1
|
Curantz C, Doody C, Horkan HR, Krasovec G, Weavers PK, DuBuc TQ, Frank U. A positive feedback loop between germ cells and gonads induces and maintains sexual reproduction in a cnidarian. SCIENCE ADVANCES 2025; 11:eadq8220. [PMID: 39772697 PMCID: PMC11708894 DOI: 10.1126/sciadv.adq8220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025]
Abstract
The fertile gonad includes cells of two distinct developmental origins: the somatic mesoderm and the germ line. How somatic and germ cells interact to develop and maintain fertility is not well understood. Here, using grafting experiments and transgenic reporter animals, we find that a specific part of the gonad-the germinal zone-acts as a sexual organizer to induce and maintain de novo germ cells and somatic gonads in the cnidarian Hydractinia symbiolongicarpus. Germ cells express a member of the transforming growth factor-β family, Gonadless (Gls), that induces gonad morphogenesis. Loss of Gls resulted in animals lacking gonads but having nonproliferative germ cells. We propose that primary germ cells drive gonad development though Gls secretion. The germinal zone in the newly formed gonad provides positive feedback to induce secondary germ cells by activating Tfap2 in resident pluripotent stem cells. The contribution of germ cell signaling to the patterning of somatic gonadal tissue may be a general animal feature.
Collapse
Affiliation(s)
- Camille Curantz
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, University of Galway, Galway H91 W2TY, Ireland
| | - Ciara Doody
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, University of Galway, Galway H91 W2TY, Ireland
| | - Helen R. Horkan
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, University of Galway, Galway H91 W2TY, Ireland
| | - Gabriel Krasovec
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, University of Galway, Galway H91 W2TY, Ireland
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris F-75013, France
| | - Paris K. Weavers
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, University of Galway, Galway H91 W2TY, Ireland
| | - Timothy Q. DuBuc
- Department of Biology, Queens College, The City University of New York, 6530 Kissena Blvd., Flushing, NY 11367, USA
- Biology and Biochemistry PhD Programs, CUNY Graduate Center, 365 5th Ave., New York, NY 10016, USA
| | - Uri Frank
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, University of Galway, Galway H91 W2TY, Ireland
| |
Collapse
|
2
|
Hala D. The use of in silico extreme pathway (ExPa) analysis to identify conserved reproductive transcriptional-regulatory networks in humans, mice, and zebrafish. Syst Biol Reprod Med 2023; 69:271-287. [PMID: 37023256 PMCID: PMC10461611 DOI: 10.1080/19396368.2023.2188996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 04/08/2023]
Abstract
Vertebrate sex determination and differentiation are coordinated by the activations and maintenance of reproductive transcriptional-regulatory networks (TRNs). There is considerable interest in studying the conserved design principles and functions of reproductive TRNs given that their intricate regulation is susceptible to disruption by gene mutations or exposures to exogenous endocrine disrupting chemicals (or EDCs). In this manuscript, the Boolean rules describing reproductive TRNs in humans, mice, and zebrafish, were represented as a pseudo-stoichiometric matrix model. This model mathematically described the interactions of 35 transcription factors with 21 sex determination and differentiation genes across the three species. The in silico approach of Extreme Pathway (ExPa) analysis was used to predict the extent of TRN gene activations subject to the species-specific transcriptomics data, from across various developmental life-stages. A goal of this work was to identify conserved and functional reproductive TRNs across the three species. ExPa analyses predicted the sex differentiation genes, DHH, DMRT1, and AR, to be highly active in male humans, mice, and zebrafish. Whereas FOXL2 was the most active gene in female humans and mice; and CYP19A1A in female zebrafish. These results agree with the expectation that regardless of a lack of sex determination genes in zebrafish, the TRNs responsible for canalizing male vs. female sexual differentiation are conserved with mammalian taxa. ExPa analysis therefore provides a framework with which to study the TRNs that influence the development of sexual phenotypes. And the in silico predicted conservation of sex differentiation TRNs between mammals and zebrafish identifies the piscine species as an effective in vivo model to study mammalian reproductive systems under normal or perturbed pathologies.
Collapse
Affiliation(s)
- David Hala
- Department of Marine Biology, Texas A&M University at Galveston, TX, USA
| |
Collapse
|
3
|
Jung KM, Seo M, Han JY. Comparative single-cell transcriptomic analysis reveals differences in signaling pathways in gonadal primordial germ cells between chicken (Gallus gallus) and zebra finch (Taeniopygia guttata). FASEB J 2023; 37:e22706. [PMID: 36520042 DOI: 10.1096/fj.202201569r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/16/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Primordial germ cells (PGCs) have been used in avian genetic resource conservation and transgenic animal production. Despite their potential applications to numerous avian taxa facing extinction due to habitat loss and degradation, research has largely focused on poultry, such as chickens, in part owing to the difficulty in obtaining intact PGCs from other species. Recently, phenotypic differences between PGCs of chicken and zebra finch, a wild bird with vocal learning, in early embryonic development have been reported. In this study, we used advanced single-cell RNA sequencing (scRNA-seq) technology to evaluate zebra finch and chicken PGCs and surrounding cells, and to identify species-specific characteristics. We constructed single-cell transcriptome landscapes of chicken gonadal PGCs for a comparison with previously reported scRNA-seq data for zebra finch. We identified interspecific differences in several signaling pathways in gonadal PGCs and somatic cells. In particular, NODAL and insulin signaling pathway activity levels were higher in zebra finch than in chickens, whereas activity levels of the downstream FGF signaling pathway, involved in the proliferation of chicken PGCs, were higher in chickens. This study is the first cross-species single-cell transcriptomic analysis targeting birds, revealing differences in germ cell development between phylogenetically distant Galliformes and Passeriformes. Our results provide a basis for understanding the reproductive physiology of avian germ cells and for utilizing PGCs in the restoration of endangered birds and the production of transgenic birds.
Collapse
Affiliation(s)
- Kyung Min Jung
- Biomodulation Major, Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Minseok Seo
- Department of Computer and Information Science, Korea University, Sejong, South Korea
| | - Jae Yong Han
- Biomodulation Major, Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
4
|
Tveiten H, Karlsen K, Thesslund T, Johansson GS, Thiyagarajan DB, Andersen Ø. Impact of germ cell ablation on the activation of the brain-pituitary-gonadal axis in precocious Atlantic salmon (Salmo salar L.) males. Mol Reprod Dev 2022; 89:471-484. [PMID: 35830347 PMCID: PMC9796531 DOI: 10.1002/mrd.23635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/27/2022] [Accepted: 07/04/2022] [Indexed: 01/01/2023]
Abstract
The germ cells are essential for sexual reproduction by giving rise to the gametes, but the importance of germ cells for gonadal somatic functions varies among vertebrates. The RNA-binding dead end (Dnd) protein is necessary for the specification and migration of primordial germ cells to the future reproductive organs. Here, we ablated the gametes in Atlantic salmon males and females by microinjecting dnd antisense gapmer oligonucleotides at the zygotic stage. Precocious maturation was induced in above 50% of both germ cell-depleted and intact fertile males, but not in females, by exposure to an off-season photoperiod regime. Sterile and fertile males showed similar body growth, but maturing fish tended to be heavier than their immature counterparts. Pituitary fshβ messenger RNA levels strongly increased in maturing sterile and fertile males concomitant with the upregulated expression of Sertoli and Leydig cell markers. Plasma concentrations of 11-ketotestosterone and testosterone in maturing sterile males were significantly higher than the basal levels in immature fish, but lower than those in maturing fertile males. The study demonstrates that germ cells are not a prerequisite for the activation of the brain-pituitary-gonad axis and sex steroidogenesis in Atlantic salmon males, but may be important for the maintenance of gonadal somatic functions.
Collapse
Affiliation(s)
- Helge Tveiten
- Norwegian College of Fishery ScienceThe Arctic University of NorwayTromsøNorway
| | - Kristian Karlsen
- Norwegian College of Fishery ScienceThe Arctic University of NorwayTromsøNorway,Present address:
Lerøy Aurora AS, Stortorget 1N‐9267 TromsøNorway
| | | | | | | | - Øivind Andersen
- NofimaTromsøNorway,Department of Animal and Aquacultural SciencesNorwegian University of Life Sciences (NMBU)ÅsNorway
| |
Collapse
|
5
|
McGlacken-Byrne SM, Del Valle I, Le Quesne Stabej P, Bellutti L, Garcia-Alonso L, Ocaka LA, Ishida M, Suntharalingham JP, Gagunashvili A, Ogunbiyi OK, Mistry T, Buonocore F, Crespo B, Moreno N, Niola P, Brooks T, Brain CE, Dattani MT, Kelberman D, Vento-Tormo R, Lagos CF, Livera G, Conway GS, Achermann JC. Pathogenic variants in the human m6A reader YTHDC2 are associated with primary ovarian insufficiency. JCI Insight 2022; 7:154671. [PMID: 35138268 PMCID: PMC8983136 DOI: 10.1172/jci.insight.154671] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/26/2022] [Indexed: 11/17/2022] Open
Abstract
Primary ovarian insufficiency (POI) affects 1% of women and carries significant medical and psychosocial sequelae. Approximately 10% of POI has a defined genetic cause, with most implicated genes relating to biological processes involved in early fetal ovary development and function. Recently, Ythdc2, an RNA helicase and N6-methyladenosine (m6a) reader, has emerged as a novel regulator of meiosis in mice. Here, we describe homozygous pathogenic variants in YTHDC2 in three women with early-onset POI from two families: c. 2567C>G, p.P856R in the helicase-associated (HA2) domain; and c.1129G>T, p.E377*. We demonstrate that YTHDC2 is expressed in the developing human fetal ovary and is upregulated in meiotic germ cells, together with related meiosis-associated factors. The p.P856R variant results in a less flexible protein that likely disrupts downstream conformational kinetics of the HA2 domain, whereas the p.E377* variant truncates the helicase core. Taken together, our results reveal that YTHDC2 is a key new regulator of meiosis in humans and pathogenic variants within this gene are associated with POI.
Collapse
Affiliation(s)
- Sinead M McGlacken-Byrne
- Genetics and Genomics Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Ignacio Del Valle
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Polona Le Quesne Stabej
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Laura Bellutti
- Laboratory of Development of the Gonads, UMR E008, Université de Paris, Université Paris Saclay, CEA, Fontenay aux Roses, France
| | - Luz Garcia-Alonso
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Louise A Ocaka
- GOSgene, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Miho Ishida
- Genetics and Genomics Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Jenifer P Suntharalingham
- Genetics and Genomics Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Andrey Gagunashvili
- GOSgene, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Olumide K Ogunbiyi
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Talisa Mistry
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Federica Buonocore
- Genetics and Genomics Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | | | - Berta Crespo
- Developmental Biology and Cancer, UCL Great Ormond Street Institute of Child health, London, United Kingdom
| | - Nadjeda Moreno
- Developmental Biology and Cancer, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Paola Niola
- UCL Genomics, Zayed Centre for Research, London, United Kingdom
| | - Tony Brooks
- UCL Genomics, Zayed Centre for Research, London, United Kingdom
| | - Caroline E Brain
- Department of Paediatric Endocrinology, Great Ormond Street Hospital, London, United Kingdom
| | - Mehul T Dattani
- Genetics and Genomics Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Daniel Kelberman
- GOSgene, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Carlos F Lagos
- Chemical Biology & Drug Discovery Lab, Escuela de Química y Farmacia, Universidad San Sebastián, Santiago, Chile
| | - Gabriel Livera
- Laboratory of Development of the Gonads, UMR E008, Université de Paris, Université Paris Saclay, CEA, Fontenay aux Roses, France
| | - Gerard S Conway
- Institute for Women's Health, University College London, London, United Kingdom
| | - John C Achermann
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
DuBuc TQ, Schnitzler CE, Chrysostomou E, McMahon ET, Febrimarsa, Gahan JM, Buggie T, Gornik SG, Hanley S, Barreira SN, Gonzalez P, Baxevanis AD, Frank U. Transcription factor AP2 controls cnidarian germ cell induction. Science 2020; 367:757-762. [PMID: 32054756 PMCID: PMC7025884 DOI: 10.1126/science.aay6782] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/06/2019] [Indexed: 12/18/2022]
Abstract
Clonal animals do not sequester a germ line during embryogenesis. Instead, they have adult stem cells that contribute to somatic tissues or gametes. How germ fate is induced in these animals, and whether this process is related to bilaterian embryonic germline induction, is unknown. We show that transcription factor AP2 (Tfap2), a regulator of mammalian germ lines, acts to commit adult stem cells, known as i-cells, to the germ cell fate in the clonal cnidarian Hydractinia symbiolongicarpus Tfap2 mutants lacked germ cells and gonads. Transplanted wild-type cells rescued gonad development but not germ cell induction in Tfap2 mutants. Forced expression of Tfap2 in i-cells converted them to germ cells. Therefore, Tfap2 is a regulator of germ cell commitment across germ line-sequestering and germ line-nonsequestering animals.
Collapse
Affiliation(s)
- Timothy Q DuBuc
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Christine E Schnitzler
- Whitney Laboratory for Marine Bioscience, University of Florida, St. Augustine, FL 32080, USA
- Department of Biology, University of Florida, Gainesville, FL 32611, USA
| | - Eleni Chrysostomou
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Emma T McMahon
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Febrimarsa
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - James M Gahan
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Tara Buggie
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Sebastian G Gornik
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Shirley Hanley
- National Centre for Biomedical Engineering Science, National University of Ireland Galway, Galway, Ireland
| | - Sofia N Barreira
- Computational and Statistical Genomics Branch, Division of Intramural Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paul Gonzalez
- Computational and Statistical Genomics Branch, Division of Intramural Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andreas D Baxevanis
- Computational and Statistical Genomics Branch, Division of Intramural Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Uri Frank
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
9
|
N-Cadherin Is Critical for the Survival of Germ Cells, the Formation of Steroidogenic Cells, and the Architecture of Developing Mouse Gonads. Cells 2019; 8:cells8121610. [PMID: 31835801 PMCID: PMC6952792 DOI: 10.3390/cells8121610] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 01/10/2023] Open
Abstract
Normal gonad development assures the fertility of the individual. The properly functioning gonads must contain a sufficient number of the viable germ cells, possess a correct architecture and tissue structure, and assure the proper hormonal regulation. This is achieved by the interplay between the germ cells and different types of somatic cells. N-cadherin coded by the Cdh2 gene plays a critical role in this interplay. To gain an insight into the role of N-cadherin in the development of mouse gonads, we used the Cre-loxP system to knock out N-cadherin separately in two cell lines: the SF1+ somatic cells and the OCT4+ germ cells. We observed that N-cadherin plays a key role in the survival of both female and male germ cells. However, the N-cadherin is not necessary for the differentiation of the Sertoli cells or the initiation of the formation of testis cords or ovigerous cords. In the later stages of gonad development, N-cadherin is important for the maintenance of testis cord structure and is required for the formation of steroidogenic cells. In the ovaries, N-cadherin is necessary for the formation of the ovarian follicles. These results indicate that N-cadherin plays a major role in gonad differentiation, structuralization, and function.
Collapse
|
10
|
da Silva TE, Gomes NL, Lerário AM, Keegan CE, Nishi MY, Carvalho FM, Vilain E, Barseghyan H, Martinez-Aguayo A, Forclaz MV, Papazian R, Pedroso de Paula LC, Costa EC, Carvalho LR, Jorge AAL, Elias FM, Mitchell R, Costa EMF, Mendonca BB, Domenice S. Genetic Evidence of the Association of DEAH-Box Helicase 37 Defects With 46,XY Gonadal Dysgenesis Spectrum. J Clin Endocrinol Metab 2019; 104:5923-5934. [PMID: 31287541 DOI: 10.1210/jc.2019-00984] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/02/2019] [Indexed: 01/07/2023]
Abstract
CONTEXT 46,XY Gonadal dysgenesis (GD) is a heterogeneous group of disorders with a wide phenotypic spectrum, including embryonic testicular regression syndrome (ETRS). OBJECTIVE To report a gene for 46,XY GD etiology, especially for ETRS. DESIGN Screening of familial cases of 46,XY GD using whole-exome sequencing and sporadic cases by target gene-panel sequencing. SETTING Tertiary Referral Center for differences/disorders of sex development (DSD). PATIENTS AND INTERVENTIONS We selected 87 patients with 46,XY DSD (17 familial cases from 8 unrelated families and 70 sporadic cases); 55 patients had GD (among them, 10 patients from 5 families and 8 sporadic cases had ETRS), and 32 patients had 46,XY DSD of unknown etiology. RESULTS We identified four heterozygous missense rare variants, classified as pathogenic or likely pathogenic in the Asp-Glu-Ala-His-box (DHX) helicase 37 (DHX37) gene in five families (n = 11 patients) and in six sporadic cases. Two variants were recurrent: p.Arg308Gln (in two families and in three sporadic cases) and p.Arg674Trp (in two families and in two sporadic cases). The variants were specifically associated with ETRS (7/14 index cases; 50%). The frequency of rare, predicted-to-be-deleterious DHX37 variants in this cohort (14%) is significantly higher than that observed in the Genome Aggregation Database (0.4%; P < 0.001). Immunohistochemistry analysis in human testis showed that DHX37 is mainly expressed in germ cells at different stages of testis maturation, in Leydig cells, and rarely in Sertoli cells. CONCLUSION This strong genetic evidence identifies DHX37 as a player in the complex cascade of male gonadal differentiation and maintenance.
Collapse
Affiliation(s)
- Thatiana Evilen da Silva
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Nathalia Lisboa Gomes
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Antonio Marcondes Lerário
- Laboratório de Sequenciamento em Larga Escala, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Catherine Elizabeth Keegan
- Department of Pediatric Genetics, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Mirian Yumi Nishi
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Eric Vilain
- Center for Genetic Medicine Research, The Children's Research Institute, Children's National Medical Center, Children's National Health System, Washington, DC
| | - Hayk Barseghyan
- Center for Genetic Medicine Research, The Children's Research Institute, Children's National Medical Center, Children's National Health System, Washington, DC
| | - Alejandro Martinez-Aguayo
- Division de Pediatria, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | | | - Regina Papazian
- Servicio de Pediatría, Hospital Nacional Prof. Dr. A. Posadas, Buenos Aires, Argentina
| | - Leila Cristina Pedroso de Paula
- Programa de Atendimento às Desordens do Desenvolvimento Sexual, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Eduardo Corrêa Costa
- Programa de Atendimento às Desordens do Desenvolvimento Sexual, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luciani Renata Carvalho
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Alexander Augusto Lima Jorge
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Felipe Martins Elias
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Rod Mitchell
- Medical Research Council Centre for Reproductive Health, Queens Medical Research Institute, Edinburgh, United Kingdom
| | - Elaine Maria Frade Costa
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Berenice Bilharinho Mendonca
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Sequenciamento em Larga Escala, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Sorahia Domenice
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular (LIM/42) da Disciplina de Endocrinologia e Metabologia do Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
11
|
Herpin A, Schmidt C, Kneitz S, Gobé C, Regensburger M, Le Cam A, Montfort J, Adolfi MC, Lillesaar C, Wilhelm D, Kraeussling M, Mourot B, Porcon B, Pannetier M, Pailhoux E, Ettwiller L, Dolle D, Guiguen Y, Schartl M. A novel evolutionary conserved mechanism of RNA stability regulates synexpression of primordial germ cell-specific genes prior to the sex-determination stage in medaka. PLoS Biol 2019; 17:e3000185. [PMID: 30947255 PMCID: PMC6448818 DOI: 10.1371/journal.pbio.3000185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/04/2019] [Indexed: 12/20/2022] Open
Abstract
Dmrt1 is a highly conserved transcription factor, which is critically involved in regulation of gonad development of vertebrates. In medaka, a duplicate of dmrt1-acting as master sex-determining gene-has a tightly timely and spatially controlled gonadal expression pattern. In addition to transcriptional regulation, a sequence motif in the 3' UTR (D3U-box) mediates transcript stability of dmrt1 mRNAs from medaka and other vertebrates. We show here that in medaka, two RNA-binding proteins with antagonizing properties target this D3U-box, promoting either RNA stabilization in germ cells or degradation in the soma. The D3U-box is also conserved in other germ-cell transcripts, making them responsive to the same RNA binding proteins. The evolutionary conservation of the D3U-box motif within dmrt1 genes of metazoans-together with preserved expression patterns of the targeting RNA binding proteins in subsets of germ cells-suggest that this new mechanism for controlling RNA stability is not restricted to fishes but might also apply to other vertebrates.
Collapse
Affiliation(s)
- Amaury Herpin
- INRA, UR1037 Fish Physiology and Genomics, Rennes, France
- University of Wuerzburg, Physiological Chemistry, Biocenter, Wuerzburg, Germany
- * E-mail:
| | - Cornelia Schmidt
- University of Wuerzburg, Physiological Chemistry, Biocenter, Wuerzburg, Germany
| | - Susanne Kneitz
- University of Wuerzburg, Physiological Chemistry, Biocenter, Wuerzburg, Germany
| | - Clara Gobé
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France
| | | | - Aurélie Le Cam
- INRA, UR1037 Fish Physiology and Genomics, Rennes, France
| | | | - Mateus C. Adolfi
- University of Wuerzburg, Physiological Chemistry, Biocenter, Wuerzburg, Germany
| | - Christina Lillesaar
- University of Wuerzburg, Physiological Chemistry, Biocenter, Wuerzburg, Germany
| | - Dagmar Wilhelm
- University of Melbourne, Department of Anatomy & Neuroscience, Parkville, Victoria, Australia
| | - Michael Kraeussling
- University of Wuerzburg, Physiological Chemistry, Biocenter, Wuerzburg, Germany
| | | | | | - Maëlle Pannetier
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France
| | - Eric Pailhoux
- UMR BDR, INRA, ENVA, Université Paris Saclay, Jouy en Josas, France
| | - Laurence Ettwiller
- University of Heidelberg, Centre for Organismal Studies (COS), Department of Developmental Biology, Heidelberg, Germany
| | - Dirk Dolle
- University of Heidelberg, Centre for Organismal Studies (COS), Department of Developmental Biology, Heidelberg, Germany
| | - Yann Guiguen
- INRA, UR1037 Fish Physiology and Genomics, Rennes, France
| | - Manfred Schartl
- University of Wuerzburg, Physiological Chemistry, Biocenter, Wuerzburg, Germany
- Comprehensive Cancer Center Mainfranken, University Hospital, Wuerzburg, Germany
- Hagler Institute for Advanced Study and Department of Biology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
12
|
Trukhina AV, Lukina NA, Smirnov AF. Hormonal Sex Inversion and Some Aspects of Its Genetic Determination in Chicken. RUSS J GENET+ 2018. [DOI: 10.1134/s1022795418090144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
13
|
Pradhan A, Olsson PE. Germ cell depletion in zebrafish leads to incomplete masculinization of the brain. Gen Comp Endocrinol 2018; 265:15-21. [PMID: 29408375 DOI: 10.1016/j.ygcen.2018.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 11/15/2022]
Abstract
Zebrafish sex differentiation is under the control of multiple genes, but also relies on germ cell number for gonadal development. Morpholino and chemical mediated germ cell depletion leads to sterile male development in zebrafish. In this study we produced sterile males, using a dead end gene morpholino, to determine gonadal-brain interactions. Germ cell depletion following dnd inhibition downregulated the germ cell markers, vasa and ziwi, and later the larvae developed as sterile males. Despite lacking proper testis, the gonadal 11-ketotestosterone (11-KT) and estradiol (E2) levels of sterile males were similar to wild type males. Qualitative analysis of sexual behavior of sterile males demonstrated that they behaved like wild type males. Furthermore, we observed that brain 11-KT and E2 levels in sterile males remained the same as in the wild type males. In female brain, 11-KT was lower in comparison to wild type males and sterile males, while E2 was higher when compared to wild type males. qRT-PCR analysis revealed that the liver transcript profile of sterile adult males was similar to wild type males while the brain transcript profile was similar to wild type females. The results demonstrate that proper testis development may not be a prerequisite for male brain development in zebrafish but that it may be needed to fully masculinize the brain.
Collapse
Affiliation(s)
- Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, SE-701 82 Örebro, Sweden
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, SE-701 82 Örebro, Sweden.
| |
Collapse
|
14
|
Hollenbach J, Jung K, Noelke J, Gasse H, Pfarrer C, Koy M, Brehm R. Loss of connexin43 in murine Sertoli cells and its effect on blood-testis barrier formation and dynamics. PLoS One 2018; 13:e0198100. [PMID: 29856785 PMCID: PMC5983412 DOI: 10.1371/journal.pone.0198100] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
Abstract
Connexin43 (Cx43) is the predominant testicular gap junction protein and in cases of impaired spermatogenesis, Cx43 expression has been shown to be altered in several mammals. Amongst other functions, Cx43 is supposed to regulate junction formation of the blood-testis barrier (BTB). The aim of the present study was to investigate the expression pattern of different tight junction (TJ) proteins of the murine BTB using SC-specific Cx43 knockout mice (SCCx43KO). Adult homozygous male SCCx43KO mice (SCCx43KO-/-) predominantly show an arrest of spermatogenesis and SC-only tubules that might have been caused by an altered BTB assembly, composition or regulation. TJ molecules claudin-3, -5 and -11 were examined in adult wild type (WT) and SCCx43KO-/- mice using immunohistochemistry (IHC) and quantitative real-time PCR (qRT-PCR). In this context, investigation of single tubules with residual spermatogenesis in SCCx43KO-/- mice was particularly interesting to identify a potential Cx43-independent influence of germ cells (GC) on BTB composition and dynamics. In tubules without residual spermatogenesis, a diffuse cytoplasmic distribution pattern for claudin-11 protein could be demonstrated in mutant mice. Nevertheless, claudin-11 seems to form functional TJ. Claudin-3 and -5 could not be detected immunohistochemically in the seminiferous epithelium of those tubules. Correspondingly, claudin-3 and -5 mRNA expression was decreased, providing evidence of generally impaired BTB dynamics in adult KO mice. Observations of tubules with residual spermatogenesis suggested a Cx43-independent regulation of TJ proteins by GC populations. To determine initial BTB formation in peripubertal SCCx43KO-/- mice, immunohistochemical staining and qRT-PCR of claudin-11 were carried out in adolescent SCCx43KO-/- and WT mice. Additionally, BTB integrity was functionally analysed using a hypertonic glucose fixative. These analyses revealed that SCCx43KO-/- mice formed an intact BTB during puberty in the same time period as WT mice, which however seemed to be accelerated.
Collapse
Affiliation(s)
- Julia Hollenbach
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Klaus Jung
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Joanna Noelke
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hagen Gasse
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Christiane Pfarrer
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Mirja Koy
- Institute for Immunology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Ralph Brehm
- Institute for Anatomy, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
15
|
Qiu GH, Huang C, Zheng X, Yang X. The protective function of noncoding DNA in genome defense of eukaryotic male germ cells. Epigenomics 2018; 10:499-517. [PMID: 29616594 DOI: 10.2217/epi-2017-0103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Peripheral and abundant noncoding DNA has been hypothesized to protect the genome and the central protein-coding sequences against DNA damage in somatic genome. In the cytosol, invading exogenous nucleic acids may first be deactivated by small RNAs encoded by noncoding DNA via mechanisms similar to the prokaryotic CRISPR-Cas system. In the nucleus, the radicals generated by radiation in the cytosol, radiation energy and invading exogenous nucleic acids are absorbed, blocked and/or reduced by peripheral heterochromatin, and damaged DNA in heterochromatin is removed and excluded from the nucleus to the cytoplasm through nuclear pore complexes. To further strengthen the hypothesis, this review summarizes the experimental evidence supporting the protective function of noncoding DNA in the genome of male germ cells. Based on these data, this review provides evidence supporting the protective role of noncoding DNA in the genome defense of sperm genome through similar mechanisms to those of the somatic genome.
Collapse
Affiliation(s)
- Guo-Hua Qiu
- Fujian Provincial Key Laboratory for the Prevention & Control of Animal Infectious Diseases & Biotechnology; Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Fujian Province University; College of Life Sciences, Longyan University, Longyan 364012, Fujian, PR China
| | - Cuiqin Huang
- Fujian Provincial Key Laboratory for the Prevention & Control of Animal Infectious Diseases & Biotechnology; Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Fujian Province University; College of Life Sciences, Longyan University, Longyan 364012, Fujian, PR China
| | - Xintian Zheng
- Fujian Provincial Key Laboratory for the Prevention & Control of Animal Infectious Diseases & Biotechnology; Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Fujian Province University; College of Life Sciences, Longyan University, Longyan 364012, Fujian, PR China
| | - Xiaoyan Yang
- Fujian Provincial Key Laboratory for the Prevention & Control of Animal Infectious Diseases & Biotechnology; Key Laboratory of Preventive Veterinary Medicine and Biotechnology, Fujian Province University; College of Life Sciences, Longyan University, Longyan 364012, Fujian, PR China
| |
Collapse
|
16
|
Rios-Rojas C, Spiller C, Bowles J, Koopman P. Germ cells influence cord formation and leydig cell gene expression during mouse testis development. Dev Dyn 2016; 245:433-44. [DOI: 10.1002/dvdy.24371] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 10/15/2015] [Accepted: 11/18/2015] [Indexed: 11/07/2022] Open
Affiliation(s)
- Clarissa Rios-Rojas
- Institute for Molecular Bioscience; The University of Queensland; Brisbane Australia
| | - Cassy Spiller
- Institute for Molecular Bioscience; The University of Queensland; Brisbane Australia
| | - Josephine Bowles
- Institute for Molecular Bioscience; The University of Queensland; Brisbane Australia
| | - Peter Koopman
- Institute for Molecular Bioscience; The University of Queensland; Brisbane Australia
| |
Collapse
|
17
|
|