed: 12/24/2022]
Abstract
The major goal of this review is to summarize recent exciting findings that have been published within the past 10 years that, to our knowledge, have not been presented in detail in previous reviews and that may impact altered follicular development in polycystic ovarian syndrome (PCOS) and premature ovarian failure in women. Specifically, we will cover the following: (1) mouse models that have led to discovery of the derivation of two precursor populations of theca cells in the embryonic gonad; (2) the key roles of the oocyte-derived factor growth differentiation factor 9 on the hedgehog (HH) signaling pathway and theca cell functions; and (3) the impact of the HH pathway on both the specification of theca endocrine cells and theca fibroblast and smooth muscle cells in developing follicles. We will also discuss the following: (1) other signaling pathways that impact the differentiation of theca cells, not only luteinizing hormone but also insulinlike 3, bone morphogenic proteins, the circadian clock genes, androgens, and estrogens; and (2) theca-associated vascular, immune, and fibroblast cells, as well as the cytokines and matrix factors that play key roles in follicle growth. Lastly, we will integrate what is known about theca cells from mouse models, human-derived theca cell lines from patients who have PCOS and patients who do not have PCOS, and microarray analyses of human and bovine theca to understand what pathways and factors contribute to follicle growth as well as to the abnormal function of theca.
Collapse
Affiliation(s)
- JoAnne S. Richards
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Yi A. Ren
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Nicholes Candelaria
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Jaye E. Adams
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Aleksandar Rajkovic
- Department of Obstetrics, Gynecology and Reproductive Medicine, Magee-Women’s Research Institute, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
18
|
Role of the Hedgehog Signaling Pathway in Regulating the Behavior of Germline Stem Cells. Stem Cells Int 2017; 2017:5714608. [PMID: 28883837 PMCID: PMC5572616 DOI: 10.1155/2017/5714608] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/11/2017] [Accepted: 05/21/2017] [Indexed: 12/24/2022] Open
Abstract
Germline stem cells (GSCs) are adult stem cells that are responsible for the production of gametes and include spermatogonial stem cells (SSCs) and ovarian germline stem cells (OGSCs). GSCs are located in a specialized microenvironment in the gonads called the niche. Many recent studies have demonstrated that multiple signals in the niche jointly regulate the proliferation and differentiation of GSCs, which is of significance for reproductive function. Previous studies have demonstrated that the hedgehog (Hh) signaling pathway participates in the proliferation and differentiation of various stem cells, including GSCs in Drosophila and male mammals. Furthermore, the discovery of mammalian OGSCs challenged the traditional opinion that the number of primary follicles is fixed in postnatal mammals, which is of significance for the reproductive ability of female mammals and the treatment of diseases related to germ cells. Meanwhile, it still remains to be determined whether the Hh signaling pathway participates in the regulation of the behavior of OGSCs. Herein, we review the current research on the role of the Hh signaling pathway in mediating the behavior of GSCs. In addition, some suggestions for future research are proposed.
Collapse
|
19
|
Rudigier LJ, Dame C, Scholz H, Kirschner KM. Ex vivo cultures combined with vivo-morpholino induced gene knockdown provide a system to assess the role of WT1 and GATA4 during gonad differentiation. PLoS One 2017; 12:e0176296. [PMID: 28426816 PMCID: PMC5398674 DOI: 10.1371/journal.pone.0176296] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 04/07/2017] [Indexed: 12/21/2022] Open
Abstract
Gonad morphogenesis relies on the correct spatiotemporal expression of a number of genes that together fulfill the differentiation of the bipotential gonad into testes or ovaries. As such, the transcription factors WT1 and GATA4 are pivotal for proper gonadal development. Here we address the contributions of GATA4 and WT1 to the sex differentiation phase in testes and ovaries. We applied an ex vivo technique for cultivating gonads in hanging droplets of media that were supplemented with vivo-morpholinos to knockdown WT1 and GATA4 either alone or in combination at the same developmental stage. We show that WT1 is equally important for both, the initial establishment and the maintenance of the sex-specific gene expression signature in testes and ovaries. We further identified Foxl2 as a novel putative downstream target gene of WT1. Moreover, knockdown of WT1 reduced mRNA levels of several molecular components of the hedgehog signaling pathway in XY gonads, whereas Gata4 vivo-morpholino treatment increased transcripts of Dhh and Ptch1 in embryonic testes. The data suggest that for its proper function, WT1 relies on the correct expression of the GATA4 protein. Furthermore, GATA4 down-regulates several ovarian promoting genes in testes, such as Ctnnb1, Fst, and Bmp2, suggesting that this repression is required for maintaining the male phenotype. In conclusion, this study provides novel insights into the role of WT1 and GATA4 during the sex differentiation phase and represents an approach that can be applied to assess other proteins with as yet unknown functions during gonadal development.
Collapse
Affiliation(s)
- Lucas J. Rudigier
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Christof Dame
- Klinik für Neonatologie, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Holger Scholz
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
- * E-mail:
| | - Karin M. Kirschner
- Institut für Vegetative Physiologie, Charité-Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| |
Collapse
|
20
|
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
|
21
|
Dores C, Alpaugh W, Su L, Biernaskie J, Dobrinski I. Primary cilia on porcine testicular somatic cells and their role in hedgehog signaling and tubular morphogenesis in vitro. Cell Tissue Res 2016; 368:215-223. [PMID: 27841005 DOI: 10.1007/s00441-016-2523-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 09/30/2016] [Indexed: 01/06/2023]
Abstract
The primary cilium is a microtubule-based sensory organelle found on nearly all eukaryotic cells but little is understood about its function in the testis. We investigate the role of primary cilia on testis cells in vitro by inhibiting formation of the primary cilium with Ciliobrevin D, a cell-permeable, reversible chemical inhibitor of ATPase motor cytoplasmic dynein. We analyzed cultured cells for the presence of primary cilia and their involvement in hedgehog signaling. Primary cilia were present on 89.3 ± 2.3 % of untreated testicular somatic cells compared to 3.1 ± 2.5 % cells with primary cilia for Ciliobrevin D-treated cells. Protein levels of Gli-2 and Smoothened were lower on Western blots after suppression of cilia with Ciliobrevin D. The inhibitor did not affect centrosome localization or cell proliferation, indicating that changes were due to ablation of the primary cilium. Testicular somatic cells have the ability to form three-dimensional tubules in vitro. In vitro-formed tubules were significantly longer and wider in the control group than in the Ciliobrevin D-treated group (9.91 ± 0.35 vs. 5.540 ± 1.08 mm and 339.8 ± 55.78 vs. 127.2 ± 11.9 μm, respectively) indicating that primary cilia play a role in tubule formation. Our results establish that the inhibition of ATPase motor cytoplasmic dynein perturbs formation of primary cilia in testicular somatic cells, affects the hedgehog signaling pathway and impairs tubule formation in vitro. These findings provide evidence for a role of cilia in the testis in cell signaling and tubular morphogenesis in vitro.
Collapse
Affiliation(s)
- Camila Dores
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, HMRB 404, Calgary, Alberta, T2N 4N1, Canada
| | - Whitney Alpaugh
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, HMRB 404, Calgary, Alberta, T2N 4N1, Canada
| | - Lin Su
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, HMRB 404, Calgary, Alberta, T2N 4N1, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, HMRB 404, Calgary, Alberta, T2N 4N1, Canada
| | - Ina Dobrinski
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, HMRB 404, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
22
|
Wen Q, Cheng CY, Liu YX. Development, function and fate of fetal Leydig cells. Semin Cell Dev Biol 2016; 59:89-98. [PMID: 26968934 PMCID: PMC5016207 DOI: 10.1016/j.semcdb.2016.03.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 12/21/2022]
Abstract
During fetal testis development, fetal Leydig cells (FLCs) are found to be originated from multiple progenitor cells. FLC specification and function are under tight regulation of specific genes and signaling proteins. Furthermore, Sertoli cells play a crucial role to regulate FLC differentiation during fetal testis development. FLC progenitor- and FLC-produced biomolecules are also involved in the differentiation and activity of rodent FLCs. The main function of FLCs is to produce androgens to masculinize XY embryos. However, FLCs are capable of producing androstenedione but not testosterone due to the lack of 17β-HSD (17β-hydroxysteroid dehydrogenase), but fetal Sertoli cells express 17β-HSD which thus transforms androstenedione to testosterone in the fetal testis. On the other hand, FLCs produce activin A to regulate Sertoli cell proliferation, and Sertoli cells in turn modulate testis cord expansion. It is now generally accepted that adult Leydig cells (ALCs) gradually replace FLCs during postnatal development to produce testosterone to support spermatogenesis as FLCs undergo degeneration in neonatal and pre-pubertal testes. However, based on studies using genetic tracing mouse models, FLCs are found to persist in adult testes, making up ∼20% of total Leydig cells. In this review, we evaluate the latest findings regarding the development, function and fate of FLCs during fetal and adult testis development.
Collapse
Affiliation(s)
- Qing Wen
- State Key Laboratory of Stem Cells and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York 10065, United States.
| | - Yi-Xun Liu
- State Key Laboratory of Stem Cells and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
23
|
Lubik AA, Nouri M, Truong S, Ghaffari M, Adomat HH, Corey E, Cox ME, Li N, Guns ES, Yenki P, Pham S, Buttyan R. Paracrine sonic hedgehog signaling contributes significantly to acquired steroidogenesis in the prostate tumor microenvironment. Int J Cancer 2016; 140:358-369. [PMID: 27672740 DOI: 10.1002/ijc.30450] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 09/12/2016] [Indexed: 01/02/2023]
Abstract
Despite the substantial benefit of androgen deprivation therapy (ADT) for metastatic prostate cancer, patients often progress to castration-resistant disease (CRPC) that is more difficult to treat. CRPC is associated with renewed androgen receptor activity in tumor cells and restoration of tumor androgen levels through acquired intratumoral steroidogenesis (AIS). Although prostate cancer (PCa) cells have been shown to have steroidogenic capability in vitro, we previously found that benign prostate stromal cells (PrSCs) can also synthesize testosterone (T) from an adrenal precursor, DHEA, when stimulated with a hedgehog (Hh) pathway agonist, SAG. Here, we show exposure of PrSCs to a different Smoothened (Smo) agonist, Ag1.5, or to conditioned medium from sonic hedgehog overexpressing LNCaP cells induces steroidogenic enzyme expression in PrSCs and significantly increases production of T and its precursor steroids in a Smo-dependent manner from 22-OH-cholesterol substrate. Hh agonist-/ligand-treated PrSCs produced androgens at a rate similar to or greater than that of PCa cell lines. Likewise, primary bone marrow stromal cells became more steroidogenic and produced T under the influence of Smo agonist. Treatment of mice bearing LNCaP xenografts with a Smo antagonist, TAK-441, delayed the onset of CRPC after castration and substantially reduced androgen levels in residual tumors. These outcomes support the idea that stromal cells in ADT-treated primary or metastatic prostate tumors can contribute to AIS as a consequence of a paracrine Hh signaling microenvironment. As such, Smo antagonists may be useful for targeting prostate tumor stromal cell-derived AIS and delaying the onset of CRPC after ADT.
Collapse
Affiliation(s)
- Amy A Lubik
- The Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Mannan Nouri
- The Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Sarah Truong
- The Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Mazyar Ghaffari
- The Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Hans H Adomat
- The Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA
| | - Michael E Cox
- The Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Na Li
- The Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Emma S Guns
- The Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Parvin Yenki
- The Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Steven Pham
- The Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ralph Buttyan
- The Vancouver Prostate Centre and the Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
24
|
Windley SP, Wilhelm D. Signaling Pathways Involved in Mammalian Sex Determination and Gonad Development. Sex Dev 2016; 9:297-315. [PMID: 26905731 DOI: 10.1159/000444065] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2015] [Indexed: 11/19/2022] Open
Abstract
The development of any organ system requires a complex interplay of cellular signals to initiate the differentiation and development of the heterogeneous cell and tissue types required to carry out the organs' functions. In this way, an extracellular stimulus is transmitted to an intracellular target through an array of interacting protein intermediaries, ultimately enabling the target cell to elicit a response. Surprisingly, only a small number of signaling pathways are implicated throughout embryogenesis and are used over and over again. Gonadogenesis is a unique process in that 2 morphologically distinct organs, the testes and ovaries, arise from a common precursor, the bipotential genital ridge. Accordingly, most of the signaling pathways observed throughout embryogenesis also have been shown to be important for mammalian sex determination and gonad development. Here, we review the mechanisms of signal transduction within these pathways and the importance of these pathways throughout mammalian gonad development, mainly concentrating on data obtained in mouse but including other species where appropriate.
Collapse
Affiliation(s)
- Simon P Windley
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Vic., Australia
| | | |
Collapse
|
25
|
Chen SR, Liu YX. Testis Cord Maintenance in Mouse Embryos: Genes and Signaling1. Biol Reprod 2016; 94:42. [DOI: 10.1095/biolreprod.115.137117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/12/2016] [Indexed: 12/12/2022] Open
|
26
|
Romereim SM, Cupp AS. Mesonephric Cell Migration into the Gonads and Vascularization Are Processes Crucial for Testis Development. Results Probl Cell Differ 2016; 58:67-100. [PMID: 27300176 DOI: 10.1007/978-3-319-31973-5_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Testis morphogenesis requires the integration and reorganization of multiple cell types from several sources, one of the more notable being the mesonephric-derived cell population. One of the earliest sex-specific morphogenetic events in the gonad is a wave of endothelial cell migration from the mesonephros that is crucial for (1) partitioning the gonad into domains for testis cords, (2) providing the vasculature of the testis, and (3) signaling to cells both within the gonad and beyond it to coordinately regulate testis development. In addition to endothelial cell migration, there is evidence that precursors of peritubular myoid cells migrate from the mesonephros, an event which is also important for testis cord architecture. Investigation of the mesonephric cell migration event has utilized histology, lineage tracing with mouse genetic markers, and many studies of the signaling molecules/pathways involved. Some of the more well-studied signaling molecules involved include vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), and neurotrophins. In this chapter, the morphogenetic events, relevant signaling pathways, mechanisms underlying the migration, and the role of the migratory cells within the testis will be discussed. Overall, the migration of mesonephric cells into the early testis is indispensable for its development and future functionality.
Collapse
|
27
|
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
|
28
|
Bashamboo A, McElreavey K. Human sex-determination and disorders of sex-development (DSD). Semin Cell Dev Biol 2015; 45:77-83. [DOI: 10.1016/j.semcdb.2015.10.030] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/19/2015] [Accepted: 10/19/2015] [Indexed: 11/28/2022]
|
29
|
Werner R, Merz H, Birnbaum W, Marshall L, Schröder T, Reiz B, Kavran JM, Bäumer T, Capetian P, Hiort O. 46,XY Gonadal Dysgenesis due to a Homozygous Mutation in Desert Hedgehog (DHH) Identified by Exome Sequencing. J Clin Endocrinol Metab 2015; 100:E1022-9. [PMID: 25927242 PMCID: PMC4490300 DOI: 10.1210/jc.2015-1314] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND 46,XY disorders of sex development (DSD) comprise a heterogeneous group of congenital conditions. Mutations in a variety of genes can affect gonadal development or androgen biosynthesis/action and thereby influence the development of the internal and external genital organs. OBJECTIVE The objective of the study was to identify the genetic cause in two 46,XY sisters of a consanguineous family with DSD and gonadal tumor formation. METHODS We used a next-generation sequencing approach by exome sequencing. Electrophysiological and high-resolution ultrasound examination of peripheral nerves as well as histopathological examination of the gonads were performed. RESULTS We identified a novel homozygous R124Q mutation in the desert hedgehog gene (DHH), which alters a conserved residue among the three mammalian Hedgehog ligands sonic hedgehog, Indian hedgehog, and desert hedgehog. No other relevant mutations in DSD-related genes were encountered. The gonads of one patient showed partial gonadal dysgenesis with loss of Leydig cells in tubular areas with seminoma in situ and a hyperplasia of Leydig cell-like cells expressing CYP17A1 in more dysgenetic parts of the gonad. In addition, both patients suffer from a polyneuropathy. High-resolution ultrasound revealed a structural change of peripheral nerve structure that fits well to a minifascicle formation of peripheral nerves. CONCLUSION Mutations in DHH play a role in 46,XY gonadal dysgenesis and are associated with seminoma formation and a neuropathy with minifascicle formation. Gonadal dysgenesis in these cases may be due to impairment of Sertoli cell-Leydig cell interaction during gonadal development.
Collapse
Affiliation(s)
- Ralf Werner
- Department of Paediatric and Adolescent Medicine, and Division of Experimental Paediatric Endocrinology and Diabetes (R.W., W.B., L.M., O.H.), Departments of Pathology (H.M.), Gynecology (T.S.), Neurology and Institute of Neurogenetics (P.C.), and Paediatric and Adult Movement Disorders and Neuropsychiatry and Institute of Neurogenetics (T.B.), Institute of Integrative and Experimental Genomics (B.R.), University of Luebeck, 23538 Luebeck, Germany; and Department of Biophysics and Biophysical Chemistry (J.M.K.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Hartmut Merz
- Department of Paediatric and Adolescent Medicine, and Division of Experimental Paediatric Endocrinology and Diabetes (R.W., W.B., L.M., O.H.), Departments of Pathology (H.M.), Gynecology (T.S.), Neurology and Institute of Neurogenetics (P.C.), and Paediatric and Adult Movement Disorders and Neuropsychiatry and Institute of Neurogenetics (T.B.), Institute of Integrative and Experimental Genomics (B.R.), University of Luebeck, 23538 Luebeck, Germany; and Department of Biophysics and Biophysical Chemistry (J.M.K.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Wiebke Birnbaum
- Department of Paediatric and Adolescent Medicine, and Division of Experimental Paediatric Endocrinology and Diabetes (R.W., W.B., L.M., O.H.), Departments of Pathology (H.M.), Gynecology (T.S.), Neurology and Institute of Neurogenetics (P.C.), and Paediatric and Adult Movement Disorders and Neuropsychiatry and Institute of Neurogenetics (T.B.), Institute of Integrative and Experimental Genomics (B.R.), University of Luebeck, 23538 Luebeck, Germany; and Department of Biophysics and Biophysical Chemistry (J.M.K.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Louise Marshall
- Department of Paediatric and Adolescent Medicine, and Division of Experimental Paediatric Endocrinology and Diabetes (R.W., W.B., L.M., O.H.), Departments of Pathology (H.M.), Gynecology (T.S.), Neurology and Institute of Neurogenetics (P.C.), and Paediatric and Adult Movement Disorders and Neuropsychiatry and Institute of Neurogenetics (T.B.), Institute of Integrative and Experimental Genomics (B.R.), University of Luebeck, 23538 Luebeck, Germany; and Department of Biophysics and Biophysical Chemistry (J.M.K.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Tatjana Schröder
- Department of Paediatric and Adolescent Medicine, and Division of Experimental Paediatric Endocrinology and Diabetes (R.W., W.B., L.M., O.H.), Departments of Pathology (H.M.), Gynecology (T.S.), Neurology and Institute of Neurogenetics (P.C.), and Paediatric and Adult Movement Disorders and Neuropsychiatry and Institute of Neurogenetics (T.B.), Institute of Integrative and Experimental Genomics (B.R.), University of Luebeck, 23538 Luebeck, Germany; and Department of Biophysics and Biophysical Chemistry (J.M.K.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Benedikt Reiz
- Department of Paediatric and Adolescent Medicine, and Division of Experimental Paediatric Endocrinology and Diabetes (R.W., W.B., L.M., O.H.), Departments of Pathology (H.M.), Gynecology (T.S.), Neurology and Institute of Neurogenetics (P.C.), and Paediatric and Adult Movement Disorders and Neuropsychiatry and Institute of Neurogenetics (T.B.), Institute of Integrative and Experimental Genomics (B.R.), University of Luebeck, 23538 Luebeck, Germany; and Department of Biophysics and Biophysical Chemistry (J.M.K.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Jennifer M Kavran
- Department of Paediatric and Adolescent Medicine, and Division of Experimental Paediatric Endocrinology and Diabetes (R.W., W.B., L.M., O.H.), Departments of Pathology (H.M.), Gynecology (T.S.), Neurology and Institute of Neurogenetics (P.C.), and Paediatric and Adult Movement Disorders and Neuropsychiatry and Institute of Neurogenetics (T.B.), Institute of Integrative and Experimental Genomics (B.R.), University of Luebeck, 23538 Luebeck, Germany; and Department of Biophysics and Biophysical Chemistry (J.M.K.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Tobias Bäumer
- Department of Paediatric and Adolescent Medicine, and Division of Experimental Paediatric Endocrinology and Diabetes (R.W., W.B., L.M., O.H.), Departments of Pathology (H.M.), Gynecology (T.S.), Neurology and Institute of Neurogenetics (P.C.), and Paediatric and Adult Movement Disorders and Neuropsychiatry and Institute of Neurogenetics (T.B.), Institute of Integrative and Experimental Genomics (B.R.), University of Luebeck, 23538 Luebeck, Germany; and Department of Biophysics and Biophysical Chemistry (J.M.K.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Philipp Capetian
- Department of Paediatric and Adolescent Medicine, and Division of Experimental Paediatric Endocrinology and Diabetes (R.W., W.B., L.M., O.H.), Departments of Pathology (H.M.), Gynecology (T.S.), Neurology and Institute of Neurogenetics (P.C.), and Paediatric and Adult Movement Disorders and Neuropsychiatry and Institute of Neurogenetics (T.B.), Institute of Integrative and Experimental Genomics (B.R.), University of Luebeck, 23538 Luebeck, Germany; and Department of Biophysics and Biophysical Chemistry (J.M.K.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Olaf Hiort
- Department of Paediatric and Adolescent Medicine, and Division of Experimental Paediatric Endocrinology and Diabetes (R.W., W.B., L.M., O.H.), Departments of Pathology (H.M.), Gynecology (T.S.), Neurology and Institute of Neurogenetics (P.C.), and Paediatric and Adult Movement Disorders and Neuropsychiatry and Institute of Neurogenetics (T.B.), Institute of Integrative and Experimental Genomics (B.R.), University of Luebeck, 23538 Luebeck, Germany; and Department of Biophysics and Biophysical Chemistry (J.M.K.), Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
30
|
McClelland KS, Bell K, Larney C, Harley VR, Sinclair AH, Oshlack A, Koopman P, Bowles J. Purification and Transcriptomic Analysis of Mouse Fetal Leydig Cells Reveals Candidate Genes for Specification of Gonadal Steroidogenic Cells1. Biol Reprod 2015; 92:145. [DOI: 10.1095/biolreprod.115.128918] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/02/2015] [Indexed: 01/12/2023] Open
|
31
|
Nygaard MB, Almstrup K, Lindbæk L, Christensen ST, Svingen T. Cell context-specific expression of primary cilia in the human testis and ciliary coordination of Hedgehog signalling in mouse Leydig cells. Sci Rep 2015; 5:10364. [PMID: 25992706 PMCID: PMC4438617 DOI: 10.1038/srep10364] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/09/2015] [Indexed: 12/04/2022] Open
Abstract
Primary cilia are sensory organelles that coordinate numerous cellular signalling pathways during development and adulthood. Defects in ciliary assembly or function lead to a series of developmental disorders and diseases commonly referred to as ciliopathies. Still, little is known about the formation and function of primary cilia in the mammalian testis. Here, we characterized primary cilia in adult human testis and report a constitutive expression of cilia in peritubular myoid cells and a dynamic expression of cilia in differentiating Leydig cells. Primary cilia are generally absent from cells of mature seminiferous epithelium, but present in Sertoli cell-only tubules in Klinefelter syndrome testis. Peritubular cells in atrophic testis produce overly long cilia. Furthermore cultures of growth-arrested immature mouse Leydig cells express primary cilia that are enriched in components of Hedgehog signalling, including Smoothened, Patched-1, and GLI2, which are involved in regulating Leydig cell differentiation. Stimulation of Hedgehog signalling increases the localization of Smoothened to the cilium, which is followed by transactivation of the Hedgehog target genes, Gli1 and Ptch1. Our findings provide new information on the spatiotemporal formation of primary cilia in the testis and show that primary cilia in immature Leydig cells mediate Hedgehog signalling.
Collapse
Affiliation(s)
- Marie Berg Nygaard
- 1] University Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen DK-2100, Denmark [2] Department of Biology, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Kristian Almstrup
- University Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen DK-2100, Denmark
| | - Louise Lindbæk
- Department of Biology, University of Copenhagen, Copenhagen DK-2100, Denmark
| | | | - Terje Svingen
- 1] University Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Copenhagen DK-2100, Denmark [2] Department of Toxicology and Risk Assessment, National Food Institute, Technical University of Denmark, Søborg DK-2860, Denmark
| |
Collapse
|
32
|
Barrionuevo F, Burgos M, Jiménez R. Origin and function of embryonic Sertoli cells. Biomol Concepts 2015; 2:537-47. [PMID: 25962053 DOI: 10.1515/bmc.2011.044] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 09/16/2011] [Indexed: 11/15/2022] Open
Abstract
In the adult testis, Sertoli cells (SCs) are the epithelial supporting cells of the seminiferous tubules that provide germ cells (GCs) with the required nutrients and structural and regulatory support to complete spermatogenesis. SCs also form the blood-testis barrier, phagocytose apoptotic spermatocytes and cell debris derived from spermiogenesis, and produce and secrete numerous paracrine and endocrine signals involved in different regulatory processes. In addition to their essential functions in the adult testis, SCs play a pivotal role during testis development. They are the first cells to differentiate in the embryonic XY gonadal primordium and are involved in the regulation of testis-specific differentiation processes, such as prevention of GC entry into meiosis, Leydig and peritubular myoid cell differentiation, and regression of the Müllerian duct, the anlagen of the uterus, oviducts, and the upper part of the vagina. Expression of the Y-linked gene SRY in pre-SCs initiates a genetic cascade that leads to SC differentiation and subsequently to testis development. Since the identification of the SRY gene, many Sertoli-specific transcription factors and signals underlying the molecular mechanisms of early testis differentiation have been identified. Here, we review the state of the art of the molecular interactions that commit the supporting cell lineage of the gonadal primordium to differentiate as SCs and the subsequent Sertoli-specific signaling pathways involved in early testis differentiation.
Collapse
|
33
|
Wainwright EN, Svingen T, Ng ET, Wicking C, Koopman P. Primary cilia function regulates the length of the embryonic trunk axis and urogenital field in mice. Dev Biol 2014; 395:342-54. [DOI: 10.1016/j.ydbio.2014.08.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 08/20/2014] [Accepted: 08/27/2014] [Indexed: 01/06/2023]
|
34
|
Liu Y, Wei Z, Huang Y, Bai C, Zan L, Li G. Cyclopamine did not affect mouse oocyte maturation in vitro but decreased early embryonic development. Anim Sci J 2014; 85:840-7. [PMID: 24889396 DOI: 10.1111/asj.12220] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 01/30/2014] [Indexed: 12/31/2022]
Abstract
Hedgehog (Hh) pathway has been studied in various animal body life procedures and is suggested to be important for the development of multiple organs. The genes involved in the Hh signaling pathway were expressed in the ovary of mice, pigs and cattle. However, the function of Hh signaling pathway on oocyte maturation and early embryonic development is still controversial. We detected the effect of sonic hedgehog (Shh) and cyclopamine on the in vitro maturation of mouse oocytes and embryo development. The results showed that the presence of Shh or cyclopamine resulted in similar oocyte maturation to control groups. Shh did not improve early embryonic development. However, the supplement of cyclopamine depressed early embryo development. The mRNA of shh, ptch1, smo and gli1 were less detected in the denuded oocytes. The expression levels of ptch1 ascended from the uncleaved zygote to blastocyst stage. Smo or gli1 were expressed on a higher level at the two-cell or four-cell stage in early embryonic development separately. Therefore, Shh did not affect mouse oocyte maturation and early embryo development, but cyclopamine led to inhibited development of mouse early embryo. The effects of Hh signaling on the oocyte maturation and early embryo development might be species-specific.
Collapse
Affiliation(s)
- Yang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | | | | | | | | | | |
Collapse
|
35
|
Chen M, Wang X, Wang Y, Zhang L, Xu B, Lv L, Cui X, Li W, Gao F. Wt1 Is Involved in Leydig Cell Steroid Hormone Biosynthesis by Regulating Paracrine Factor Expression in Mice1. Biol Reprod 2014; 90:71. [DOI: 10.1095/biolreprod.113.114702] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
36
|
Svingen T, Koopman P. Building the mammalian testis: origins, differentiation, and assembly of the component cell populations. Genes Dev 2013; 27:2409-26. [PMID: 24240231 PMCID: PMC3841730 DOI: 10.1101/gad.228080.113] [Citation(s) in RCA: 265] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Development of testes in the mammalian embryo requires the formation and assembly of several cell types that allow these organs to achieve their roles in male reproduction and endocrine regulation. Testis development is unusual in that several cell types such as Sertoli, Leydig, and spermatogonial cells arise from bipotential precursors present in the precursor tissue, the genital ridge. These cell types do not differentiate independently but depend on signals from Sertoli cells that differentiate under the influence of transcription factors SRY and SOX9. While these steps are becoming better understood, the origins and roles of many testicular cell types and structures-including peritubular myoid cells, the tunica albuginea, the arterial and venous blood vasculature, lymphatic vessels, macrophages, and nerve cells-have remained unclear. This review synthesizes current knowledge of how the architecture of the testis unfolds and highlights the questions that remain to be explored, thus providing a roadmap for future studies that may help illuminate the causes of XY disorders of sex development, infertility, and testicular cancers.
Collapse
Affiliation(s)
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| |
Collapse
|
37
|
Goswami D, Kusre G, Dutta HK, Sarma A. The prune belly syndrome in a female foetus with urorectal septum malformation sequence: a case report on a rare entity with an unusual association. J Clin Diagn Res 2013; 7:1727-9. [PMID: 24086893 DOI: 10.7860/jcdr/2013/5672.3243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 04/11/2013] [Indexed: 11/24/2022]
Abstract
The prune belly syndrome is a rare congenital anomaly which is characterized by the triad of an absent or a deficient development of the abdominal muscle, bilateral cryptorchidism and an anomalous urinary tract. In its full form, this condition occurs only in males. However, a similar condition occurs in females in the absence of cryptorchidism. On the other hand, the urorectal septum malformation sequence is a lethal congenital malformation which is characterized by the development of a phallus like structure, a smooth perineum and the absence of urethral, vaginal and anal openings. We are reporting a case of a female foetus with the prune belly syndrome, which was associated with a urorectal septum malformation sequence. A dead foetus with a protruded abdomen and ambiguous genitalia, was born at 32 weeks of pregnancy. On autopsy, it was found to have female internal genital organs. The left kidney, the urinary bladder and the rectum were absent. The sigmoid colon, the ureters and the fallopian tubes opened into a common cloacal sac. The histopathological examination of the ovary showed the presence of Leydig's cells. The occurrence of the female counterpart of the prune belly syndrome is extremely rare and only few of such cases were found to be discussed in the details in the indexed English literature so far. Hence, we hope that this case report will contribute to the existing knowledge on the prune belly syndrome.
Collapse
Affiliation(s)
- Dibyajyoti Goswami
- Post Graduate Trainee, Department of Anatomy, Assam Medical College and Hospital , Dibrugarh, Assam-786002, India
| | | | | | | |
Collapse
|
38
|
Miyabayashi K, Katoh-Fukui Y, Ogawa H, Baba T, Shima Y, Sugiyama N, Kitamura K, Morohashi KI. Aristaless related homeobox gene, Arx, is implicated in mouse fetal Leydig cell differentiation possibly through expressing in the progenitor cells. PLoS One 2013; 8:e68050. [PMID: 23840809 PMCID: PMC3695952 DOI: 10.1371/journal.pone.0068050] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 05/24/2013] [Indexed: 11/19/2022] Open
Abstract
Development of the testis begins with the expression of the SRY gene in pre-Sertoli cells. Soon after, testis cords containing Sertoli and germ cells are formed and fetal Leydig cells subsequently develop in the interstitial space. Studies using knockout mice have indicated that multiple genes encoding growth factors and transcription factors are implicated in fetal Leydig cell differentiation. Previously, we demonstrated that the Arx gene is implicated in this process. However, how ARX regulates Leydig cell differentiation remained unknown. In this study, we examined Arx KO testes and revealed that fetal Leydig cell numbers largely decrease throughout the fetal life. Since our study shows that fetal Leydig cells rarely proliferate, this decrease in the KO testes is thought to be due to defects of fetal Leydig progenitor cells. In sexually indifferent fetal gonads of wild type, ARX was expressed in the coelomic epithelial cells and cells underneath the epithelium as well as cells at the gonad-mesonephros border, both of which have been described to contain progenitors of fetal Leydig cells. After testis differentiation, ARX was expressed in a large population of the interstitial cells but not in fetal Leydig cells, raising the possibility that ARX-positive cells contain fetal Leydig progenitor cells. When examining marker gene expression, we observed cells as if they were differentiating into fetal Leydig cells from the progenitor cells. Based on these results, we propose that ARX acts as a positive factor for differentiation of fetal Leydig cells through functioning at the progenitor stage.
Collapse
Affiliation(s)
- Kanako Miyabayashi
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuko Katoh-Fukui
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hidesato Ogawa
- Advanced ICT Research Institute Kobe, National Institute of Information and Communications Technology, Kobe, Japan
| | - Takashi Baba
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichi Shima
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriyuki Sugiyama
- Department of Anatomy and Developmental Biology, Graduate School of Medical Science, Kyoto Prefecture University of Medicine, Kyoto, Japan
| | - Kunio Kitamura
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Ken-ichirou Morohashi
- Department of Molecular Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- * E-mail:
| |
Collapse
|
39
|
Barsoum IB, Kaur J, Ge RS, Cooke PS, Yao HHC. Dynamic changes in fetal Leydig cell populations influence adult Leydig cell populations in mice. FASEB J 2013; 27:2657-66. [PMID: 23568777 DOI: 10.1096/fj.12-225060] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Testes contain two distinct Leydig cell populations during development: fetal and adult Leydig cells (FLCs and ALCs, respectively). ALCs are not derived from FLCs, and it is unknown whether these two populations share common progenitors. We discovered that hedgehog (Hh) signaling is responsible for transforming steroidogenic factor 1-positive (SF1(+)) progenitors into FLCs. However, not all SF1(+) progenitors become FLCs, and some remain undifferentiated through fetal development. We therefore hypothesized that if FLCs and ALCs share SF1(+) progenitors, increased Hh pathway activation in SF1(+) progenitor cells could change the dynamics and distribution of SF1(+) progenitors, FLCs, and ALCs. Using a genetic model involving constitutive activation of Hh pathway in SF1(+) cells, we observed reduced numbers of SF1(+) progenitor cells and increased FLCs. Conversely, increased Hh activation led to decreased ALC populations prepubertally, while adult ALC numbers were comparable to control testes. Hence, reduction in SF1(+) progenitors temporarily affects ALC numbers, suggesting that SF1(+) progenitors in fetal testes are a potential source of both FLCs and ALCs. Besides transient ALC defects, adult animals with Hh activation in SF1(+) progenitors had reduced testicular weight, oligospermia, and decreased sperm mobility. These defects highlight the importance of properly regulated Hh signaling in Leydig cell development and testicular functions.
Collapse
Affiliation(s)
- Ivraym B Barsoum
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | | | | | | | | |
Collapse
|
40
|
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
|
41
|
Bashamboo A, McElreavey K. Gene Mutations Associated with Anomalies of Human Gonad Formation. Sex Dev 2013; 7:126-46. [DOI: 10.1159/000342188] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
42
|
Ahmed SF, Bashamboo A, Lucas-Herald A, McElreavey K. Understanding the genetic aetiology in patients with XY DSD. Br Med Bull 2013; 106:67-89. [PMID: 23529942 DOI: 10.1093/bmb/ldt008] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Disorders of sex development (DSD) consist of a wide range of disorders and are commoner in those with an XY karyotype. In over half of these cases who have a 46,XY karyotype and who are raised as boys, the underlying aetiology remains unclear. AREAS OF AGREEMENT Identification of the underlying genetic abnormality may predict long-term outcome. However, genetic abnormalities that are associated with XY DSD manifest themselves with a wide range of phenotype. To understand the aetiology as well as the phenotypic variation, there is a need to harness the advanced genetic technology that is now available. AREAS OF CONTROVERSY The point at which genetic analysis should be undertaken in the course of investigations is unclear. In addition, there is little agreement on the most effective approach for genetic analysis that will be of clinical benefit to the patient. AREAS TIMELY FOR DEVELOPING RESEARCH There is a need to understand and improve the clinical utility of genetic analysis in the clinical setting of the patient with a suspected DSD. This will be even more important when parallel gene sequencing identifies variations in multiple genes.
Collapse
Affiliation(s)
- S F Ahmed
- School of Medicine, University of Glasgow, Royal Hospital for Sick Children, Yorkhill, Glasgow, UK.
| | | | | | | |
Collapse
|
43
|
Ren Y, Cowan RG, Migone FF, Quirk SM. Overactivation of hedgehog signaling alters development of the ovarian vasculature in mice. Biol Reprod 2012; 86:174. [PMID: 22402963 DOI: 10.1095/biolreprod.112.099176] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The hedgehog (HH) signaling pathway is critical for ovarian function in Drosophila, but its role in the mammalian ovary has not been defined. Previously, expression of a dominant active allele of the HH signal transducer protein smoothened (SMO) in Amhr2(cre/+)SmoM2 mice caused anovulation in association with a lack of smooth muscle in the theca of developing follicles. The current study examined events during the first 2 wk of life in Amhr2(cre/+)SmoM2 mice to gain insight into the cause of anovulation. Expression of transcriptional targets of HH signaling, Gli1, Ptch1, and Hhip, which are used as measures of pathway activity, were elevated during the first several days of life in Amhr2(cre/+)SmoM2 mice compared to controls but were similar to controls in older mice. Microarray analysis showed that genes with increased expression in 2-day-old mutants compared to controls were enriched for the processes of vascular and tube development and steroidogenesis. The density of platelet endothelial cell adhesion molecule (PECAM)-labeled endothelial tubes was increased in the cortex of newborn ovaries of mutant mice. Costaining of preovulatory follicles for PECAM and smooth muscle actin showed that muscle-type vascular support cells are deficient in theca of mutant mice. Expression of genes for steroidogenic enzymes that are normally expressed in the fetal adrenal gland were elevated in newborn ovaries of mutant mice. In summary, overactivation of HH signaling during early life alters gene expression and vascular development and this is associated with the lifelong development of anovulatory follicles in which the thecal vasculature fails to mature appropriately.
Collapse
Affiliation(s)
- Yi Ren
- Department of Animal Science, Cornell University, Ithaca, New York 14853, USA
| | | | | | | |
Collapse
|
44
|
Franco HL, Yao HHC. Sex and hedgehog: roles of genes in the hedgehog signaling pathway in mammalian sexual differentiation. Chromosome Res 2012; 20:247-58. [PMID: 22105695 DOI: 10.1007/s10577-011-9254-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The chromosome status of the mammalian embryo initiates a multistage process of sexual development in which the bipotential reproductive system establishes itself as either male or female. These events are governed by intricate cell-cell and interorgan communication that is regulated by multiple signaling pathways. The hedgehog signaling pathway was originally identified for its key role in the development of Drosophila, but is now recognized as a critical developmental regulator in many species, including humans. In addition to its developmental roles, the hedgehog signaling pathway also modulates adult organ function, and misregulation of this pathway often leads to diseases, such as cancer. The hedgehog signaling pathway acts through its morphogenetic ligands that signal from ligand-producing cells to target cells over a specified distance. The target cells then respond in a graded manner based on the concentration of the ligands that they are exposed to. Through this unique mechanism of action, the hedgehog signaling pathway elicits cell fate determination, epithelial-mesenchymal interactions, and cellular homeostasis. Here, we review current findings on the roles of hedgehog signaling in the sexually dimorphic development of the reproductive organs with an emphasis on mammals and comparative evidence in other species.
Collapse
Affiliation(s)
- Heather L Franco
- Reproductive Developmental Biology Group, Laboratory of Reproductive and Developmental Toxicity, National Institute of Environmental Health Sciences, 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| | | |
Collapse
|
45
|
Levina E, Chen M, Carkner R, Shtutman M, Buttyan R. Paracrine Hedgehog increases the steroidogenic potential of prostate stromal cells in a Gli-dependent manner. Prostate 2012; 72:817-24. [PMID: 22025366 DOI: 10.1002/pros.21500] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Accepted: 09/19/2011] [Indexed: 01/03/2023]
Abstract
Acquired intratumoral steroidogenesis is involved in progression of prostate cancer to castration resistant disease (CRPC) and a target for improved therapeutics. Recent work has shown that prostate cancer cells can acquire steroidogenic activity as they progress to a therapeutic-resistant state. However, benign prostate stromal cells (PrSCs) also have steroidogenic potential though they are often overlooked as a source of intratumoral androgens. Here, we present preliminary studies showing that the steroidogenic activity of primary human PrSCs is significantly increased by exposure to a Hedgehog agonist (SAG) or by transduction of PrSCs with lentiviruses that expresses active Gli2 (Gli2ΔN), a transcription factor that is triggered by Hh signaling. Comparative gene expression profiling on Chips, that was confirmed by quantitative real-time PCR, revealed that hedgehog agonist treatment induced in these cells expressions of hedgehog target genes (Gli1, Ptch1, and SCUBE1) plus a specific cadre of genes involved in cholesterol/steroid biosynthesis, metabolism, and transport. Genes involved downstream in steroid hormone generation, including CYP17A1 and CYP19A1 were also induced. Both the hedgehog agonist and the Gli2-expressing lentivirus significantly increased the output of testosterone (T) from PrSCs that were supplemented with dihydroepiandrosterone (DHEA), an adrenal precursor of T. Finally, knockdown of Gli2 by siRNA suppressed the ability of SAG to induce this response. Collectively, our data indicate that hedgehog/Gli signaling may be a factor in acquired intratumoral steroidogenesis of a prostate tumor through its actions on stromal cells in the tumor microenvironment and an influence for the development of CRPC.
Collapse
Affiliation(s)
- Elina Levina
- South Carolina College of Pharmacy, Department of Biomedical and Pharmaceutical Sciences, Columbia, South Carolina, USA
| | | | | | | | | |
Collapse
|
46
|
|
47
|
Ungewitter EK, Yao HHC. How to make a gonad: cellular mechanisms governing formation of the testes and ovaries. Sex Dev 2012; 7:7-20. [PMID: 22614391 PMCID: PMC3474884 DOI: 10.1159/000338612] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Sex determination of the gonad is an extraordinary process by which a single organ anlage is directed to form one of two different structures, a testis or an ovary. Morphogenesis of these two organs utilizes many common cellular events; differences in the timing and execution of these events must combine to generate sexually dimorphic structures. In this chapter, we review recent research on the cellular processes of gonad morphogenesis, focusing on data from mouse models. We highlight the shared cellular mechanisms in testis and ovary morphogenesis and examine the differences that enable formation of the two organs responsible for the perpetuation of all sexually reproducing species.
Collapse
Affiliation(s)
- E K Ungewitter
- Reproductive Developmental Biology Group, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | |
Collapse
|
48
|
Hayrabedyan S, Todorova K, Pashova S, Mollova M, Fernández N. Sertoli Cell Quiescence - New Insights. Am J Reprod Immunol 2012; 68:451-5. [DOI: 10.1111/j.1600-0897.2012.01137.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 03/13/2012] [Indexed: 12/26/2022] Open
Affiliation(s)
- Soren Hayrabedyan
- Institute of Biology and Immunology of Reproduction; BAS; Sofia; Bulgaria
| | | | - Shina Pashova
- Institute of Biology and Immunology of Reproduction; BAS; Sofia; Bulgaria
| | - Margarita Mollova
- Institute of Biology and Immunology of Reproduction; BAS; Sofia; Bulgaria
| | - Nelson Fernández
- School of Biological Sciences; University of Essex; Colchester; UK
| |
Collapse
|
49
|
Abstract
Disorders of sex development often arise from anomalies in the molecular or cellular networks that guide the differentiation of the embryonic gonad into either a testis or an ovary, two functionally distinct organs. The activation of the Y-linked gene Sry (sex-determining region Y) and its downstream target Sox9 (Sry box-containing gene 9) triggers testis differentiation by stimulating the differentiation of Sertoli cells, which then direct testis morphogenesis. Once engaged, a genetic pathway promotes the testis development while actively suppressing genes involved in ovarian development. This review focuses on the events of testis determination and the struggle to maintain male fate in the face of antagonistic pressure from the underlying female programme.
Collapse
|
50
|
O'Hara WA, Azar WJ, Behringer RR, Renfree MB, Pask AJ. Desert hedgehog is a mammal-specific gene expressed during testicular and ovarian development in a marsupial. BMC DEVELOPMENTAL BIOLOGY 2011; 11:72. [PMID: 22132805 PMCID: PMC3293750 DOI: 10.1186/1471-213x-11-72] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 12/01/2011] [Indexed: 12/26/2022]
Abstract
BACKGROUND Desert hedgehog (DHH) belongs to the hedgehog gene family that act as secreted intercellular signal transducers. DHH is an essential morphogen for normal testicular development and function in both mice and humans but is not present in the avian lineage. Like other hedgehog proteins, DHH signals through the patched (PTCH) receptors 1 and 2. Here we examine the expression and protein distribution of DHH, PTCH1 and PTCH2 in the developing testes of a marsupial mammal (the tammar wallaby) to determine whether DHH signalling is a conserved factor in gonadal development in all therian mammals. RESULTS DHH, PTCH1 and PTCH2 were present in the marsupial genome and highly conserved with their eutherian orthologues. Phylogenetic analyses indicate that DHH has recently evolved and is a mammal-specific hedgehog orthologue. The marsupial PTCH2 receptor had an additional exon (exon 21a) not annotated in eutherian PTCH2 proteins. Interestingly we found evidence of this exon in humans and show that its translation would result in a truncated protein with functions similar to PTCH1. We also show that DHH expression was not restricted to the testes during gonadal development (as in mice), but was also expressed in the developing ovary. Expression of DHH, PTCH1 and PTCH2 in the adult tammar testis and ovary was consistent with findings in the adult mouse. CONCLUSIONS These data suggest that there is a highly conserved role for DHH signalling in the differentiation and function of the mammalian testis and that DHH may be necessary for marsupial ovarian development. The receptors PTCH1 and PTCH2 are highly conserved mediators of hedgehog signalling in both the developing and adult marsupial gonads. Together these findings indicate DHH is an essential therian mammal-specific morphogen in gonadal development and gametogenesis.
Collapse
Affiliation(s)
- William A O'Hara
- Department of Molecular and Cellular Biology, The University of Connecticut, Storrs, CT 06269, USA
| | | | | | | | | |
Collapse
|