1
|
Chen T, Zhang B, He G, Shen C, Wang N, Zong J, Chen X, Chen L, Li C, Zhou X. Exosomes-mediated retinoic acid disruption: A link between gut microbiota depletion and impaired spermatogenesis. Toxicology 2024; 508:153907. [PMID: 39121937 DOI: 10.1016/j.tox.2024.153907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Gut microbiota symbiosis faces enormous challenge with increasing exposure to drugs such as environmental poisons and antibiotics. The gut microbiota is an important component of the host microbiota and has been proven to be involved in regulating spermatogenesis, but the molecular mechanism is still unclear. A male mouse model with gut microbiota depletion/dysbiosis was constructed by adding combined antibiotics to free drinking water, and reproductive parameters such as epididymal sperm count, testicular weight and paraffin sections were measured. Testicular transcriptomic and serum metabolomic analyses were performed to reveal the molecular mechanism of reproductive dysfunction induced by gut microbiota dysbiosis in male mice.This study confirms that antibiotic induced depletion of gut microbiota reduces sperm count in the epididymis and reduces germ cells in the seminiferous tubules in male mice. Further study showed that exosomes isolated from microbiota-depleted mice led to abnormally high levels of retinoic acid and decrease in the number of germ cells in the seminiferous tubules and sperm in the epididymis. Finally, abnormally high levels of retinoic acid was confirmed to disrupted meiotic processes, resulting in spermatogenesis disorders. This study proposed the concept of the gut microbiota-exosome-retinoic acid-testicular axis and demonstrated that depletion of the gut microbiota caused changes in the function of exosomes, which led to abnormal retinoic acid metabolism in the testis, thereby impairing meiosis and spermatogenesis processes.
Collapse
Affiliation(s)
- Tong Chen
- College of Animal Sciences, Jilin University, No. 5333 Xi'an Road, Lvyuan District, Changchun, Jilin 130062, China
| | - Boqi Zhang
- College of Animal Sciences, Jilin University, No. 5333 Xi'an Road, Lvyuan District, Changchun, Jilin 130062, China
| | - Guitian He
- College of Animal Sciences, Jilin University, No. 5333 Xi'an Road, Lvyuan District, Changchun, Jilin 130062, China
| | - Caomeihui Shen
- College of Animal Sciences, Jilin University, No. 5333 Xi'an Road, Lvyuan District, Changchun, Jilin 130062, China
| | - Nan Wang
- College of Animal Sciences, Jilin University, No. 5333 Xi'an Road, Lvyuan District, Changchun, Jilin 130062, China
| | - Jinxin Zong
- College of Animal Sciences, Jilin University, No. 5333 Xi'an Road, Lvyuan District, Changchun, Jilin 130062, China
| | - Xue Chen
- College of Animal Sciences, Jilin University, No. 5333 Xi'an Road, Lvyuan District, Changchun, Jilin 130062, China
| | - Lu Chen
- College of Animal Sciences, Jilin University, No. 5333 Xi'an Road, Lvyuan District, Changchun, Jilin 130062, China
| | - Chunjin Li
- College of Animal Sciences, Jilin University, No. 5333 Xi'an Road, Lvyuan District, Changchun, Jilin 130062, China.
| | - Xu Zhou
- College of Animal Sciences, Jilin University, No. 5333 Xi'an Road, Lvyuan District, Changchun, Jilin 130062, China.
| |
Collapse
|
2
|
Perrotta G, Condrea D, Ghyselinck NB. Meiosis and retinoic acid in the mouse fetal gonads: An unforeseen twist. Curr Top Dev Biol 2024; 161:59-88. [PMID: 39870439 DOI: 10.1016/bs.ctdb.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
In mammals, differentiation of germ cells is crucial for sexual reproduction, involving complex signaling pathways and environmental cues defined by the somatic cells of the gonads. This review examines the long-standing model positing that all-trans retinoic acid (ATRA) acts as a meiosis-inducing substance (MIS) in the fetal ovary by inducing expression of STRA8 in female germ cells, while CYP26B1 serves as a meiosis-preventing substance (MPS) in the fetal testis by degrading ATRA and preventing STRA8 expression in the male germ cells until postnatal development. Recent genetic studies in the mouse challenge this paradigm, revealing that meiosis initiation in female germ cells can occur independently of ATRA signaling, with key roles played by other intrinsic factors like DAZL and DMRT1, and extrinsic signals such as BMPs and vitamin C. Thus, ATRA can no longer be considered as 'the' long-searched MIS. Furthermore, evidence indicates that CYP26B1 does not prevent meiosis by degrading ATRA in the fetal testis, but acts by degrading an unidentified MIS or synthesizing an equally unknown MPS. By emphasizing the necessity of genetic loss-of-function approaches to accurately delineate the roles of signaling molecules such ATRA in vivo, this chapter calls for a reevaluation of the mechanisms instructing and preventing meiosis initiation in the fetal ovary and testis, respectively. It highlights the need for further research into the molecular identities of the signals involved in these processes.
Collapse
Affiliation(s)
- Giulia Perrotta
- Université de Strasbourg, IGBMC UMR 7104, Illkirch, France; CNRS, UMR 7104, Illkirch, France; Inserm, UMR-S 1258, Illkirch, France; IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Diana Condrea
- Université de Strasbourg, IGBMC UMR 7104, Illkirch, France; CNRS, UMR 7104, Illkirch, France; Inserm, UMR-S 1258, Illkirch, France; IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Norbert B Ghyselinck
- Université de Strasbourg, IGBMC UMR 7104, Illkirch, France; CNRS, UMR 7104, Illkirch, France; Inserm, UMR-S 1258, Illkirch, France; IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.
| |
Collapse
|
3
|
Alexander AK, Rodriguez KF, Chen YY, Amato CM, Estermann MA, Nicol B, Xu X, Hung-Chang Yao H. Single-nucleus multiomics reveals the gene-regulatory networks underlying sex determination of murine primordial germ cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.19.581036. [PMID: 39386556 PMCID: PMC11463670 DOI: 10.1101/2024.02.19.581036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Accurate specification of female and male germ cells during embryonic development is critical for sexual reproduction. Primordial germ cells (PGCs) are the bipotential precursors of mature gametes that commit to an oogenic or spermatogenic fate in response to sex-determining cues from the fetal gonad. The critical processes required for PGCs to integrate and respond to signals from the somatic environment in gonads are not understood. In this study, we developed the first single-nucleus multiomics map of chromatin accessibility and gene expression during murine PGC development in both XX and XY embryos. Profiling of cell-type specific transcriptomes and regions of open chromatin from the same cell captured the molecular signatures and gene networks underlying PGC sex determination. Joint RNA and ATAC data for single PGCs resolved previously unreported PGC subpopulations and cataloged a multimodal reference atlas of differentiating PGC clusters. We discovered that regulatory element accessibility precedes gene expression during PGC development, suggesting that changes in chromatin accessibility may prime PGC lineage commitment prior to differentiation. Similarly, we found that sexual dimorphism in chromatin accessibility and gene expression increased temporally in PGCs. Combining single-nucleus sequencing data, we computationally mapped the cohort of transcription factors that regulate the expression of sexually dimorphic genes in PGCs. For example, the gene regulatory networks of XX PGCs are enriched for the transcription factors, TFAP2c, TCFL5, GATA2, MGA, NR6A1, TBX4, and ZFX. Sex-specific enrichment of the forkhead-box and POU6 families of transcription factors was also observed in XY PGCs. Finally, we determined the temporal expression patterns of WNT, BMP, and RA signaling during PGC sex determination, and our discovery analyses identified potentially new cell communication pathways between supporting cells and PGCs. Our results illustrate the diversity of factors involved in programming PGCs towards a sex-specific fate.
Collapse
Affiliation(s)
- Adriana K. Alexander
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Karina F. Rodriguez
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Yu-Ying Chen
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Ciro M. Amato
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Martin A. Estermann
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Barbara Nicol
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Xin Xu
- Epigenetics & Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Humphrey Hung-Chang Yao
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| |
Collapse
|
4
|
Wu D, Zhang K, Guan K, Tan J, Huang C, Sun F. Retinoic acid tiers mitochondrial metabolism to Sertoli Cell-Mediated efferocytosis via a non-RAR-dependent mechanism. Biochem Pharmacol 2024; 225:116281. [PMID: 38744379 DOI: 10.1016/j.bcp.2024.116281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/03/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024]
Abstract
Efferocytosis of massive non-viable germ cells by Sertoli cells (SCs), the specialized phagocytes, is essential for maintaining testis homeostasis. What elusive is the contribution of mitochondrial metabolism to this energy-consuming process, as SC has a preference of aerobic glycolysis. All-trans retinoic acid (ATRA, hereafter referred to as RA) is a well-known morphogen that primarily acts through the nuclear RA receptor (RAR). It sustains SC blood-testisbarrier integrity, and it's SC-derived RA sets the timing of meiotic commitment. In this study, we revisited RA in SC biology, from the perspective of SC-mediated efferocytosis. We provide evidence that RA induces transcriptional programming of multiple regulators involved in efferocytosis, which thereby represses SC-mediated efferocytosis, via a RAR-independent mechanism, as blocking pan-RAR activity fails to rescue RA-induced defective efferocytosis. RA-treated SCs exhibit alternations in mitochondrial dynamics and metabolism, and the hindered efferocytosis can be rescued by stimulating mitochondrial OXPHOS via pharmacological targeting of AMPK and PDK. We thus prefer to propose a signaling axis of RA-mitochondrial metabolism-efferocytosis. Our study uncovers a hitherto unappreciated role of RA in SC biology and tiers mitochondria metabolism to SC-mediated efferocytosis, contributing a deeper understanding of SC in male reproduction.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China
| | - Jiachen Tan
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China; School of Basic Medical Sciences, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
5
|
Li Y, Song P, Zhao J, Zhang W, Liu X, Lv X, Zhao J. Neonatal vitamin A supplementation improves sheep fertility potential. Front Vet Sci 2024; 11:1370576. [PMID: 38756517 PMCID: PMC11097686 DOI: 10.3389/fvets.2024.1370576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
This study aimed to explore the effects of neonatal vitamin A (VA) supplementation on testis development and spermatogenesis. A total of 32 newborn lambs were intramuscularly injected with corn oil (control group) or corn oil + 2500 IU/kg BW VA (VA group). They were slaughtered and sampled at 3 weeks and 8 months of age to analyze spermatogenesis, cell proliferation, hormone secretion, antioxidant status of the testis, and adult sheep sperm parameters. Compared with the control group, the expression of spermatogonial differentiation-related genes in VA group was up-regulated (P < 0.05). Testis weight, seminiferous tubule diameter, number of spermatogonium and spermatocyte, and sperm density increased significantly in VA group at 8 months of age (P < 0.05). Neonatal VA injection upregulated the expression of the cell proliferation marker PCNA and cell cycle-related genes in the testis (P < 0.05). VA increased the concentrations of testosterone (T), luteinizing hormone (LH), and follicle-stimulating hormone (FSH) in the serum and upregulated steroidogenesis-related genes in the testis (P < 0.05). The antioxidant levels in the VA group were maintained at high levels. The total antioxidant capacity (T-AOC), antioxidant enzyme content and antioxidant-related genes were increased in the testis (P < 0.05). Furthermore, neonatal VA injection activated retinoic acid (RA) signaling to maintain the blood-testosterone barrier (BTB) in the testis of 3-week-old sheep. AMP-activated protein kinase (AMPK) and protein kinase B (AKT) signaling were also modulated in the sheep testis (P < 0.05). Taken together, VA supplementation in newborn rams promotes testis development and spermatogenesis to improve fertility.
Collapse
Affiliation(s)
- Yating Li
- College of Animal Science, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Pengkang Song
- College of Animal Science, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Jiamin Zhao
- College of Animal Science, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Weipeng Zhang
- College of Animal Science, Shanxi Agricultural University, Jinzhong, Shanxi, China
| | - Xiangdong Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Xiaoyang Lv
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou, Jiangsu, China
| | - Junxing Zhao
- College of Animal Science, Shanxi Agricultural University, Jinzhong, Shanxi, China
| |
Collapse
|
6
|
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
|
7
|
Green ES, Chan HY, Frost E, Griffiths M, Hutchison J, Martin JH, Mihalas BP, Newman T, Dunleavy JEM. Recent advances in reproductive research in Australia and New Zealand: highlights from the Annual Meeting of the Society for Reproductive Biology, 2022. Reprod Fertil Dev 2024; 36:RD23213. [PMID: 38346692 DOI: 10.1071/rd23213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/09/2024] [Indexed: 04/11/2024] Open
Abstract
In 2022, the Society for Reproductive Biology came together in Christchurch New Zealand (NZ), for its first face-to-face meeting since the global COVID-19 pandemic. The meeting showcased recent advancements in reproductive research across a diverse range of themes relevant to human health and fertility, exotic species conservation, and agricultural breeding practices. Here, we highlight the key advances presented across the main themes of the meeting, including advances in addressing opportunities and challenges in reproductive health related to First Nations people in Australia and NZ; increasing conservation success of exotic species, including ethical management of invasive species; improvements in our understanding of developmental biology, specifically seminal fluid signalling, ovarian development and effects of environmental impacts such as endocrine-disrupting chemicals; and leveraging scientific breakthroughs in reproductive engineering to drive solutions for fertility, including in assisted reproductive technologies in humans and agricultural industries, and for regenerative medicine.
Collapse
Affiliation(s)
- Ella S Green
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Hon Y Chan
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Emily Frost
- Fertility & Research Centre, Discipline of Women's Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Meaghan Griffiths
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Vic., Australia; and Gynaecology Research Centre, Royal Women's Hospital, Parkville, Vic., Australia
| | - Jennifer Hutchison
- School of BioSciences, Faculty of Science, The University of Melbourne, Melbourne, Vic., Australia; and Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton Vic., Australia; and Department of Molecular and Translational Science, Monash University, Clayton, Vic., Australia
| | - Jacinta H Martin
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, Australia; and Infertility and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Bettina P Mihalas
- The Oocyte Biology Research Unit, Discipline of Women's Health, School of Clinical Medicine, Faculty of Medicine and Health, The University of NSW Sydney, Randwick, NSW, Australia
| | - Trent Newman
- School of BioSciences, Faculty of Science, The University of Melbourne, Melbourne, Vic., Australia
| | - Jessica E M Dunleavy
- School of BioSciences and Bio21 Institute, Faculty of Science, The University of Melbourne, Melbourne, Vic., Australia
| |
Collapse
|
8
|
Wang G, Lu R, Gao Y, Zhang H, Shi X, Ma W, Wu L, Tian X, Liu H, Jiang H, Li X, Ma X. Molecular characterization and potential function of Rxrγ in gonadal differentiation of Chinese soft-shelled turtle (Pelodiscus sinensis). J Steroid Biochem Mol Biol 2023; 233:106360. [PMID: 37429547 DOI: 10.1016/j.jsbmb.2023.106360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 07/12/2023]
Abstract
Retinoid X receptor (RXR) is a member of the ligand-dependent nuclear receptor family. Previous studies revealed that RXRs are involved in reproduction in vertebrates. However, information on the function of RXRs in turtles is scarce. In this study, the Rxrγ cDNA sequence of Pelodiscus sinensis was cloned and analyzed, and a polyclonal antibody was constructed. RXRγ protein showed a positive signal in both mature and differentiated gonads of the turtle. Subsequently, the function of the Rxrγ gene in gonadal differentiation was confirmed using short interfering RNA (RNAi). The full-length cDNA sequence of the Rxrγ gene in P. sinensis was 2152 bp, encoding 407 amino acids and containing typical nuclear receptor family domains, including the DNA-binding domain (DBD), ligand-binding domain (LBD), and activation function 1 (AF1). Moreover, gonadal Ps-Rxrγ showed sexual dimorphism expression patterns in differentiated gonads. Real-time quantitative PCR results revealed that the Rxrγ gene was highly expressed in the turtle ovary. RNAi treatment increased the number of Sertoli cells in ZZ embryonic gonads. Furthermore, RNA interference upregulated Dmrt1 and Sox9 in ZZ and ZW embryonic gonads. However, Foxl2, Cyp19a1, Stra8, and Cyp26b1 were downregulated in embryonic gonads. The results indicated that Rxrγ participated in gonadal differentiation and development in P. sinensis.
Collapse
Affiliation(s)
- Guiyu Wang
- College of Fisheries Henan Normal University, Xinxiang 453007, China
| | - Ruiyi Lu
- College of Fisheries Henan Normal University, Xinxiang 453007, China
| | - Yijie Gao
- College of Fisheries Henan Normal University, Xinxiang 453007, China
| | - Haoran Zhang
- College of Fisheries Henan Normal University, Xinxiang 453007, China
| | - Xi Shi
- College of Fisheries Henan Normal University, Xinxiang 453007, China
| | - Wenge Ma
- College of Fisheries Henan Normal University, Xinxiang 453007, China
| | - Limin Wu
- College of Fisheries Henan Normal University, Xinxiang 453007, China
| | - Xue Tian
- College of Fisheries Henan Normal University, Xinxiang 453007, China
| | - Huifen Liu
- College of Fisheries Henan Normal University, Xinxiang 453007, China
| | - Hongxia Jiang
- College of Fisheries Henan Normal University, Xinxiang 453007, China
| | - Xuejun Li
- College of Fisheries Henan Normal University, Xinxiang 453007, China.
| | - Xiao Ma
- College of Fisheries Henan Normal University, Xinxiang 453007, China.
| |
Collapse
|
9
|
Cintrón-Rivera LG, Oulette G, Prakki A, Burns NM, Patel R, Cyr R, Plavicki J. Exposure to the persistent organic pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, dioxin) disrupts development of the zebrafish inner ear. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 259:106539. [PMID: 37086653 PMCID: PMC10519160 DOI: 10.1016/j.aquatox.2023.106539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/07/2023] [Accepted: 04/09/2023] [Indexed: 05/03/2023]
Abstract
Dioxins are a class of highly toxic and persistent environmental pollutants that have been shown through epidemiological and laboratory-based studies to act as developmental teratogens. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), the most potent dioxin congener, has a high affinity for the aryl hydrocarbon receptor (AHR), a ligand activated transcription factor. TCDD-induced AHR activation during development impairs nervous system, cardiac, and craniofacial development. Despite the robust phenotypes previously reported, the characterization of developmental malformations and our understanding of the molecular targets mediating TCDD-induced developmental toxicity remains limited. In zebrafish, TCDD-induced craniofacial malformations are produced, in part, by the downregulation of SRY-box transcription factor 9b (sox9b), a member of the SoxE gene family. sox9b, along with fellow SoxE gene family members sox9a and sox10, have important functions in the development of the otic placode, the otic vesicle, and, ultimately, the inner ear. Given that sox9b is a known target of TCDD and that transcriptional interactions exist among SoxE genes, we asked whether TCDD exposure impaired the development of the zebrafish auditory system, specifically the otic vesicle, which gives rise to the sensory components of the inner ear. Using immunohistochemistry, in vivo confocal imaging, and time-lapse microscopy, we assessed the impact of TCDD exposure on zebrafish otic vesicle development. We found exposure resulted in structural deficits, including incomplete pillar fusion and altered pillar topography, leading to defective semicircular canal development. The observed structural deficits were accompanied by reduced collagen type II expression in the ear. Together, our findings reveal the otic vesicle as a novel target of TCDD-induced toxicity, suggest that the function of multiple SoxE genes may be affected by TCDD exposure, and provide insight into how environmental contaminants contribute to congenital malformations.
Collapse
Affiliation(s)
- Layra G Cintrón-Rivera
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Gabrielle Oulette
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Aishwarya Prakki
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Nicole M Burns
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA
| | - Ratna Patel
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Rachel Cyr
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Jessica Plavicki
- Department of Pathology and Laboratory Medicine, Brown University, 70 Ship St, Providence, RI, 02903, USA.
| |
Collapse
|
10
|
Cai P, Yuan H, Gao Z, Qiao H, Zhang W, Jiang S, Xiong Y, Gong Y, Wu Y, Jin S, Fu H. 17β-Estradiol Induced Sex Reversal and Gonadal Transcriptome Analysis in the Oriental River Prawn ( Macrobrachium nipponense): Mechanisms, Pathways, and Potential Harm. Int J Mol Sci 2023; 24:ijms24108481. [PMID: 37239827 DOI: 10.3390/ijms24108481] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/04/2023] [Accepted: 05/07/2023] [Indexed: 05/28/2023] Open
Abstract
Sex reversal induced by 17β-estradiol (E2) has shown the potential possibility for monoculture technology development. The present study aimed to determine whether dietary supplementation with different concentrations of E2 could induce sex reversal in M. nipponense, and select the sex-related genes by performing the gonadal transcriptome analysis of normal male (M), normal female (FM), sex-reversed male prawns (RM), and unreversed male prawns (NRM). Histology, transcriptome analysis, and qPCR were performed to compare differences in gonad development, key metabolic pathways, and genes. Compared with the control, after 40 days, feeding E2 with 200 mg/kg at PL25 (PL: post-larvae developmental stage) resulted in the highest sex ratio (female: male) of 2.22:1. Histological observations demonstrated the co-existence of testis and ovaries in the same prawn. Male prawns from the NRM group exhibited slower testis development without mature sperm. RNA sequencing revealed 3702 differentially expressed genes (DEGs) between M vs. FM, 3111 between M vs. RM, and 4978 between FM vs. NRM. Retinol metabolism and nucleotide excision repair pathways were identified as the key pathways for sex reversal and sperm maturation, respectively. Sperm gelatinase (SG) was not screened in M vs. NRM, corroborating the results of the slice D. In M vs. RM, reproduction-related genes such as cathepsin C (CatC), heat shock protein cognate (HSP), double-sex (Dsx), and gonadotropin-releasing hormone receptor (GnRH) were expressed differently from the other two groups, indicating that these are involved in the process of sex reversal. Exogenous E2 can induce sex reversal, providing valuable evidence for the establishment of monoculture in this species.
Collapse
Affiliation(s)
- Pengfei Cai
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Huwei Yuan
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Zijian Gao
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Hui Qiao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Wenyi Zhang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Sufei Jiang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Yiwei Xiong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Yongsheng Gong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Yan Wu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Shubo Jin
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Hongtuo Fu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| |
Collapse
|
11
|
Coen S, Keogh K, Lonergan P, Fair S, Kenny DA. Early life nutrition affects the molecular ontogeny of testicular development in the young bull calf. Sci Rep 2023; 13:6748. [PMID: 37185277 PMCID: PMC10130005 DOI: 10.1038/s41598-022-23743-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 11/04/2022] [Indexed: 05/17/2023] Open
Abstract
Enhanced early life nutrition accelerates sexual development in the bull calf through neuroendocrine-signalling mediated via the hypothalamic-pituitary-testicular axis. Our aim was to assess the impact of contrasting feeding regimes in bull calves during the first 12 weeks of life on the testes transcriptome and proteome. Holstein-Friesian bull calves were offered either a high (HI) or moderate (MOD) plane of nutrition, designed to support target growth rates of 1.0 and 0.5 kg/day, respectively. At 12 weeks of age all calves were euthanized, testicular parenchyma sampled, and global transcriptome (miRNAseq and mRNAseq) and proteome analyses undertaken. Bioinformatic analyses revealed 7 differentially expressed (DE) miRNA and 20 DE mRNA. There were no differentially abundant proteins between the two dietary groups. Integration of omics results highlighted a potential role for the cadherin gene, CDH13, in earlier reproductive development. Furthermore, co-regulatory network analysis of the proteomic data revealed CDH13 as a hub protein within a network enriched for processes related to insulin, IGF-1, androgen and Sertoli cell junction signalling pathways as well as cholesterol biosynthesis. Overall, results highlight a potential role for CDH13 in mediating earlier reproductive development as a consequence of enhanced early life nutrition in the bull calf.
Collapse
Affiliation(s)
- Stephen Coen
- Teagasc Animal and Grassland Research and Innovation Centre, Grange, Dunsany, Co. Meath, Ireland
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kate Keogh
- Teagasc Animal and Grassland Research and Innovation Centre, Grange, Dunsany, Co. Meath, Ireland
| | - Pat Lonergan
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sean Fair
- Laboratory of Animal Reproduction, Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - David A Kenny
- Teagasc Animal and Grassland Research and Innovation Centre, Grange, Dunsany, Co. Meath, Ireland.
| |
Collapse
|
12
|
Zhang B, Chen F, Xu T, Tian Y, Zhang Y, Cao M, Guo X, Yin D. The crosstalk effects of polybrominated diphenyl ethers on the retinoic acid and thyroid hormone signaling pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 883:163590. [PMID: 37088389 DOI: 10.1016/j.scitotenv.2023.163590] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/11/2023] [Accepted: 04/15/2023] [Indexed: 05/03/2023]
Abstract
The toxicological and pathological influences of polybrominated diphenyl ethers (PBDEs) on the animal central nervous system have attracted worldwide attention. However, their mechanism of action has not been completely elucidated. Given that retinoic acid (RA) and thyroid hormone (TH) signaling pathway are closely related to neurodevelopment, the crosstalk between the two signaling pathways at the levels of metabolite conversion, gene expression and ligand-receptor interaction after exposure to two representative PBDE congeners (BDE-47 and BDE-209) using zebrafish larvae, dual reporter gene assay, and docking simulation was studied. Our results clarified that BDE-47 could disrupt the transport and metabolism of retinoids, induce changes in expression of key genes, bind with the seven nuclear receptors, and activate RA signaling pathway. BDE-47 exhibited more effects on the indicators of the two signaling pathways than BDE-209. Furthermore, BDE-47 may disrupt TH signaling pathway by disrupting RA signaling pathway, indicating that RA signal is priorly influenced than TH signal. This work offered a new perspective to elucidate TH signal disruption mechanism induced by PBDEs from RA signaling pathway, which is of great significance to elucidate the health effects of PBDEs.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China; Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Fu Chen
- School of Environmental and Geographical Sciences, Shanghai Normal University, Shanghai 200234, China
| | - Ting Xu
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai 200092, China.
| | - Yijun Tian
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China
| | - Yajie Zhang
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Miao Cao
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Xueping Guo
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China
| | - Daqiang Yin
- Key Laboratory of Yangtze River Water Environment, Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai 200092, China; Shanghai Institute of Pollution Control and Ecological Security, Shanghai 200092, China
| |
Collapse
|
13
|
Cintr N-Rivera LG, Oulette G, Prakki A, Burns NM, Patel R, Cyr R, Plavicki J. Exposure to the persistent organic pollutant 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD, dioxin) disrupts development of the zebrafish inner ear. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532434. [PMID: 36993549 PMCID: PMC10054988 DOI: 10.1101/2023.03.14.532434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Dioxins are a class of highly toxic and persistent environmental pollutants that have been shown through epidemiological and laboratory-based studies to act as developmental teratogens. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), the most potent dioxin congener, has a high affinity for the aryl hydrocarbon receptor (AHR), a ligand activated transcription factor. TCDD-induced AHR activation during development impairs nervous system, cardiac, and craniofacial development. Despite the robust phenotypes previously reported, the characterization of developmental malformations and our understanding of the molecular targets mediating TCDD-induced developmental toxicity remains limited. In zebrafish, TCDD-induced craniofacial malformations are produced, in part, by the downregulation of SRY-box transcription factor 9b ( sox9b ), a member of the SoxE gene family. sox9b , along with fellow SoxE gene family members sox9a and sox10 , have important functions in the development of the otic placode, the otic vesicle, and, ultimately, the inner ear. Given that sox9b in a known target of TCDD and that transcriptional interactions exist among SoxE genes, we asked whether TCDD exposure impaired the development of the zebrafish auditory system, specifically the otic vesicle, which gives rise to the sensory components of the inner ear. Using immunohistochemistry, in vivo confocal imaging, and time-lapse microscopy, we assessed the impact of TCDD exposure on zebrafish otic vesicle development. We found exposure resulted in structural deficits, including incomplete pillar fusion and altered pillar topography, leading to defective semicircular canal development. The observed structural deficits were accompanied by reduced collagen type II expression in the ear. Together, our findings reveal the otic vesicle as a novel target of TCDD-induced toxicity, suggest that the function of multiple SoxE genes may be affected by TCDD exposure, and provide insight into how environmental contaminants contribute to congenital malformations. Highlights The zebrafish ear is necessary to detect changes in motion, sound, and gravity.Embryos exposed to TCDD lack structural components of the developing ear.TCDD exposure impairs formation of the fusion plate and alters pillar topography.The semicircular canals of the ear are required to detect changes in movement.Following TCDD exposure embryos fail to establish semicircular canals.
Collapse
|
14
|
Abstract
Vitamin A (retinol) is a critical micronutrient required for the control of stem cell functions, cell differentiation, and cell metabolism in many different cell types, both during embryogenesis and in the adult organism. However, we must obtain vitamin A from food sources. Thus, the uptake and metabolism of vitamin A by intestinal epithelial cells, the storage of vitamin A in the liver, and the metabolism of vitamin A in target cells to more biologically active metabolites, such as retinoic acid (RA) and 4-oxo-RA, must be precisely regulated. Here, I will discuss the enzymes that metabolize vitamin A to RA and the cytochrome P450 Cyp26 family of enzymes that further oxidize RA. Because much progress has been made in understanding the regulation of ALDH1a2 (RALDH2) actions in the intestine, one focus of this review is on the metabolism of vitamin A in intestinal epithelial cells and dendritic cells. Another focus is on recent data that 4-oxo-RA is a ligand required for the maintenance of hematopoietic stem cell dormancy and the important role of RARβ (RARB) in these stem cells. Despite this progress, many questions remain in this research area, which links vitamin A metabolism to nutrition, immune functions, developmental biology, and nuclear receptor pharmacology.
Collapse
Affiliation(s)
- Lorraine J Gudas
- Department of Pharmacology, and Revlon Pharmaceutical Professor of Pharmacology and Toxicology, Pharmacology Department, and the Meyer Cancer Center of Weill Cornell Medicine of Cornell University, 1300 York Ave, New York, NY 10065
| |
Collapse
|
15
|
Identification and Expression Pattern of cyp26b1 Gene in Gonad of the Chinese Tongue Sole ( Cynoglossus semilaevis). Animals (Basel) 2022; 12:ani12192652. [PMID: 36230393 PMCID: PMC9559488 DOI: 10.3390/ani12192652] [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: 07/07/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022] Open
Abstract
Simple Summary In fish, it is obvious that the asynchronous development of the gonads and sexual dimorphism limit the development of aquaculture, so the research into sex-differentiation and gonadal growth is very important. Due to the sexual reversal phenomenon (genetic females becoming phenotypic males), the Chinese tongue sole (Cynoglossus semilaevis) is a great model for investigating sex-differentiation. Herein, we report one gene involved in sex-differentiation and gonadal growth of the Chinese Tongue Sole. The gene cyp26b1 (cytochrome P450 family 26 subfamily b member 1) is a metabolizing Retinoic Acid (RA) enzyme. Since it regulates RA to control sex determination and differentiation, cyp26b1 is considered a critical part of mammals’ ovary-antagonizing and testis-determining downstream passageway of Sry (sex-determining region Y) and Sox9 (sry-box transcription factor 9). In fish, the related research is reported only on the Japanese flounder (Paralichthys olivaceus) and zebrafish (Danio rerio). In the current investigation, the identification and expression pattern of the cyp26b1 gene in the Chinese tongue sole suggested that cyp26b1 might impact sex-differentiation and gonadal development. Abstract As an RA-metabolizing enzyme, cyp26b1 has a substantial impact on RA-signaling pathways. The cyp26b1 gene from the Chinese tongue sole was cloned and identified in this investigation. The cyp26b1 ORF was 1536 bp in length and encoded a 512 amino acid protein. A quantitative real-time PCR (qPCR) indicated that the cyp26b1 expression is no significant sexual dimorphism in the gonads at the 80 days post-hatching (dph) stages. After 4 months post-hatching (mph), the expression of cyp26b1 showed sexual dimorphism and lower level of expression in the ovaries than in the testes. An in situ hybridization demonstrated that cyp26b1 mRNA was primarily located in the testis. Interestingly, the cyp26b1 mRNA probe was also detected in the ovaries. These results suggested that cyp26b1 participates in the sex-differentiation and gonadal development of the Chinese tongue sole.
Collapse
|
16
|
Alhasnani MA, Loeb S, Hall SJ, Caruolo Z, Simmonds F, Solano AE, Spade DJ. Interaction between mono-(2-ethylhexyl) phthalate and retinoic acid alters Sertoli cell development during fetal mouse testis cord morphogenesis. Curr Res Toxicol 2022; 3:100087. [PMID: 36189433 PMCID: PMC9520016 DOI: 10.1016/j.crtox.2022.100087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/17/2022] [Accepted: 09/17/2022] [Indexed: 11/24/2022] Open
Abstract
Phthalic acid esters (phthalates) are a class of industrial chemicals that cause developmental and reproductive toxicity, but there are significant gaps in knowledge of phthalate toxicity mechanisms. There is evidence that phthalates disrupt retinoic acid signaling in the fetal testis, potentially disrupting control of spatial and temporal patterns of testis development. Our goal was to determine how a phthalate would interact with retinoic acid signaling during fetal mouse testis development. We hypothesized that mono-(2-ethylhexyl) phthalate (MEHP) would exacerbate the adverse effect of all-trans retinoic acid (ATRA) on seminiferous cord development in the mouse fetal testis. To test this hypothesis, gestational day (GD) 14 C57BL/6 mouse testes were isolated and cultured on media containing MEHP, ATRA, or a combination of both compounds. Cultured testes were collected for global transcriptome analysis after one day in culture and for histology and immunofluorescent analysis of Sertoli cell differentiation after three days in culture. ATRA disrupted seminiferous cord morphogenesis and induced aberrant FOXL2 expression. MEHP alone had no significant effect on cord development, but combined exposure to MEHP and ATRA increased the number of FOXL2-positive cells, reduced seminiferous cord number, and increased testosterone levels, beyond the effect of ATRA alone. In RNA-seq analysis, ATRA treatment and MEHP treatment resulted in differential expression of genes 510 and 134 genes, respectively, including 70 common differentially expressed genes (DEGs) between the two treatments, including genes with known roles in fetal testis development. MEHP DEGs included RAR target genes, genes involved in angiogenesis, and developmental patterning genes, including members of the homeobox superfamily. These results support the hypothesis that MEHP modulates retinoic acid signaling in the mouse fetal testis and provide insight into potential mechanisms by which phthalates disrupt seminiferous cord morphogenesis.
Collapse
Key Words
- ATRA, All-trans retinoic acid. CAS # 302-79-4
- DMSO, dimethyl sulfoxide
- Fetal testis development
- GD, gestational day
- GO, Gene Ontology
- IPA, Ingenuity Pathway Analysis
- ITCN, Image-based Tool for Counting Nuclei
- MEHP, mono-(2-ethylheyxl) phthalate. CAS # 4376-20-9
- MNGs, multinucleated germ cells
- PVC, polyvinyl chloride
- Phthalate toxicity
- Retinoic acid
- Sertoli cell
- TDS, testicular dysgenesis syndrome
Collapse
Affiliation(s)
- Maha A. Alhasnani
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Skylar Loeb
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Susan J. Hall
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Zachary Caruolo
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Faith Simmonds
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Amanda E. Solano
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| | - Daniel J. Spade
- Department of Pathology and Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912, USA
| |
Collapse
|
17
|
Svingen T, Schwartz CL, Rosenmai AK, Ramhøj L, Johansson HKL, Hass U, Draskau MK, Davidsen N, Christiansen S, Ballegaard ASR, Axelstad M. Using alternative test methods to predict endocrine disruption and reproductive adverse outcomes: do we have enough knowledge? ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 304:119242. [PMID: 35378198 DOI: 10.1016/j.envpol.2022.119242] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/12/2022] [Accepted: 03/29/2022] [Indexed: 06/14/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are a matter of great concern. They are ubiquitous in the environment, are considered harmful to humans and wildlife, yet remain challenging to identify based on current international test guidelines and regulatory frameworks. For a compound to be identified as an EDC within the EU regulatory system, a plausible link between an endocrine mode-of-action and an adverse effect outcome in an intact organism must be established. This requires in-depth knowledge about molecular pathways regulating normal development and function in animals and humans in order to elucidate causes for disease. Although our knowledge about the role of the endocrine system in animal development and function is substantial, it remains challenging to predict endocrine-related disease outcomes in intact animals based on non-animal test data. A main reason for this is that our knowledge about mechanism-of-action are still lacking for essential causal components, coupled with the sizeable challenge of mimicking the complex multi-organ endocrine system by methodological reductionism. Herein, we highlight this challenge by drawing examples from male reproductive toxicity, which is an area that has been at the forefront of EDC research since its inception. We discuss the importance of increased focus on characterizing mechanism-of-action for EDC-induced adverse health effects. This is so we can design more robust and reliable testing strategies using non-animal test methods for predictive toxicology; both to improve chemical risk assessment in general, but also to allow for considerable reduction and replacement of animal experiments in chemicals testing of the 21st Century.
Collapse
Affiliation(s)
- Terje Svingen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark.
| | | | | | - Louise Ramhøj
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| | | | - Ulla Hass
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| | - Monica Kam Draskau
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| | - Nichlas Davidsen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| | - Sofie Christiansen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| | | | - Marta Axelstad
- National Food Institute, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| |
Collapse
|
18
|
Mayère C, Regard V, Perea-Gomez A, Bunce C, Neirijnck Y, Djari C, Bellido-Carreras N, Sararols P, Reeves R, Greenaway S, Simon M, Siggers P, Condrea D, Kühne F, Gantar I, Tang F, Stévant I, Batti L, Ghyselinck NB, Wilhelm D, Greenfield A, Capel B, Chaboissier MC, Nef S. Origin, specification and differentiation of a rare supporting-like lineage in the developing mouse gonad. SCIENCE ADVANCES 2022; 8:eabm0972. [PMID: 35613264 PMCID: PMC10942771 DOI: 10.1126/sciadv.abm0972] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 04/11/2022] [Indexed: 06/15/2023]
Abstract
Gonadal sex determination represents a unique model for studying cell fate decisions. However, a complete understanding of the different cell lineages forming the developing testis and ovary remains elusive. Here, we investigated the origin, specification, and subsequent sex-specific differentiation of a previously uncharacterized population of supporting-like cells (SLCs) in the developing mouse gonads. The SLC lineage is closely related to the coelomic epithelium and specified as early as E10.5, making it the first somatic lineage to be specified in the bipotential gonad. SLC progenitors are localized within the genital ridge at the interface with the mesonephros and initially coexpress Wnt4 and Sox9. SLCs become sexually dimorphic around E12.5, progressively acquire a more Sertoli- or pregranulosa-like identity and contribute to the formation of the rete testis and rete ovarii. Last, we found that WNT4 is a crucial regulator of the SLC lineage and is required for normal development of the rete testis.
Collapse
Affiliation(s)
- Chloé Mayère
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
- iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Switzerland
| | - Violaine Regard
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
- iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Switzerland
| | - Aitana Perea-Gomez
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Corey Bunce
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yasmine Neirijnck
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Cyril Djari
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | | | - Pauline Sararols
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Richard Reeves
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Simon Greenaway
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Michelle Simon
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Pam Siggers
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Diana Condrea
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP1014267404 ILLKIRCH CEDEX, France
| | - Françoise Kühne
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Ivana Gantar
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Furong Tang
- Université Côte d’Azur, Inserm, CNRS, Institut de Biologie Valrose (iBV), 06108 Nice, France
| | - Isabelle Stévant
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
| | - Laura Batti
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Norbert B. Ghyselinck
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP1014267404 ILLKIRCH CEDEX, France
| | - Dagmar Wilhelm
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Andy Greenfield
- Mammalian Genetics Unit, Medical Research Council Harwell Institute, Oxfordshire OX11 0RD, UK
| | - Blanche Capel
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, 1211 Geneva, Switzerland
- iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, Switzerland
| |
Collapse
|
19
|
AOP Key Event Relationship report: Linking decreased retinoic acid levels with disrupted meiosis in developing oocytes. Curr Res Toxicol 2022; 3:100069. [PMID: 35345548 PMCID: PMC8957012 DOI: 10.1016/j.crtox.2022.100069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/24/2022] [Accepted: 03/17/2022] [Indexed: 12/03/2022] Open
Abstract
The first case study to develop and publish an individual KER as a stand-alone unit of information under the AOP framework overseen by the OECD. Full description of a KER linking decreased all-trans retinoic acid (atRA) levels in developing ovaries with disrupted meiotic entry of oogonia. KER described is associated with an intended AOP linking inhibition of the atRA producing ALDH1A enzymes with reduced fertility in women.
The Adverse Outcome Pathway (AOP) concept is an emerging tool in regulatory toxicology that uses simplified descriptions to show cause-effect relationships between stressors and toxicity outcomes in intact organisms. The AOP structure is a modular framework, with Key Event Relationships (KERs) representing the unit of causal relationship based on existing knowledge, describing the connection between two Key Events. Because KERs are the only unit to support inference it has been argued recently that KERs should be recognized as the core building blocks of knowledge assembly within the AOP-Knowledge Base. Herein, we present a first case to support this proposal and provide a full description of a KER linking decreased all-trans retinoic acid (atRA) levels in developing ovaries with disrupted meiotic entry of oogonia. We outline the evidence to support a role for atRA in inducing meiosis in oogonia across mammals; this is important because elements of the RA synthesis/degradation pathway are recognized targets for numerous environmental chemicals. The KER we describe will be used to support an intended AOP linking inhibition of the atRA producing ALDH1A enzymes with reduced fertility in women.
Collapse
|
20
|
Kulibin AY, Malolina EA. The Rete Testis: Development and Role in Testis Function. Russ J Dev Biol 2021. [DOI: 10.1134/s1062360421060072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Abstract
The rete testis connects seminiferous tubules in which germ cells develop to the efferent ducts and the epididymis, where gametes mature and gain mobility. Several recent studies have thoroughly explored the morphogenesis of this structure in mice during embryonic and postnatal periods. A part of the rete testis has been shown to derive from the precursors of gonad somatic cells before sex determination. The other part forms from embryonal Sertoli cells of testis cords adjacent to the mesonephros. The transformation of Sertoli cells into rete testis cells is apparently not limited to the embryonic stage of development and continues during postnatal testis development. Recently, it was found that the rete testis participates in the formation and maintenance of specialized Sertoli cells in terminal segments of seminiferous tubules, transitional zones. Current views suggest that the transitional zones of the seminiferous tubules may represent a niche for spermatogonial stem cells, the site of the prolonged proliferation of Sertoli cells in the pubertal and postpubertal periods of testis development, and also could be a generator of spermatogenic waves. To sum up, the rete testis transports gametes from the testis to the epididymis, maintains pressure within seminiferous tubules, regulates the composition of the testicular fluid, and impacts the spermatogenic process itself.
Collapse
|
21
|
Estermann MA, Major AT, Smith CA. Genetic Regulation of Avian Testis Development. Genes (Basel) 2021; 12:1459. [PMID: 34573441 PMCID: PMC8470383 DOI: 10.3390/genes12091459] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 11/30/2022] Open
Abstract
As in other vertebrates, avian testes are the site of spermatogenesis and androgen production. The paired testes of birds differentiate during embryogenesis, first marked by the development of pre-Sertoli cells in the gonadal primordium and their condensation into seminiferous cords. Germ cells become enclosed in these cords and enter mitotic arrest, while steroidogenic Leydig cells subsequently differentiate around the cords. This review describes our current understanding of avian testis development at the cell biology and genetic levels. Most of this knowledge has come from studies on the chicken embryo, though other species are increasingly being examined. In chicken, testis development is governed by the Z-chromosome-linked DMRT1 gene, which directly or indirectly activates the male factors, HEMGN, SOX9 and AMH. Recent single cell RNA-seq has defined cell lineage specification during chicken testis development, while comparative studies point to deep conservation of avian testis formation. Lastly, we identify areas of future research on the genetics of avian testis development.
Collapse
Affiliation(s)
| | | | - Craig Allen Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (M.A.E.); (A.T.M.)
| |
Collapse
|
22
|
Jiménez R, Burgos M, Barrionuevo FJ. Sex Maintenance in Mammals. Genes (Basel) 2021; 12:genes12070999. [PMID: 34209938 PMCID: PMC8303465 DOI: 10.3390/genes12070999] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/26/2021] [Accepted: 06/26/2021] [Indexed: 12/30/2022] Open
Abstract
The crucial event in mammalian sexual differentiation occurs at the embryonic stage of sex determination, when the bipotential gonads differentiate as either testes or ovaries, according to the sex chromosome constitution of the embryo, XY or XX, respectively. Once differentiated, testes produce sexual hormones that induce the subsequent differentiation of the male reproductive tract. On the other hand, the lack of masculinizing hormones in XX embryos permits the formation of the female reproductive tract. It was long assumed that once the gonad is differentiated, this developmental decision is irreversible. However, several findings in the last decade have shown that this is not the case and that a continuous sex maintenance is needed. Deletion of Foxl2 in the adult ovary lead to ovary-to-testis transdifferentiation and deletion of either Dmrt1 or Sox9/Sox8 in the adult testis induces the opposite process. In both cases, mutant gonads were genetically reprogrammed, showing that both the male program in ovaries and the female program in testes must be actively repressed throughout the individual's life. In addition to these transcription factors, other genes and molecular pathways have also been shown to be involved in this antagonism. The aim of this review is to provide an overview of the genetic basis of sex maintenance once the gonad is already differentiated.
Collapse
|
23
|
Sararols P, Stévant I, Neirijnck Y, Rebourcet D, Darbey A, Curley MK, Kühne F, Dermitzakis E, Smith LB, Nef S. Specific Transcriptomic Signatures and Dual Regulation of Steroidogenesis Between Fetal and Adult Mouse Leydig Cells. Front Cell Dev Biol 2021; 9:695546. [PMID: 34262907 PMCID: PMC8273516 DOI: 10.3389/fcell.2021.695546] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/08/2021] [Indexed: 12/20/2022] Open
Abstract
Leydig cells (LC) are the main testicular androgen-producing cells. In eutherian mammals, two types of LCs emerge successively during testicular development, fetal Leydig cells (FLCs) and adult Leydig cells (ALCs). Both display significant differences in androgen production and regulation. Using bulk RNA sequencing, we compared the transcriptomes of both LC populations to characterize their specific transcriptional and functional features. Despite similar transcriptomic profiles, a quarter of the genes show significant variations in expression between FLCs and ALCs. Non-transcriptional events, such as alternative splicing was also observed, including a high rate of intron retention in FLCs compared to ALCs. The use of single-cell RNA sequencing data also allowed the identification of nine FLC-specific genes and 50 ALC-specific genes. Expression of the corticotropin-releasing hormone 1 (Crhr1) receptor and the ACTH receptor melanocortin type 2 receptor (Mc2r) specifically in FLCs suggests a dual regulation of steroidogenesis. The androstenedione synthesis by FLCs is stimulated by luteinizing hormone (LH), corticotrophin-releasing hormone (CRH), and adrenocorticotropic hormone (ACTH) whereas the testosterone synthesis by ALCs is dependent exclusively on LH. Overall, our study provides a useful database to explore LC development and functions.
Collapse
Affiliation(s)
- Pauline Sararols
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Isabelle Stévant
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Yasmine Neirijnck
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Diane Rebourcet
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW, Australia
| | - Annalucia Darbey
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW, Australia
| | - Michael K Curley
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Françoise Kühne
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Emmanouil Dermitzakis
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Faculty of Medicine, Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
| | - Lee B Smith
- College of Engineering, Science and Environment, The University of Newcastle, Callaghan, NSW, Australia.,Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Serge Nef
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
24
|
Rore H, Owen N, Piña-Aguilar RE, Docherty K, Sekido R. Testicular somatic cell-like cells derived from embryonic stem cells induce differentiation of epiblasts into germ cells. Commun Biol 2021; 4:802. [PMID: 34183774 PMCID: PMC8239049 DOI: 10.1038/s42003-021-02322-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 06/09/2021] [Indexed: 02/06/2023] Open
Abstract
Regeneration of the testis from pluripotent stem cells is a real challenge, reflecting the complexity of the interaction of germ cells and somatic cells. Here we report the generation of testicular somatic cell-like cells (TesLCs) including Sertoli cell-like cells (SCLCs) from mouse embryonic stem cells (ESCs) in xeno-free culture. We find that Nr5a1/SF1 is critical for interaction between SCLCs and PGCLCs. Intriguingly, co-culture of TesLCs with epiblast-like cells (EpiLCs), rather than PGCLCs, results in self-organised aggregates, or testicular organoids. In the organoid, EpiLCs differentiate into PGCLCs or gonocyte-like cells that are enclosed within a seminiferous tubule-like structure composed of SCLCs. Furthermore, conditioned medium prepared from TesLCs has a robust inducible activity to differentiate EpiLCs into PGCLCs. Our results demonstrate conditions for in vitro reconstitution of a testicular environment from ESCs and provide further insights into the generation of sperm entirely in xeno-free culture.
Collapse
Affiliation(s)
- Holly Rore
- Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen, UK
| | - Nicholas Owen
- Institute of Ophthalmology, University College London, London, UK
| | | | - Kevin Docherty
- Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen, UK
| | - Ryohei Sekido
- Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen, UK.
- Institute of Ophthalmology, University College London, London, UK.
| |
Collapse
|
25
|
Banisch TU, Slaidina M, Gupta S, Ho M, Gilboa L, Lehmann R. A transitory signaling center controls timing of primordial germ cell differentiation. Dev Cell 2021; 56:1742-1755.e4. [PMID: 34081907 PMCID: PMC8330407 DOI: 10.1016/j.devcel.2021.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 03/07/2021] [Accepted: 05/10/2021] [Indexed: 12/16/2022]
Abstract
Organogenesis requires exquisite spatiotemporal coordination of cell morphogenesis, migration, proliferation, and differentiation of multiple cell types. For gonads, this involves complex interactions between somatic and germline tissues. During Drosophila ovary morphogenesis, primordial germ cells (PGCs) either are sequestered in stem cell niches and are maintained in an undifferentiated germline stem cell state or transition directly toward differentiation. Here, we identify a mechanism that links hormonal triggers of somatic tissue morphogenesis with PGC differentiation. An early ecdysone pulse initiates somatic swarm cell (SwC) migration, positioning these cells close to PGCs. A second hormone peak activates Torso-like signal in SwCs, which stimulates the Torso receptor tyrosine kinase (RTK) signaling pathway in PGCs promoting their differentiation by de-repression of the differentiation gene, bag of marbles. Thus, systemic temporal cues generate a transitory signaling center that coordinates ovarian morphogenesis with stem cell self-renewal and differentiation programs, highlighting a more general role for such centers in reproductive and developmental biology.
Collapse
Affiliation(s)
- Torsten U Banisch
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA.
| | - Maija Slaidina
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Selena Gupta
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Megan Ho
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Lilach Gilboa
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ruth Lehmann
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
26
|
Dai S, Qi S, Wei X, Liu X, Li Y, Zhou X, Xiao H, Lu B, Wang D, Li M. Germline sexual fate is determined by the antagonistic action of dmrt1 and foxl3/foxl2 in tilapia. Development 2021; 148:dev.199380. [PMID: 33741713 DOI: 10.1242/dev.199380] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/04/2021] [Indexed: 12/21/2022]
Abstract
Germline sexual fate has long been believed to be determined by the somatic environment, but this idea is challenged by recent studies of foxl3 mutants in medaka. Here, we demonstrate that the sexual fate of tilapia germline is determined by the antagonistic interaction of dmrt1 and foxl3, which are transcriptionally repressed in male and female germ cells, respectively. Loss of dmrt1 rescued the germ cell sex reversal in foxl3Δ7/Δ7 XX fish, and loss of foxl3 partially rescued germ cell sex reversal but not somatic cell fate in dmrt1Δ5/Δ5 XY fish. Interestingly, germ cells lost sexual plasticity in dmrt1Δ5/Δ5 XY and foxl3Δ7/Δ7 XX single mutants, as aromatase inhibitor (AI) and estrogen treatment failed to rescue the respective phenotypes. However, recovery of germ cell sexual plasticity was observed in dmrt1/foxl3 double mutants. Importantly, mutation of somatic cell-specific foxl2 resulted in testicular development in foxl3Δ7/Δ7 or dmrt1Δ5/Δ5 mutants. Our findings demonstrate that sexual plasticity of germ cells relies on the presence of both dmrt1 and foxl3. The existence of dmrt1 and foxl3 allows environmental factors to influence the sex fate decision in vertebrates.
Collapse
Affiliation(s)
- Shengfei Dai
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Shuangshuang Qi
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xueyan Wei
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xingyong Liu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Yibing Li
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Xin Zhou
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Hesheng Xiao
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Baoyue Lu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Deshou Wang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Minghui Li
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
27
|
Feng CW, Burnet G, Spiller CM, Cheung FKM, Chawengsaksophak K, Koopman P, Bowles J. Identification of regulatory elements required for Stra8 expression in fetal ovarian germ cells of the mouse. Development 2021; 148:dev.194977. [PMID: 33574039 DOI: 10.1242/dev.194977] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 02/04/2021] [Indexed: 12/14/2022]
Abstract
In mice, the entry of germ cells into meiosis crucially depends on the expression of stimulated by retinoic acid gene 8 (Stra8). Stra8 is expressed specifically in pre-meiotic germ cells of females and males, at fetal and postnatal stages, respectively, but the mechanistic details of its spatiotemporal regulation are yet to be defined. In particular, there has been considerable debate regarding whether retinoic acid is required, in vivo, to initiate Stra8 expression in the mouse fetal ovary. We show that the distinctive anterior-to-posterior pattern of Stra8 initiation, characteristic of germ cells in the fetal ovary, is faithfully recapitulated when 2.9 kb of the Stra8 promoter is used to drive eGFP expression. Using in vitro transfection assays of cutdown and mutant constructs, we identified two functional retinoic acid responsive elements (RAREs) within this 2.9 kb regulatory element. We also show that the transcription factor DMRT1 enhances Stra8 expression, but only in the presence of RA and the most proximal RARE. Finally, we used CRISPR/Cas9-mediated targeted mutation studies to demonstrate that both RAREs are required for optimal Stra8 expression levels in vivo.
Collapse
Affiliation(s)
- Chun-Wei Feng
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Guillaume Burnet
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Cassy M Spiller
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Fiona Ka Man Cheung
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Kallayanee Chawengsaksophak
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia.,Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i. Vídenská 1083, 4 14220 Prague, Czech Republic
| | - Peter Koopman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Josephine Bowles
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia .,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
28
|
Radovic Pletikosic SM, Starovlah IM, Miljkovic D, Bajic DM, Capo I, Nef S, Kostic TS, Andric SA. Deficiency in insulin-like growth factors signalling in mouse Leydig cells increase conversion of testosterone to estradiol because of feminization. Acta Physiol (Oxf) 2021; 231:e13563. [PMID: 32975906 DOI: 10.1111/apha.13563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
Abstract
AIM A growing body of evidence pointed correlation between insulin-resistance, testosterone level and infertility, but there is scarce information about mechanisms. The aim of this study was to identify the possible mechanism linking the insulin-resistance with testosterone-producing-Leydig-cells functionality. METHODS We applied in vivo and in vitro approaches. The in vivo model of functional genomics is represented by INSR/IGF1R-deficient-testosterone-producing Leydig cells obtained from the prepubertal (P21) and adult (P80) male mice with insulin + IGF1-receptors deletion in steroidogenic cells (Insr/Igf1r-DKO). The in vitro model of INSR/IGF1R-deficient-cell was mimicked by blockade of insulin/IGF1-receptors on the primary culture of P21 and P80 Leydig cells. RESULTS Leydig-cell-specific-insulin-resistance induce the development of estrogenic characteristics of progenitor Leydig cells in prepubertal mice and mature Leydig cells in adult mice, followed with a dramatic reduction of androgen phenotype. Level of androgens in serum, testes and Leydig cells decrease as a consequence of the dramatic reduction of steroidogenic capacity and activity as well as all functional markers of Leydig cell. Oppositely, the markers for female-steroidogenic-cell differentiation and function increase. The physiological significances are the higher level of testosterone-to-estradiol-conversion in double-knock-out-mice of both ages and few spermatozoa in adults. Intriguingly, the transcription of pro-male sexual differentiation markers Sry/Sox9 increased in P21-Leydig-cells, questioning the current view about the antagonistic genetic programs underlying gonadal sex determination. CONCLUSION The results provide new molecular mechanisms leading to the development of the female phenotype in Leydig cells from Insr/Igf1r-DKO mice and could help to better understand the correlation between insulin resistance, testosterone and male (in)fertility.
Collapse
Affiliation(s)
- Sava M. Radovic Pletikosic
- Laboratory for Reproductive Endocrinology and Signalling Laboratory for Chronobiology and Aging CeRES DBE Faculty of Sciences University of Novi Sad Novi Sad Serbia
| | - Isidora M. Starovlah
- Laboratory for Reproductive Endocrinology and Signalling Laboratory for Chronobiology and Aging CeRES DBE Faculty of Sciences University of Novi Sad Novi Sad Serbia
| | - Dejan Miljkovic
- Center for Medical‐Pharmaceutical Research and Quality Control Department for Histology and Embryology Faculty of Medicine University of Novi Sad Novi Sad Serbia
| | - Dragana M. Bajic
- Laboratory for Reproductive Endocrinology and Signalling Laboratory for Chronobiology and Aging CeRES DBE Faculty of Sciences University of Novi Sad Novi Sad Serbia
| | - Ivan Capo
- Center for Medical‐Pharmaceutical Research and Quality Control Department for Histology and Embryology Faculty of Medicine University of Novi Sad Novi Sad Serbia
| | - Serge Nef
- Department of Genetic Medicine and Development Medical Faculty University of Geneva Geneva Switzerland
| | - Tatjana S. Kostic
- Laboratory for Reproductive Endocrinology and Signalling Laboratory for Chronobiology and Aging CeRES DBE Faculty of Sciences University of Novi Sad Novi Sad Serbia
| | - Silvana A. Andric
- Laboratory for Reproductive Endocrinology and Signalling Laboratory for Chronobiology and Aging CeRES DBE Faculty of Sciences University of Novi Sad Novi Sad Serbia
| |
Collapse
|
29
|
Draskau MK, Spiller CM, Boberg J, Bowles J, Svingen T. Developmental biology meets toxicology: contributing reproductive mechanisms to build adverse outcome pathways. Mol Hum Reprod 2021; 26:111-116. [PMID: 31943113 DOI: 10.1093/molehr/gaaa001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
An adverse outcome pathway (AOP) is a simplified description of the sequence of mechanistic events that lead to a particular toxicological effect, from initial trigger to adverse outcome. Although designed to inform regulatory risk assessors, the AOP framework also provides a platform for innovative collaborations between experts from relevant research fields and the regulatory community. The underpinning for any AOP is basic knowledge about molecular and developmental processes; such knowledge can only be attained by solid bioscientific research. Starting with this fundamental knowledge, the objective is to devise novel testing strategies that focus on key events in a causative pathway. It is anticipated that such a knowledge-based approach will ultimately alleviate many of the burdens associated with classical chemical testing strategies that typically involve large-scale animal toxicity regimens. This hails from the notion that a solid understanding of the underlying mechanisms will allow the development and use of alternative test methods, including both in vitro and in silico approaches. This review is specifically targeted at professionals working in bioscientific fields, such as developmental and reproductive biology, and aims to (i) inform on the existence of the AOP framework and (ii) encourage new cross-disciplinary collaborations. It is hoped that fundamental biological knowledge can thus be better exploited for applied purposes: firstly, an improved understanding of how our perpetual exposure to environmental chemicals is causing human reproductive disease and, secondly, new approaches to screen for harmful chemicals more efficiently. This is not an instructional manual on how to create AOPs; rather, we discuss how to harness fundamental knowledge from the biosciences to assist regulatory toxicologists in their efforts to protect humans against chemicals that harm human reproductive development and function.
Collapse
Affiliation(s)
- Monica Kam Draskau
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Cassy M Spiller
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Julie Boberg
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Josephine Bowles
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Terje Svingen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
30
|
Nagahama Y, Chakraborty T, Paul-Prasanth B, Ohta K, Nakamura M. Sex determination, gonadal sex differentiation, and plasticity in vertebrate species. Physiol Rev 2020; 101:1237-1308. [PMID: 33180655 DOI: 10.1152/physrev.00044.2019] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A diverse array of sex determination (SD) mechanisms, encompassing environmental to genetic, have been found to exist among vertebrates, covering a spectrum from fixed SD mechanisms (mammals) to functional sex change in fishes (sequential hermaphroditic fishes). A major landmark in vertebrate SD was the discovery of the SRY gene in 1990. Since that time, many attempts to clone an SRY ortholog from nonmammalian vertebrates remained unsuccessful, until 2002, when DMY/dmrt1by was discovered as the SD gene of a small fish, medaka. Surprisingly, however, DMY/dmrt1by was found in only 2 species among more than 20 species of medaka, suggesting a large diversity of SD genes among vertebrates. Considerable progress has been made over the last 3 decades, such that it is now possible to formulate reasonable paradigms of how SD and gonadal sex differentiation may work in some model vertebrate species. This review outlines our current understanding of vertebrate SD and gonadal sex differentiation, with a focus on the molecular and cellular mechanisms involved. An impressive number of genes and factors have been discovered that play important roles in testicular and ovarian differentiation. An antagonism between the male and female pathway genes exists in gonads during both sex differentiation and, surprisingly, even as adults, suggesting that, in addition to sex-changing fishes, gonochoristic vertebrates including mice maintain some degree of gonadal sexual plasticity into adulthood. Importantly, a review of various SD mechanisms among vertebrates suggests that this is the ideal biological event that can make us understand the evolutionary conundrums underlying speciation and species diversity.
Collapse
Affiliation(s)
- Yoshitaka Nagahama
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki, Japan.,South Ehime Fisheries Research Center, Ehime University, Ainan, Japan.,Faculty of Biological Science and Technology, Kanazawa University, Ishikawa, Japan
| | - Tapas Chakraborty
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki, Japan.,South Ehime Fisheries Research Center, Ehime University, Ainan, Japan.,Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukouka, Japan.,Karatsu Satellite of Aqua-Bioresource Innovation Center, Kyushu University, Karatsu, Japan
| | - Bindhu Paul-Prasanth
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki, Japan.,Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidapeetham, Kochi, Kerala, India
| | - Kohei Ohta
- Laboratory of Marine Biology, Faculty of Agriculture, Kyushu University, Fukouka, Japan
| | - Masaru Nakamura
- Sesoko Station, Tropical Biosphere Research Center, University of the Ryukyus, Okinawa, Japan.,Research Center, Okinawa Churashima Foundation, Okinawa, Japan
| |
Collapse
|
31
|
Yan RG, Li BY, Yang QE. Function and transcriptomic dynamics of Sertoli cells during prospermatogonia development in mouse testis. Reprod Biol 2020; 20:525-535. [PMID: 32952085 DOI: 10.1016/j.repbio.2020.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 09/02/2020] [Accepted: 09/05/2020] [Indexed: 10/23/2022]
Abstract
In mammals, spermatogonial stem cells (SSCs) arise from a subpopulation of prospermatogonia during neonatal testis development. Currently, molecular mechanisms directing the prospermatogonia to spermatogonial transition are not well understood. In the study, we found that reducing Sertoli cells number by Amh-cre mediated expression of diphtheria toxin (AC;DTA) in murine fetal testis caused defects in prospermatogonia fate decisions. Histological and immunohistochemical analyses confirmed that Sertoli cells loss occurred at embryonic day (E) 14.5. Prospermatogonia maintained mitotic arrest at E16.5 in control animals, in contrast, 13.4% of germ cells in AC;DTA testis reentered cell cycle and expressed gH2A.X and Sycp3, indicating the commitment to meiosis. After birth, the number of prospermatogonia resuming mitosis was significantly affected by Sertoli cell loss in AC;DTA animals. Lastly, we isolated primary Sertoli cells using a Sertoli cell specific GFP reporter line and showed dynamics of Sertoli cell transcriptomes at E12.5, E13.5, E16.5 and P1. By further analysis, we revealed unique gene expression patterns and potential candidate genes regulating Sertoli cell development and likely mediating interactions between Sertoli cells, prospermatogonia and other testicular cells.
Collapse
Affiliation(s)
- Rong-Ge Yan
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, QH, 810001, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bin-Ye Li
- Center for Reproductive Medicine, Qinghai Provincial People's Hospital, Xining, QH, 81001, China
| | - Qi-En Yang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, QH, 810001, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, QH, 810001, China.
| |
Collapse
|
32
|
Kulibin AY, Malolina EA. Formation of the rete testis during mouse embryonic development. Dev Dyn 2020; 249:1486-1499. [DOI: 10.1002/dvdy.242] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/18/2020] [Accepted: 08/22/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
- Andrey Yu. Kulibin
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences Moscow Russia
| | - Ekaterina A. Malolina
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences Moscow Russia
| |
Collapse
|
33
|
Putative adverse outcome pathways for female reproductive disorders to improve testing and regulation of chemicals. Arch Toxicol 2020; 94:3359-3379. [PMID: 32638039 PMCID: PMC7502037 DOI: 10.1007/s00204-020-02834-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022]
Abstract
Modern living challenges female reproductive health. We are witnessing a rise in reproductive disorders and drop in birth rates across the world. The reasons for these manifestations are multifaceted and most likely include continuous exposure to an ever-increasing number of chemicals. The cause–effect relationships between chemical exposure and female reproductive disorders, however, have proven problematic to determine. This has made it difficult to assess the risks chemical exposures pose to a woman’s reproductive development and function. To address this challenge, this review uses the adverse outcome pathway (AOP) concept to summarize current knowledge about how chemical exposure can affect female reproductive health. We have a special focus on effects on the ovaries, since they are essential for lifelong reproductive health in women, being the source of both oocytes and several reproductive hormones, including sex steroids. The AOP framework is widely accepted as a new tool for toxicological safety assessment that enables better use of mechanistic knowledge for regulatory purposes. AOPs equip assessors and regulators with a pragmatic network of linear cause–effect relationships, enabling the use of a wider range of test method data in chemical risk assessment and regulation. Based on current knowledge, we propose ten putative AOPs relevant for female reproductive disorders that can be further elaborated and potentially be included in the AOPwiki. This effort is an important step towards better safeguarding the reproductive health of all girls and women.
Collapse
|
34
|
Zeng M, Dai X, Liang Z, Sun R, Huang S, Luo L, Li Z. Critical roles of mRNA m 6A modification and YTHDC2 expression for meiotic initiation and progression in female germ cells. Gene 2020; 753:144810. [PMID: 32470506 DOI: 10.1016/j.gene.2020.144810] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/30/2020] [Accepted: 05/22/2020] [Indexed: 11/17/2022]
Abstract
Meiotic entry and progression require dynamic regulation of germline gene expression. m6A on mRNAs and recognition by YTHDC2 has been known as post-transcriptional regulatory complex, but the roles of this regulator remain unclear for meiotic initiation and progression in female germ cells (FGCs). This study showed that m6A modification occurred mainly in FGCs rather than ovarian somatic cells (SOMAs), and m6A levels in FGCs increased significantly with meiotic initiation. m6A inhibition suppressed expression of the meiotic markers and affected the percent of FGCs at zygotene, pachytene and diplotene stage respectively. YTHDC2 expression also increased in the same pattern with m6A. Ythdc2 knockdown decreased the percent of STRA8-positive FGCs and altered the percent of FGCs at zygotene and pachytene stage respectively. Taken together, these results suggest that mRNA m6A modification and YTHDC2 expression are essential for meiotic initiation and progression in FGCs.
Collapse
Affiliation(s)
- Ming Zeng
- Medical Research Institute, Baoan Women's and Children's Hospital, Jinan University, Shenzhen 518133, China
| | - Xin Dai
- Medical Research Institute, Baoan Women's and Children's Hospital, Jinan University, Shenzhen 518133, China
| | - Zhibing Liang
- Medical Research Institute, Baoan Women's and Children's Hospital, Jinan University, Shenzhen 518133, China
| | - Ruliang Sun
- Department of Pathology, Baoan Women's and Children's Hospital, Jinan University, Shenzhen 518133, China
| | - Sui Huang
- Department of Pathology, Baoan Women's and Children's Hospital, Jinan University, Shenzhen 518133, China
| | - Liangping Luo
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Zhongxiang Li
- Medical Research Institute, Baoan Women's and Children's Hospital, Jinan University, Shenzhen 518133, China.
| |
Collapse
|
35
|
Vernet N, Condrea D, Mayere C, Féret B, Klopfenstein M, Magnant W, Alunni V, Teletin M, Souali-Crespo S, Nef S, Mark M, Ghyselinck NB. Meiosis occurs normally in the fetal ovary of mice lacking all retinoic acid receptors. SCIENCE ADVANCES 2020; 6:eaaz1139. [PMID: 32917583 PMCID: PMC7244263 DOI: 10.1126/sciadv.aaz1139] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/13/2020] [Indexed: 05/27/2023]
Abstract
Gametes are generated through a specialized cell differentiation process, meiosis, which, in ovaries of most mammals, is initiated during fetal life. All-trans retinoic acid (ATRA) is considered as the molecular signal triggering meiosis initiation. In the present study, we analyzed female fetuses ubiquitously lacking all ATRA nuclear receptors (RAR), obtained through a tamoxifen-inducible cre recombinase-mediated gene targeting approach. Unexpectedly, mutant oocytes robustly expressed meiotic genes, including the meiotic gatekeeper STRA8. In addition, ovaries from mutant fetuses grafted into adult recipient females yielded offspring bearing null alleles for all Rar genes. Thus, our results show that RAR are fully dispensable for meiotic initiation, as well as for the production of functional oocytes. Assuming that the effects of ATRA all rely on RAR, our study goes against the current model according to which meiosis is triggered by endogenous ATRA in the developing ovary. It therefore revives the search for the meiosis-inducing substance.
Collapse
Affiliation(s)
- Nadège Vernet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Diana Condrea
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Chloé Mayere
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Betty Féret
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Muriel Klopfenstein
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - William Magnant
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Violaine Alunni
- GenomEast platform, France Génomique consortium, IGBMC, 1 rue Laurent Fries, F-67404 Illkirch Cedex, France
| | - Marius Teletin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
- Service de Biologie de la Reproduction, Hôpitaux Universitaires de Strasbourg (HUS), France
| | - Sirine Souali-Crespo
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Manuel Mark
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France
- Service de Biologie de la Reproduction, Hôpitaux Universitaires de Strasbourg (HUS), France
| | - Norbert B Ghyselinck
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, BP-10142, F-67404 Illkirch Cedex, France.
| |
Collapse
|
36
|
Spade DJ, Hall SJ, Wortzel JD, Reyes G, Boekelheide K. All-trans Retinoic Acid Disrupts Development in Ex Vivo Cultured Fetal Rat Testes. II: Modulation of Mono-(2-ethylhexyl) Phthalate Toxicity. Toxicol Sci 2020; 168:149-159. [PMID: 30476341 DOI: 10.1093/toxsci/kfy283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Humans are universally exposed to low levels of phthalate esters (phthalates), which are used to plasticize polyvinyl chloride. Phthalates exert adverse effects on the development of seminiferous cords in the fetal testis through unknown toxicity pathways. To investigate the hypothesis that phthalates alter seminiferous cord development by disrupting retinoic acid (RA) signaling in the fetal testis, gestational day 15 fetal rat testes were exposed for 1-3 days to 10-6 M all-trans retinoic acid (ATRA) alone or in combination with 10-6-10-4 M mono-(2-ethylhexyl) phthalate (MEHP) in ex vivo culture. As previously reported, exogenous ATRA reduced seminiferous cord number. This effect was attenuated in a concentration-dependent fashion by MEHP co-exposure. ATRA and MEHP-exposed testes were depleted of DDX4-positive germ cells but not Sertoli cells. MEHP alone enhanced the expression of the RA receptor target Rbp1 and the ovary development-associated genes Wnt4 and Nr0b1, and suppressed expression of the Leydig cell marker, Star, and the germ cell markers, Ddx4 and Pou5f1. In co-exposures, MEHP predominantly enhanced the gene expression effects of ATRA, but the Wnt4 and Nr0b1 concentration-responses were nonlinear. Similarly, ATRA increased the number of cells expressing the granulosa cell marker FOXL2 in testis cultures, but this induction was attenuated by addition of MEHP. These results indicate that MEHP can both enhance and inhibit actions of ATRA during fetal testis development and provide evidence that RA signaling is a target for phthalate toxicity in the fetal testis.
Collapse
Affiliation(s)
- Daniel J Spade
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| | - Susan J Hall
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| | - Jeremy D Wortzel
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| | - Gerardo Reyes
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912.,Division of Natural Sciences, College of Mount Saint Vincent, Riverdale, New York 10471
| | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
37
|
Roberts C. Regulating Retinoic Acid Availability during Development and Regeneration: The Role of the CYP26 Enzymes. J Dev Biol 2020; 8:jdb8010006. [PMID: 32151018 PMCID: PMC7151129 DOI: 10.3390/jdb8010006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/17/2020] [Accepted: 02/17/2020] [Indexed: 12/16/2022] Open
Abstract
This review focuses on the role of the Cytochrome p450 subfamily 26 (CYP26) retinoic acid (RA) degrading enzymes during development and regeneration. Cyp26 enzymes, along with retinoic acid synthesising enzymes, are absolutely required for RA homeostasis in these processes by regulating availability of RA for receptor binding and signalling. Cyp26 enzymes are necessary to generate RA gradients and to protect specific tissues from RA signalling. Disruption of RA homeostasis leads to a wide variety of embryonic defects affecting many tissues. Here, the function of CYP26 enzymes is discussed in the context of the RA signalling pathway, enzymatic structure and biochemistry, human genetic disease, and function in development and regeneration as elucidated from animal model studies.
Collapse
Affiliation(s)
- Catherine Roberts
- Developmental Biology of Birth Defects, UCL-GOS Institute of Child Health, 30 Guilford St, London WC1N 1EH, UK;
- Institute of Medical and Biomedical Education St George’s, University of London, Cranmer Terrace, Tooting, London SW17 0RE, UK
| |
Collapse
|
38
|
Divergent Roles of CYP26B1 and Endogenous Retinoic Acid in Mouse Fetal Gonads. Biomolecules 2019; 9:biom9100536. [PMID: 31561560 PMCID: PMC6843241 DOI: 10.3390/biom9100536] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/17/2019] [Accepted: 09/24/2019] [Indexed: 11/19/2022] Open
Abstract
In female mammals, germ cells enter meiosis in the fetal ovaries, while in males, meiosis is prevented until postnatal development. Retinoic acid (RA) is considered the main inducer of meiotic entry, as it stimulates Stra8 which is required for the mitotic/meiotic switch. In fetal testes, the RA-degrading enzyme CYP26B1 prevents meiosis initiation. However, the role of endogenous RA in female meiosis entry has never been demonstrated in vivo. In this study, we demonstrate that some effects of RA in mouse fetal gonads are not recapitulated by the invalidation or up-regulation of CYP26B1. In organ culture of fetal testes, RA stimulates testosterone production and inhibits Sertoli cell proliferation. In the ovaries, short-term inhibition of RA-signaling does not decrease Stra8 expression. We develop a gain-of-function model to express CYP26A1 or CYP26B1. Only CYP26B1 fully prevents STRA8 induction in female germ cells, confirming its role as part of the meiotic prevention machinery. CYP26A1, a very potent RA degrading enzyme, does not impair the formation of STRA8-positive cells, but decreases Stra8 transcription. Collectively, our data reveal that CYP26B1 has other activities apart from metabolizing RA in fetal gonads and suggest a role of endogenous RA in amplifying Stra8, rather than being the initial inducer of Stra8. These findings should reactivate the quest to identify meiotic preventing or inducing substances.
Collapse
|
39
|
Yadu N, Kumar PG. Retinoic acid signaling in regulation of meiosis during embryonic development in mice. Genesis 2019; 57:e23327. [PMID: 31313882 DOI: 10.1002/dvg.23327] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 06/23/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022]
Abstract
In the embryonic gonads of mice, the genetic and epigenetic regulatory programs for germ cell sex specification and meiosis induction or suppression are intertwined. The quest for garnering comprehensive understanding of these programs has led to the emergence of retinoic acid (RA) as an important extrinsic factor, which regulates initiation of meiosis in female fetal germ cells that have attained a permissive epigenetic ground state. In contrast, germ cells in fetal testis are protected from the exposure to RA due to the activity of CYP26B1, an RA metabolizing enzyme, which is highly expressed in fetal testis. In this review, we provide an overview of the molecular mechanisms operating in fetal gonads of mice, which enable regulation of meiosis via RA signaling.
Collapse
Affiliation(s)
- Nomesh Yadu
- Division of Molecular Reproduction, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Pradeep G Kumar
- Division of Molecular Reproduction, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
40
|
Macdonald J, Kilcoyne KR, Sharpe RM, Kavanagh Á, Anderson RA, Brown P, Smith LB, Jørgensen A, Mitchell RT. DMRT1 repression using a novel approach to genetic manipulation induces testicular dysgenesis in human fetal gonads. Hum Reprod 2019; 33:2107-2121. [PMID: 30272154 PMCID: PMC6195803 DOI: 10.1093/humrep/dey289] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/04/2018] [Indexed: 01/16/2023] Open
Abstract
STUDY QUESTION Does loss of DMRT1 in human fetal testis alter testicular development and result in testicular dysgenesis? SUMMARY ANSWER DMRT1 repression in human fetal testis alters the expression of key testicular and ovarian determining genes, and leads to focal testicular dysgenesis. WHAT IS KNOWN ALREADY Testicular dysgenesis syndrome (TDS) is associated with common testicular disorders in young men, but its etiology is unknown. DMRT1 has been shown to play a role in the regulation of sex differentiation in the vertebrate gonad. Downregulation of DMRT1 in male mice results in trans-differentiation of Sertoli cells into granulosa (FOXL2+) cells resulting in an ovarian gonadal phenotype. STUDY DESIGN, SIZE, DURATION To determine the effect of DMRT1 repression on human fetal testes, we developed a novel system for genetic manipulation, which utilizes a Lentivral delivered miRNA during short-term in vitro culture (2 weeks). A long-term (4–6 weeks) ex vivo xenograft model was used to determine the subsequent effects of DMRT1 repression on testicular development and maintenance. We included first and second-trimester testis tissue (8–20 weeks gestation; n = 12) in the study. PARTICIPANTS/MATERIALS, SETTING, METHODS Human fetal testes were cultured in vitro and exposed to either of two DMRT1 miRNAs (miR536, miR641), or to scrambled control miRNA, for 24 h. This was followed by a further 14 days of culture (n = 3–4), or xenografting (n = 5) into immunocompromised mice for 4–6 weeks. Tissues were analyzed by histology, immunohistochemistry, immunofluorescence and quantitative RT-PCR. Endpoints included histological evaluation of seminiferous cord integrity, mRNA expression of testicular, ovarian and germ cell genes, and assessment of cell number and protein expression for proliferation, apoptosis and pluripotency factors. Statistical analysis was performed using a linear mixed effect model. MAIN RESULTS AND THE ROLE OF CHANCE DMRT1 repression (miR536/miR641) resulted in a loss of DMRT1 protein expression in a sub-population of Sertoli cells of first trimester (8–11 weeks gestation) human fetal testis; however, this did not affect the completion of seminiferous cord formation or morphological appearance. In second-trimester testis (12–20 weeks gestation), DMRT1 repression (miR536/miR641) resulted in disruption of seminiferous cords with absence of DMRT1 protein expression in Sertoli (SOX9+) cells. No differences in proliferation (Ki67+) were observed and apoptotic cells (CC3+) were rare. Expression of the Sertoli cell associated gene, SOX8, was significantly reduced (miR536, 34% reduction, P = 0.031; miR641 36% reduction, P = 0.026), whilst SOX9 expression was unaffected. Changes in expression of AMH (miR536, 100% increase, P = 0.033), CYP26B1 (miR641, 38% reduction, P = 0.05) and PTGDS (miR642, 30% reduction, P = 0.0076) were also observed. Amongst granulosa cell associated genes, there was a significant downregulation in R-spondin 1 expression (miR536, 76% reduction, P < 0.0001; miR641, 49% reduction, P = 0.046); however, there were no changes in expression of the granulosa cell marker, FOXL2. Analysis of germ cell associated genes demonstrated a significant increase in the expression of the pluripotency gene OCT4 (miR536, 233%, P < 0.001). We used the xenograft system to investigate the longer-term effects of seminiferous cord disruption via DMRT1 repression. As was evident in vitro for second-trimester samples, DMRT1 repression resulted in focal testicular dysgenesis similar to that described in adults with TDS. These dysgenetic areas were devoid of germ cells, whilst expression of FOXL2 within the dysgenetic areas, indicated trans-differentiation from a male (Sertoli cell) to female (granulosa cell) phenotype. LIMITATIONS, REASONS FOR CAUTION Human fetal testis tissue is a limited resource; however, we were able to demonstrate significant effects of DMRT1 repression on the expression of germ and somatic cell genes, in addition to the induction of focal testicular dysgenesis, using these limited samples. In vitro culture may not reflect all aspects of human fetal testis development and function; however, the concurrent use of the xenograft model which represents a more physiological system supports the validity of the in vitro findings. WIDER IMPLICATIONS OF THE FINDINGS Our findings have important implications for understanding the role of DMRT1 in human testis development and in the origin of testicular dysgenesis. In addition, we provide validation of a novel system that can be used to determine the effects of repression of genes that have been implicated in gonadal development and associated human reproductive disorders. STUDY FUNDING/COMPETING INTEREST(S) This project was funded by a Wellcome Trust Intermediate Clinical Fellowship (Grant No. 098522) awarded to RTM. LBS was supported by MRC Programme Grant MR/N002970/1. RAA was supported by MRC Programme Grant G1100357/1. RMS was supported by MRC Programme Grant G33253. This work was undertaken in the MRC Centre for Reproductive Health which is funded by the MRC Centre grant MR/N022556/1. The funding bodies had no input into the conduct of the research or the production of this manuscript. The authors have declared no conflicts of interest.
Collapse
Affiliation(s)
- Joni Macdonald
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK
| | - Karen R Kilcoyne
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK
| | - Richard M Sharpe
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK
| | - Áine Kavanagh
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK
| | - Pamela Brown
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK.,School of Environmental and Life Sciences, Faculty of Science, University of Newcastle, Callaghan, NSW, Australia
| | - Anne Jørgensen
- University Department of Growth and Reproduction, Rigshospitalet, Blegdamsvej 9, Copenhagen, Denmark
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, Scotland, UK.,Edinburgh Royal Hospital for Sick Children, 9 Sciennes Road, Edinburgh, Scotland, UK
| |
Collapse
|
41
|
Regulation of Leydig cell steroidogenesis: intriguing network of signaling pathways and mitochondrial signalosome. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.coemr.2019.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
42
|
Helsel A, Griswold MD. Retinoic acid signaling and the cycle of the seminiferous epithelium. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2019; 6:1-6. [PMID: 32832726 PMCID: PMC7442248 DOI: 10.1016/j.coemr.2019.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Affiliation(s)
- Aileen Helsel
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, WA, 99164-7520, United States
| | - Michael D Griswold
- School of Molecular Biosciences and the Center for Reproductive Biology, Washington State University, Pullman, WA, 99164-7520, United States
| |
Collapse
|
43
|
Sonic Hedgehog Signaling Is Required for Cyp26 Expression during Embryonic Development. Int J Mol Sci 2019; 20:ijms20092275. [PMID: 31072004 PMCID: PMC6540044 DOI: 10.3390/ijms20092275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 02/06/2023] Open
Abstract
Deciphering how signaling pathways interact during development is necessary for understanding the etiopathogenesis of congenital malformations and disease. In several embryonic structures, components of the Hedgehog and retinoic acid pathways, two potent players in development and disease are expressed and operate in the same or adjacent tissues and cells. Yet whether and, if so, how these pathways interact during organogenesis is, to a large extent, unclear. Using genetic and experimental approaches in the mouse, we show that during development of ontogenetically different organs, including the tail, genital tubercle, and secondary palate, Sonic hedgehog (SHH) loss-of-function causes anomalies phenocopying those induced by enhanced retinoic acid signaling and that SHH is required to prevent supraphysiological activation of retinoic signaling through maintenance and reinforcement of expression of the Cyp26 genes. Furthermore, in other tissues and organs, disruptions of the Hedgehog or the retinoic acid pathways during development generate similar phenotypes. These findings reveal that rigidly calibrated Hedgehog and retinoic acid activities are required for normal organogenesis and tissue patterning.
Collapse
|
44
|
Parekh PA, Garcia TX, Waheeb R, Jain V, Gandhi P, Meistrich ML, Shetty G, Hofmann MC. Undifferentiated spermatogonia regulate Cyp26b1 expression through NOTCH signaling and drive germ cell differentiation. FASEB J 2019; 33:8423-8435. [PMID: 30991836 DOI: 10.1096/fj.201802361r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cytochrome P450 family 26 subfamily B member 1 (CYP26B1) regulates the concentration of all-trans retinoic acid (RA) and plays a key role in germ cell differentiation by controlling local distribution of RA. The mechanisms regulating Cyp26b1 expression in postnatal Sertoli cells, the main components of the stem cell niche, are so far unknown. During gonad development, expression of Cyp26b1 is maintained by Steroidogenic Factor 1 (SF-1) and Sex-Determining Region Y Box-9 (SOX9), which ensure that RA is degraded and germ cell differentiation is blocked. Here, we show that the NOTCH target Hairy/Enhancer-of-Split Related with YRPW Motif 1 (HEY1), a transcriptional repressor, regulates germ cell differentiation via direct binding to the Cyp26b1 promoter and thus inhibits its expression in Sertoli cells. Further, using in vivo germ cell ablation, we demonstrate that undifferentiated type A spermatogonia are the cells that activate NOTCH signaling in Sertoli cells through their expression of the NOTCH ligand JAGGED-1 (JAG1) at stage VIII of the seminiferous epithelium cycle, therefore mediating germ cell differentiation by a ligand concentration-dependent process. These data therefore provide more insights into the mechanisms of germ cell differentiation after birth and potentially explain the spatiotemporal RA pulses driving the transition between undifferentiated to differentiating spermatogonia.-Parekh, P. A., Garcia, T. X., Waheeb, R., Jain, V., Gandhi, P., Meistrich, M. L., Shetty, G., Hofmann, M.-C. Undifferentiated spermatogonia regulate Cyp26b1 expression through NOTCH signaling and drive germ cell differentiation.
Collapse
Affiliation(s)
- Parag A Parekh
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Thomas X Garcia
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA.,Department of Biology and Biotechnology, University of Houston-Clear Lake, Houston, Texas, USA
| | - Reham Waheeb
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA.,Department of Theriogenology, University of Alexandria, Alexandria, Egypt
| | - Vivek Jain
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA.,Department of Biology and Biotechnology, University of Houston-Clear Lake, Houston, Texas, USA
| | - Pooja Gandhi
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Marvin L Meistrich
- Department of Experimental Radiation Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Gunapala Shetty
- Department of Experimental Radiation Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Marie-Claude Hofmann
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
45
|
Abstract
Germ cells are the stem cells of the species. Thus, it is critical that we have a good understanding of how they are specified, how the somatic cells instruct and support them, how they commit to one or other sex, and how they ultimately develop into functional gametes. Here, we focus on specifics of how sexual fate is determined during fetal life. Because the majority of relevant experimental work has been done using the mouse model, we focus on that species. We review evidence regarding the identity of instructive signals from the somatic cells, and the molecular responses that occur in germ cells in response to those extrinsic signals. In this way we aim to clarify progress to date regarding the mechanisms underlying the mitotic to meiosis switch in germ cells of the fetal ovary, and those involved in adopting and securing male fate in germ cells of the fetal testis.
Collapse
Affiliation(s)
- Cassy Spiller
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Josephine Bowles
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia; Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|