1
|
He J, Wang Z, Yang L, Jiang Y, Yan G, Pan Y, Gao F, Yuan J, Gao Y. Unveiling the role of FOXL2 in female differentiation and disease: a comprehensive review†. Biol Reprod 2025; 112:600-613. [PMID: 39976382 DOI: 10.1093/biolre/ioaf013] [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: 08/26/2024] [Revised: 12/30/2024] [Accepted: 02/20/2025] [Indexed: 02/21/2025] Open
Abstract
Ovarian differentiation relies on the accurate and orderly expression of numerous related genes. Forkhead box protein L2 (FOXL2) is one of the earliest ovarian differentiation markers and transcription factors. In sex determination, FOXL2 maintains the differentiation of the female pathway by inhibiting male differentiation genes, including SOX9 and SF1. In addition, FOXL2 promotes the synthesis of follicle-stimulating hormone and anti-Müllerian hormone to support follicle development. Mutations in FOXL2 are associated with numerous female reproductive diseases. A comprehensive and in-depth study of FOXL2 provides novel strategies for the diagnosis and treatment of such diseases. This review discusses the mechanism of FOXL2 in female sex differentiation and maintenance, hormone synthesis, and disease occurrence and reveals the role of FOXL2 as a central factor in female sex development and fertility maintenance. This review will serve as a reference for identifying novel targets of other regulatory factors interacting with FOXL2 in female sex determination and follicle development and for the diagnosis and treatment of female reproductive diseases.
Collapse
Affiliation(s)
- Jia He
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| | - Zican Wang
- College of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Lici Yang
- College of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Yongjian Jiang
- College of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Ge Yan
- College of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Yongwei Pan
- College of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Fei Gao
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jinxiang Yuan
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong, China
| | - Yang Gao
- College of Basic Medicine, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
2
|
Predescu DN, Mokhlesi B, Predescu SA. X-inactive-specific transcript: a long noncoding RNA with a complex role in sex differences in human disease. Biol Sex Differ 2024; 15:101. [PMID: 39639337 PMCID: PMC11619133 DOI: 10.1186/s13293-024-00681-5] [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: 09/04/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
In humans, the X and Y chromosomes determine the biological sex, XX specifying for females and XY for males. The long noncoding RNA X-inactive specific transcript (lncRNA XIST) plays a crucial role in the process of X chromosome inactivation (XCI) in cells of the female, a process that ensures the balanced expression of X-linked genes between sexes. Initially, it was believed that XIST can be expressed only from the inactive X chromosome (Xi) and is considered a typically female-specific transcript. However, accumulating evidence suggests that XIST can be detected in male cells as well, and it participates in the development of cancers and other human diseases by regulating gene expression at epigenetic, chromatin remodeling, transcriptional, and translational levels. XIST is abnormally expressed in many sexually dimorphic diseases, including autoimmune and neurological diseases, pulmonary arterial hypertension (PAH), and some types of cancers. However, the underlying mechanisms are not fully understood. Escape from XCI and skewed XCI also contributes to sex-biased diseases and their severity. Interestingly, in humans, similar to experimental animal models of human disease, the males with the XIST gene activated display the sex-biased disease condition at a rate close to females, and significantly greater than males who had not been genetically modified. For instance, the men with supernumerary X chromosomes, such as men with Klinefelter syndrome (47, XXY), are predisposed toward autoimmunity similar to females (46, XX), and have increased risk for strongly female biased diseases, compared to 46, XY males. Interestingly, chromosome X content has been linked to a longer life span, and the presence of two chromosome X contributes to increased longevity regardless of the hormonal status. In this review, we summarize recent knowledge about XIST structure/function correlation and involvement in human disease with focus on XIST abnormal expression in males. Many human diseases show differences between males and females in penetrance, presentation, progression, and survival. In humans, the X and Y sex chromosomes determine the biological sex, XX specifying for females and XY for males. This numeric imbalance, two X chromosomes in females and only one in males, known as sex chromosome dosage inequality, is corrected in the first days of embryonic development by inactivating one of the X chromosomes in females. While this "dosage compensation" should in theory solve the difference in the number of genes between sexes, the expressed doses of X genes are incompletely compensated by X chromosome inactivation in females. In this review we try to highlight how abnormal expression and function of XIST, a gene on the X chromosome responsible for this inactivation process, may explain the sex differences in human health and disease. A better understanding of the molecular mechanisms of XIST participation in the male-female differences in disease is highly relevant since it would allow for improving the personalization of diagnosis and sex-specific treatment of patients.
Collapse
Affiliation(s)
- Dan N Predescu
- Department of Internal Medicine, Pulmonary, Critical Care, and Sleep Medicine, Rush University Medical Center, Chicago, IL, 60612, USA.
| | - Babak Mokhlesi
- Department of Internal Medicine, Pulmonary, Critical Care, and Sleep Medicine, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Sanda A Predescu
- Department of Internal Medicine, Pulmonary, Critical Care, and Sleep Medicine, Rush University Medical Center, Chicago, IL, 60612, USA
| |
Collapse
|
3
|
King AC, Zenker AK. Sex blind: bridging the gap between drug exposure and sex-related gene expression in Danio rerio using next-generation sequencing (NGS) data and a literature review to find the missing links in pharmaceutical and environmental toxicology studies. FRONTIERS IN TOXICOLOGY 2023; 5:1187302. [PMID: 37398910 PMCID: PMC10312089 DOI: 10.3389/ftox.2023.1187302] [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: 03/15/2023] [Accepted: 06/01/2023] [Indexed: 07/04/2023] Open
Abstract
The sex of both humans and Danio rerio has previously been shown to affect the way individuals respond to drug exposure. Genes which allow identification of sex in juvenile zebrafish show potential to reveal these confounding variables between sex in toxicological and preclinical trials but the link between these is so far missing. These sex-specific, early expressed genes where expression is not altered by drug exposure must be carefully selected for this purpose. We aimed to discover genes which can be used in pharmaceutical trials and environmental toxicology studies to uncover sex-related variations in gene expression with drug application using the model organism Danio rerio. Previously published early sex determining genes from King et al. were evaluated as well as additional genes selected from our zebrafish Next-generation sequencing (NGS) data which are known from previously published works not to be susceptible to changes in expression with drug exposure. NGS revealed a further ten female-specific genes (vtg1, cyp17a1, cyp19a1a, igf3, ftz-f1, gdf9, foxl2a, Nr0b1, ipo4, lhcgr) and five male related candidate genes (FKBP5, apobb1, hbaa1, dmrt1, spata6) which are also expressed in juvenile zebrafish, 28 days post fertilisation (dpf). Following this, a literature review was performed to classify which of these early-expressed sex specific genes are already known to be affected by drug exposure in order to determine candidate genes to be used in pharmaceutical trials or environmental toxicology testing studies. Discovery of these early sex-determining genes in Danio rerio will allow identification of sex-related responses to drug testing to improve sex-specific healthcare and the medical treatment of human patients.
Collapse
Affiliation(s)
| | - Armin K. Zenker
- University of Applied Sciences and Arts North-Western Switzerland (FHNW), Muttenz, Switzerland
| |
Collapse
|
4
|
Chen HA, Grimshaw AA, Taylor-Giorlando M, Vijayakumar P, Li D, Margetts M, Pelosi E, Vash-Margita A. Ovarian absence: a systematic literature review and case series report. J Ovarian Res 2023; 16:13. [PMID: 36642704 PMCID: PMC9841619 DOI: 10.1186/s13048-022-01090-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 12/24/2022] [Indexed: 01/17/2023] Open
Abstract
Ovarian absence is an uncommon condition that most frequently presents unilaterally. Several etiologies for the condition have been proposed, including torsion, vascular accident, and embryological defect. A systematic review was conducted to describe the clinical presentation of ovarian absence, as well as its associations with other congenital anomalies, through a systematic search of Cochrane Library, ClinicalTrials.gov, Google Scholar, Ovid Embase, Ovid Medline, PubMed, Scopus, and Web of Science. Exclusion criteria included cases with suspicion for Differences of Sex Development, lack of surgically-confirmed ovarian absence, and karyotypes other than 46XX. Our search yielded 12,120 citations, of which 79 studies were included. 10 additional studies were found by citation chasing resulting in a total 113 cases including two unpublished cases presented in this review. Abdominal/pelvic pain (30%) and infertility/subfertility (19%) were the most frequent presentations. Ovarian abnormalities were not noted in 28% of cases with pre-operative ovarian imaging results. Approximately 17% of cases had concomitant uterine abnormalities, while 22% had renal abnormalities. Renal abnormalities were more likely in patients with uterine abnormalities (p < 0.005). Torsion or vascular etiology was the most frequently suspected etiology of ovarian absence (52%), followed by indeterminate (27%) and embryologic etiology (21%). Most cases of ovarian absence are likely attributable to torsion or vascular accidents, despite many references to the condition as "agenesis" in the literature. Imaging may fail to correctly diagnose ovarian absence, and diagnostic laparoscopy may be preferable in many cases as genitourinary anatomy and fertility considerations can be assessed during the procedure. Fertility is likely minimally or not affected in women with unilateral ovarian absence.
Collapse
Affiliation(s)
| | - Alyssa A Grimshaw
- Yale University, Harvey Cushing/John Hay Whitney Medical Library, New Haven, CT, USA
| | | | - Pavithra Vijayakumar
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Dan Li
- Yale University School of Medicine, New Haven, CT, USA
| | - Miranda Margetts
- Center for American Indian and Rural Health Equity, Montana State University, Bozeman, MT, USA
| | - Emanuele Pelosi
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Alla Vash-Margita
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale New Haven Hospital, New Haven, CT, USA.
- Yale Department of Obstetrics, Gynecology & Reproductive Medicine, Farnam Memorial Building, 310 Cedar Street, Fl 3, Rm 329, New Haven, CT, 06510, USA.
| |
Collapse
|
5
|
Lucas-Herald AK, Mitchell RT. Testicular Sertoli Cell Hormones in Differences in Sex Development. Front Endocrinol (Lausanne) 2022; 13:919670. [PMID: 35909548 PMCID: PMC9329667 DOI: 10.3389/fendo.2022.919670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
The Sertoli cells of the testes play an essential role during gonadal development, in addition to supporting subsequent germ cell survival and spermatogenesis. Anti-Müllerian hormone (AMH) is a member of the TGF-β superfamily, which is secreted by immature Sertoli cells from the 8th week of fetal gestation. lnhibin B is a glycoprotein, which is produced by the Sertoli cells from early in fetal development. In people with a Difference or Disorder of Sex Development (DSD), these hormones may be useful to determine the presence of testicular tissue and potential for spermatogenesis. However, fetal Sertoli cell development and function is often dysregulated in DSD conditions and altered production of Sertoli cell hormones may be detected throughout the life course in these individuals. As such this review will consider the role of AMH and inhibin B in individuals with DSD.
Collapse
Affiliation(s)
- Angela K. Lucas-Herald
- Developmental Endocrinology Research Group, University of Glasgow, Glasgow, United Kingdom
| | - Rod T. Mitchell
- MRC Centre for Reproductive Health, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Department of Paediatric Endocrinology, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
| |
Collapse
|
6
|
Fang F, Iaquinta PJ, Xia N, Liu L, Diao L, Reijo Pera RA. Transcriptional control of human gametogenesis. Hum Reprod Update 2022; 28:313-345. [PMID: 35297982 PMCID: PMC9071081 DOI: 10.1093/humupd/dmac002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 11/22/2021] [Indexed: 11/14/2022] Open
Abstract
The pathways of gametogenesis encompass elaborate cellular specialization accompanied by precise partitioning of the genome content in order to produce fully matured spermatozoa and oocytes. Transcription factors are an important class of molecules that function in gametogenesis to regulate intrinsic gene expression programs, play essential roles in specifying (or determining) germ cell fate and assist in guiding full maturation of germ cells and maintenance of their populations. Moreover, in order to reinforce or redirect cell fate in vitro, it is transcription factors that are most frequently induced, over-expressed or activated. Many reviews have focused on the molecular development and genetics of gametogenesis, in vivo and in vitro, in model organisms and in humans, including several recent comprehensive reviews: here, we focus specifically on the role of transcription factors. Recent advances in stem cell biology and multi-omic studies have enabled deeper investigation into the unique transcriptional mechanisms of human reproductive development. Moreover, as methods continually improve, in vitro differentiation of germ cells can provide the platform for robust gain- and loss-of-function genetic analyses. These analyses are delineating unique and shared human germ cell transcriptional network components that, together with somatic lineage specifiers and pluripotency transcription factors, function in transitions from pluripotent stem cells to gametes. This grand theme review offers additional insight into human infertility and reproductive disorders that are linked predominantly to defects in the transcription factor networks and thus may potentially contribute to the development of novel treatments for infertility.
Collapse
Affiliation(s)
- Fang Fang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Phillip J Iaquinta
- Division of Research, Economic Development, and Graduate Education, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Ninuo Xia
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Lei Liu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Lei Diao
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Renee A Reijo Pera
- Division of Research, Economic Development, and Graduate Education, California Polytechnic State University, San Luis Obispo, CA, USA
- McLaughlin Research Institute, Great Falls, MT, USA
| |
Collapse
|
7
|
King AC, Gut M, Zenker AK. Shedding new light on early sex determination in zebrafish. Arch Toxicol 2020; 94:4143-4158. [PMID: 32975586 PMCID: PMC7655572 DOI: 10.1007/s00204-020-02915-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 09/17/2020] [Indexed: 01/10/2023]
Abstract
In contrast to established zebrafish gene annotations, the question of sex determination has still not been conclusively clarified for developing zebrafish, Danio rerio, larvae, 28 dpf or earlier. Recent studies indicate polygenic sex determination (PSD), with the genes being distributed throughout the genome. Early genetic markers of sex in zebrafish help unravel co-founding sex-related differences to apply to human health and environmental toxicity studies. A qPCR-based method was developed for six genes: cytochrome P450, family 17, subfamily A, polypeptide 1 (cyp17a1); cytochrome P450, family 19, subfamily A, polypeptide 1a (cyp19a1a); cytochrome P450, family 19, subfamily A, polypeptides 1b (cyp19a1b); vitellogenin 1 (vtg1); nuclear receptor subfamily 0, group B, member 1 (nr0b1), sry (sex-determining region Y)-box 9b (sox9b) and actin, beta 1 (actb1), the reference gene. Sry-box 9a (Sox9a), insulin-like growth factor 3 (igf3) and double sex and mab-3 related transcription factor 1 (dmrt1), which are also known to be associated with sex determination, were used in gene expression tests. Additionally, Next-Generation-Sequencing (NGS) sequenced the genome of two adult female and male and two juveniles. PCR analysis of adult zebrafish revealed sex-specific expression of cyp17a1, cyp19a1a, vtg1, igf3 and dmrt1, the first four strongly expressed in female zebrafish and the last one highly expressed in male conspecifics. From NGS, nine female and four male-fated genes were selected as novel for assessing zebrafish sex, 28 dpf. Differences in transcriptomes allowed allocation of sex-specific genes also expressed in juvenile zebrafish.
Collapse
Affiliation(s)
- Alex C King
- FHNW, University of Applied Sciences and Arts North-Western Switzerland, School of Life Sciences, Institute for Ecopreneurship, Hofackerstrasse 30, 4132, Muttenz, Switzerland
| | - Michelle Gut
- FHNW, University of Applied Sciences and Arts North-Western Switzerland, School of Life Sciences, Institute for Ecopreneurship, Hofackerstrasse 30, 4132, Muttenz, Switzerland
| | - Armin K Zenker
- FHNW, University of Applied Sciences and Arts North-Western Switzerland, School of Life Sciences, Institute for Ecopreneurship, Hofackerstrasse 30, 4132, Muttenz, Switzerland.
| |
Collapse
|
8
|
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: 147] [Impact Index Per Article: 29.4] [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
|
9
|
Lobo IKC, Nascimento ÁRD, Yamagishi MEB, Guiguen Y, Silva GFD, Severac D, Amaral ADC, Reis VR, Almeida FLD. Transcriptome of tambaqui Colossoma macropomum during gonad differentiation: Different molecular signals leading to sex identity. Genomics 2020; 112:2478-2488. [PMID: 32027957 DOI: 10.1016/j.ygeno.2020.01.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 01/11/2020] [Accepted: 01/31/2020] [Indexed: 12/13/2022]
Abstract
Tambaqui (Colossoma macropomum) is the major native species in Brazilian aquaculture, and we have shown that females exhibit a higher growth compared to males, opening up the possibility for the production of all-female population. To date, there is no information on the sex determination and differentiation molecular mechanisms of tambaqui. In the present study, transcriptome sequencing of juvenile trunks was performed to understand the molecular network involved in the gonadal sex differentiation. The results showed that before differentiation, components of the Wnt/β-catenin pathway, fox and fst genes imprint female sex development, whereas antagonistic pathways (gsk3b, wt1 and fgfr2), sox9 and genes for androgen synthesis indicate male differentiation. Hence, in undifferentiated tambaqui, the Wnt/β-catenin exerts a role on sex differentiation, either upregulated in female-like individuals, or antagonized in male-like individuals.
Collapse
Affiliation(s)
| | | | | | - Yann Guiguen
- INRA, UR1037 LPGP, Campus de Beaulieu, Rennes, France.
| | | | - Dany Severac
- MGX, Univ Montpellier, CNRS, INSERM, Montpellier, France.
| | - Aldessandro da Costa Amaral
- Programa de Pós-graduação em Ciências Pesqueiras nos Trópicos, Universidade Federal do Amazonas, Manaus, Brazil
| | - Vanessa Ribeiro Reis
- Programa de Pós-graduação em Biotecnologia, Universidade Federal do Amazonas, Manaus, Brazil
| | | |
Collapse
|
10
|
Only two sex forms but multiple gender variants: How to explain? Commun Integr Biol 2018; 11:e1427399. [PMID: 29497472 PMCID: PMC5824932 DOI: 10.1080/19420889.2018.1427399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/04/2018] [Indexed: 11/29/2022] Open
Abstract
Are sex and gender interchangeable terms? In classical biology, both are sometimes but not always used on an equal basis for some groups of animals. However, for our own species the Homo sapiens, they are not. A major question is why are there only two types of gametes (sperm- and egg cells), two types of sex steroids, (androgens and estrogens in vertebrates, and two types of ecdysteroids in insects), while the reproduction-related behaviour of the gamete producers displays a much greater variability than just two prominent forms, namely heterosexual males and heterosexual females? It indicates that in addition to a few sex-determining genes ( = the first pillar), other factors play a role. A second possible pillar is the still poorly understood cognitive memory system in which electrical phenomena and its association with the plasma membrane membrane-cytoskeletal complex of cells play a major role (learning, imitation and imprinting). This paper advances a third pillar, that hitherto has been almost completely ignored, namely the cellular Ca2+-homeostasis system, more specifically its sex-specific differences. Differential male-female genetics- and hormone-based Ca2+-homeostasis with effects on gender-related processes has been named Calcigender before. It will be argued that it follows from the principles of Ca2+- physiology and homeostasis that all individuals of a sexually reproducing animal population have a personalized gender behaviour. Thus, subdividing gender-behaviours in hetero-, homo-, bi-, trans- etc. which all result from a differential use of the very same basic physiological principles, is too primitive a system that may yield false sociological interpretations.
Collapse
|
11
|
Morphogenesis-related gene-expression profile in porcine oocytes before and after in vitro maturation. ZYGOTE 2017; 25:331-340. [PMID: 28669375 DOI: 10.1017/s096719941700020x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mammalian oocyte maturation is achieved when oocytes reach metaphase II (MII) stage, and accumulate mRNA and proteins in the cytoplasm following fertilization. It has been shown that oocytes investigated before and after in vitro maturation (IVM) differ significantly in transcriptomic and proteomic profiles. Additionally, folliculogenesis and oogenesis is accompanied by morphogenetic changes, which significantly influence further zygote formation and embryo growth. This study aimed to determine new transcriptomic markers of porcine oocyte morphogenesis that are associated with cell maturation competence. An Affymetrix microarray assay was performed on an RNA template isolated from porcine oocytes before (n = 150) and after (n = 150) IVM. The brilliant cresyl blue (BCB) staining test was used for identification of cells with the highest developmental capacity. DAVID (Database for Annotation, Visualization, and Integrated Discovery) software was used for the extraction of the genes belonging to a cell morphogenesis Gene Ontology group. The control group consisted of freshly isolated oocytes. In total, 12,000 different transcripts were analysed, from which 379 genes were downregulated and 40 were upregulated in oocytes following IVM. We found five genes, SOX9, MAP1B, DAB2, FN1, and CXCL12, that were significantly upregulated in oocytes after IVM (in vitro group) compared with oocytes analysed before IVM (in vivo group). In conclusion, we found new transcriptomic markers of oocyte morphogenesis, which may be also recognized as significant mediators of cellular maturation capacity in pigs. Genes SOX9, MAP1B, DAB2, FN1, and CXCL12 may be involved in the regulation of the MII stage oocyte formation and several other processes that are crucial for porcine reproductive competence.
Collapse
|
12
|
Jeong YH, Lu H, Park CH, Li M, Luo H, Kim JJ, Liu S, Ko KH, Huang S, Hwang IS, Kang MN, Gong D, Park KB, Choi EJ, Park JH, Jeong YW, Moon C, Hyun SH, Kim NH, Jeung EB, Yang H, Hwang WS, Gao F. Stochastic anomaly of methylome but persistent SRY hypermethylation in disorder of sex development in canine somatic cell nuclear transfer. Sci Rep 2016; 6:31088. [PMID: 27501986 PMCID: PMC4977463 DOI: 10.1038/srep31088] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 07/13/2016] [Indexed: 12/30/2022] Open
Abstract
Somatic cell nuclear transfer (SCNT) provides an excellent model for studying epigenomic reprogramming during mammalian development. We mapped the whole genome and whole methylome for potential anomalies of mutations or epimutations in SCNT-generated dogs with XY chromosomal sex but complete gonadal dysgenesis, which is classified as 78, XY disorder of sex development (DSD). Whole genome sequencing revealed no potential genomic variations that could explain the pathogenesis of DSD. However, extensive but stochastic anomalies of genome-wide DNA methylation were discovered in these SCNT DSD dogs. Persistent abnormal hypermethylation of the SRY gene was observed together with its down-regulated mRNA and protein expression. Failure of SRY expression due to hypermethylation was further correlated with silencing of a serial of testis determining genes, including SOX9, SF1, SOX8, AMH and DMRT1 in an early embryonic development stage at E34 in the XY(DSD) gonad, and high activation of the female specific genes, including FOXL2, RSPO1, CYP19A1, WNT4, ERα and ERβ, after one postnatal year in the ovotestis. Our results demonstrate that incomplete demethylation on the SRY gene is the driving cause of XY(DSD) in these XY DSD dogs, indicating a central role of epigenetic regulation in sex determination.
Collapse
Affiliation(s)
| | - Hanlin Lu
- BGI-Shenzhen, Shenzhen, China.,Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Chi-Hun Park
- Sooam Biotech Research Foundation, Seoul 152-904, Korea.,Animal Bioscience and Biotechnology Laboratory, United States Department of Agriculture, Beltsville, MD, 20705, USA.,Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | | | | | - Joung Joo Kim
- Sooam Biotech Research Foundation, Seoul 152-904, Korea
| | | | - Kyeong Hee Ko
- Sooam Biotech Research Foundation, Seoul 152-904, Korea
| | | | - In Sung Hwang
- Sooam Biotech Research Foundation, Seoul 152-904, Korea
| | - Mi Na Kang
- Sooam Biotech Research Foundation, Seoul 152-904, Korea
| | - Desheng Gong
- BGI-Shenzhen, Shenzhen, China.,Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Kang Bae Park
- Sooam Biotech Research Foundation, Seoul 152-904, Korea
| | - Eun Ji Choi
- Sooam Biotech Research Foundation, Seoul 152-904, Korea
| | | | | | - Changjong Moon
- College of Veterinary Medicine, Chonnam National University, Gwangju 500-757, Korea
| | - Sang-Hwan Hyun
- Sooam Biotech Research Foundation, Seoul 152-904, Korea.,College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, Korea
| | - Nam Hyung Kim
- College of Animal Sciences, Chungbuk National University, Cheongju, Chungbuk 361-763, Korea
| | - Eui-Bae Jeung
- College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 361-763, Korea
| | | | - Woo Suk Hwang
- Sooam Biotech Research Foundation, Seoul 152-904, Korea
| | - Fei Gao
- BGI-Shenzhen, Shenzhen, China.,Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| |
Collapse
|
13
|
Taketo T. The role of sex chromosomes in mammalian germ cell differentiation: can the germ cells carrying X and Y chromosomes differentiate into fertile oocytes? Asian J Androl 2016; 17:360-6. [PMID: 25578929 PMCID: PMC4430933 DOI: 10.4103/1008-682x.143306] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The sexual differentiation of germ cells into spermatozoa or oocytes is strictly regulated by their gonadal environment, testis or ovary, which is determined by the presence or absence of the Y chromosome, respectively. Hence, in normal mammalian development, male germ cells differentiate in the presence of X and Y chromosomes, and female germ cells do so in the presence of two X chromosomes. However, gonadal sex reversal occurs in humans as well as in other mammalian species, and the resultant XX males and XY females can lead healthy lives, except for a complete or partial loss of fertility. Germ cells carrying an abnormal set of sex chromosomes are efficiently eliminated by multilayered surveillance mechanisms in the testis, and also, though more variably, in the ovary. Studying the molecular basis for sex-specific responses to a set of sex chromosomes during gametogenesis will promote our understanding of meiotic processes contributing to the evolution of sex determining mechanisms. This review discusses the fate of germ cells carrying various sex chromosomal compositions in mouse models, the limitation of which may be overcome by recent successes in the differentiation of functional germ cells from embryonic stem cells under experimental conditions.
Collapse
Affiliation(s)
- Teruko Taketo
- Department of Surgery, Research Institute of MUHC; Department of Biology; Department of Obstetrics and Gynecology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
14
|
Zhou L, Charkraborty T, Zhou Q, Mohapatra S, Nagahama Y, Zhang Y. Rspo1-activated signalling molecules are sufficient to induce ovarian differentiation in XY medaka (Oryzias latipes). Sci Rep 2016; 6:19543. [PMID: 26782368 PMCID: PMC4726049 DOI: 10.1038/srep19543] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 09/23/2015] [Indexed: 11/15/2022] Open
Abstract
In contrast to our understanding of testicular differentiation, ovarian differentiation is less well understood in vertebrates. In mammals, R-spondin1 (Rspo1), an activator of Wnt/β-catenin signaling pathway, is located upstream of the female sex determination pathway. However, the functions of Rspo1 in ovarian differentiation remain unclear in non-mammalian species. In order to elucidate the detailed functions of Rspo/Wnt signaling pathway in fish sex determination/differentiation, the ectopic expression of the Rspo1 gene was performed in XY medaka (Oryzias latipes). The results obtained demonstrated that the gain of Rspo1 function induced femininity in XY fish. The overexpression of Rspo1 enhanced Wnt4b and β-catenin transcription, and completely suppressed the expression of male-biased genes (Dmy, Gsdf, Sox9a2 and Dmrt1) as well as testicular differentiation. Gonadal reprograming of Rspo1-over-expressed-XY (Rspo1-OV-XY) fish, induced the production of female-biased genes (Cyp19a1a and Foxl2), estradiol-17β production and further female type secondary sexuality. Moreover, Rspo1-OV-XY females were fertile and produced successive generations. Promoter analyses showed that Rspo1 transcription was directly regulated by DM domain genes (Dmy, the sex-determining gene, and Dmrt1) and remained unresponsive to Foxl2. Taken together, our results strongly suggest that Rspo1 is sufficient to activate ovarian development and plays a decisive role in the ovarian differentiation in medaka.
Collapse
Affiliation(s)
- Linyan Zhou
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing 400715, P.R. China.,SORST, Japan Science Technology Corporation, Kawaguchi, Saitama 332-0012, Japan
| | - Tapas Charkraborty
- SORST, Japan Science Technology Corporation, Kawaguchi, Saitama 332-0012, Japan.,South Ehime Fisheries Research Center, Ehime University, Ainan, Ehime, 798-4206, Japan
| | - Qian Zhou
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 305-8577, Ibaraki, Japan
| | - Sipra Mohapatra
- South Ehime Fisheries Research Center, Ehime University, Ainan, Ehime, 798-4206, Japan
| | - Yoshitaka Nagahama
- SORST, Japan Science Technology Corporation, Kawaguchi, Saitama 332-0012, Japan.,South Ehime Fisheries Research Center, Ehime University, Ainan, Ehime, 798-4206, Japan.,Institution for Collaborative Relations, Ehime University, 790-8577, Matsuyama, Japan
| | - Yueguang Zhang
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, School of Life Science, Southwest University, Chongqing 400715, P.R. China
| |
Collapse
|
15
|
Pelosi E, Forabosco A, Schlessinger D. Genetics of the ovarian reserve. Front Genet 2015; 6:308. [PMID: 26528328 PMCID: PMC4606124 DOI: 10.3389/fgene.2015.00308] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/24/2015] [Indexed: 11/13/2022] Open
Abstract
Primordial follicles or non-growing follicles (NGFs) are the functional unit of reproduction, each comprising a single germ cell surrounded by supporting somatic cells. NGFs constitute the ovarian reserve (OR), prerequisite for germ cell ovulation and the continuation of the species. The dynamics of the reserve is determined by the number of NGFs formed and their complex subsequent fates. During the reproductive lifespan, the OR progressively diminishes due to follicle atresia as well as recruitment, maturation, and ovulation. The depletion of the OR is the major determining driver of menopause, which ensues when the number of primordial follicles falls below a threshold of ∼1,000. Therefore, genes and processes involved in follicle dynamics are particularly important to understand the process of menopause, both in the typical reproductive lifespan and in conditions like primary ovarian insufficiency, defined as menopause before age 40. Genes and their variants that affect the timing of menopause thereby provide candidates for diagnosis of and intervention in problems of reproductive lifespan. We review the current knowledge of processes and genes involved in the development of the OR and in the dynamics of ovarian follicles.
Collapse
Affiliation(s)
- Emanuele Pelosi
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | | | - David Schlessinger
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
16
|
Suzuki H, Kanai-Azuma M, Kanai Y. From Sex Determination to Initial Folliculogenesis in Mammalian Ovaries: Morphogenetic Waves along the Anteroposterior and Dorsoventral Axes. Sex Dev 2015; 9:190-204. [DOI: 10.1159/000440689] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2015] [Indexed: 11/19/2022] Open
|
17
|
Marongiu M, Marcia L, Pelosi E, Lovicu M, Deiana M, Zhang Y, Puddu A, Loi A, Uda M, Forabosco A, Schlessinger D, Crisponi L. FOXL2 modulates cartilage, skeletal development and IGF1-dependent growth in mice. BMC DEVELOPMENTAL BIOLOGY 2015; 15:27. [PMID: 26134413 PMCID: PMC4489133 DOI: 10.1186/s12861-015-0072-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 05/05/2015] [Indexed: 11/10/2022]
Abstract
BACKGROUND Haploinsufficiency of the FOXL2 transcription factor in humans causes Blepharophimosis/Ptosis/Epicanthus Inversus syndrome (BPES), characterized by eyelid anomalies and premature ovarian failure. Mice lacking Foxl2 recapitulate human eyelid/forehead defects and undergo female gonadal dysgenesis. We report here that mice lacking Foxl2 also show defects in postnatal growth and embryonic bone and cartilage formation. METHODS Foxl2 (-/-) male mice at different stages of development have been characterized and compared to wild type. Body length and weight were measured and growth curves were created. Skeletons were stained with alcian blue and/or alizarin red. Bone and cartilage formation was analyzed by Von Kossa staining and immunofluorescence using anti-FOXL2 and anti-SOX9 antibodies followed by confocal microscopy. Genes differentially expressed in skull vaults were evaluated by microarray analysis. Analysis of the GH/IGF1 pathway was done evaluating the expression of several hypothalamic-pituitary-bone axis markers by RT-qPCR. RESULTS Compared to wild-type, Foxl2 null mice are smaller and show skeletal abnormalities and defects in cartilage and bone mineralization, with down-regulation of the GH/IGF1 axis. Consistent with these effects, we find FOXL2 expressed in embryos at 9.5 dpc in neural tube epithelium, in head mesenchyme near the neural tube, and within the first branchial arch; then, starting at 12.5 dpc, expressed in cartilaginous tissue; and at PO and P7, in hypothalamus. CONCLUSIONS Our results support FOXL2 as a master transcription factor in a spectrum of developmental processes, including growth, cartilage and bone formation. Its action overlaps that of SOX9, though they are antagonistic in female vs male gonadal sex determination but conjoint in cartilage and skeletal development.
Collapse
Affiliation(s)
- Mara Marongiu
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cittadella Universitaria di Monserrato, SS 554 km 4500, Monserrato, 09042, Italy.
| | - Loredana Marcia
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cittadella Universitaria di Monserrato, SS 554 km 4500, Monserrato, 09042, Italy. .,Università degli Studi di Sassari, Sassari, Italy.
| | | | - Mario Lovicu
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cittadella Universitaria di Monserrato, SS 554 km 4500, Monserrato, 09042, Italy.
| | - Manila Deiana
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cittadella Universitaria di Monserrato, SS 554 km 4500, Monserrato, 09042, Italy.
| | - Yonqing Zhang
- Laboratory of Genetics, NIA-IRP, NIH, Baltimore, MD, USA.
| | - Alessandro Puddu
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cittadella Universitaria di Monserrato, SS 554 km 4500, Monserrato, 09042, Italy. .,Università degli Studi di Cagliari, Cagliari, Italy.
| | - Angela Loi
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cittadella Universitaria di Monserrato, SS 554 km 4500, Monserrato, 09042, Italy.
| | - Manuela Uda
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cittadella Universitaria di Monserrato, SS 554 km 4500, Monserrato, 09042, Italy.
| | - Antonino Forabosco
- Cante di Montevecchio Association, Genomic Research Center, Fano, Italy.
| | | | - Laura Crisponi
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Cittadella Universitaria di Monserrato, SS 554 km 4500, Monserrato, 09042, Italy.
| |
Collapse
|
18
|
Liu H, Mu X, Gui L, Su M, Li H, Zhang G, Liu Z, Zhang J. Characterization and gonadal expression of FOXL2 relative to Cyp19a genes in spotted scat Scatophagus argus. Gene 2014; 561:6-14. [PMID: 25550048 DOI: 10.1016/j.gene.2014.12.060] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 12/16/2014] [Accepted: 12/24/2014] [Indexed: 01/12/2023]
Abstract
In the present study, we cloned the full-length cDNAs of FOXL2, Cyp19a1a and Cyp19a1b and analyzed their expression patterns during gonadal development in spotted scat, Scatophagus argus. All three genes were expressed in ovaries and testes but showed sexual dimorphism. At early stages of gonadal development, the expression of FOXL2 in ovaries was higher than testes. FOXL2 expression deceased gradually as gonadal development continued, and reached the lowest level at the mature stage. Cyp19a1a and Cyp19a1b were expressed coordinately with FOXL2, except at the early vitellogenic stage in the ovary. The expression of FOXL2, Cyp19a1a and Cyp19a1b was mainly localized in granulosa cells of ovaries. In S. argus testes, strong expression of FOXL2 gene was observed in the interstitial cells including tubules and Leydig cells, while Cyp19a1a and Cyp19a1b were mainly expressed in Sertoli cells throughout gametogenesis. These results show that FOXL2 plays an essential role in sexual development, and imply that it may regulate Cyp19a1a and Cyp19a1b expression in S. argus.
Collapse
Affiliation(s)
- Huifen Liu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Xingjiang Mu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Lang Gui
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Maoliang Su
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Hong Li
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Guang Zhang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Zhenhao Liu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Junbin Zhang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
19
|
Simpson JL. Genetics of female infertility due to anomalies of the ovary and mullerian ducts. Methods Mol Biol 2014; 1154:39-73. [PMID: 24782005 DOI: 10.1007/978-1-4939-0659-8_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Genetic factors are pivotal in reproductive development and subsequent reproductive processes. If disturbed, infertility can occur. In the female, genetic factors affecting the ovary and the uterus are not uncommon causes of infertility. Terminal deletions on the X long arm and X short arm and X chromosomal mosaicism have long been accepted as causes of premature ovarian failure (POF). Responsible genes on the X have not yet elucidated. Attractive candidate genes for POF also exist on autosomes, and in over a dozen genes molecular perturbations are documented in non-syndromic POF. The most common single-gene cause of POF is premutation carriers for FMR1 (fragile X syndrome). As other candidate genes and additional ethnic groups are interrogated, the proportion of POF cases due to single-gene mutation will increase. Among uterine anomalies, incomplete mullerian fusion is most common. Increased recurrence risks for first-degree relatives confirm a role for genetic factors; interrogation of candidate genes is under way.
Collapse
Affiliation(s)
- Joe Leigh Simpson
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, AHC2 693, Miami, FL, 33199, USA,
| |
Collapse
|
20
|
Bashamboo A, McElreavey K. Gene Mutations Associated with Anomalies of Human Gonad Formation. Sex Dev 2013; 7:126-46. [DOI: 10.1159/000342188] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
21
|
Ahmed SF, Bashamboo A, Lucas-Herald A, McElreavey K. Understanding the genetic aetiology in patients with XY DSD. Br Med Bull 2013; 106:67-89. [PMID: 23529942 DOI: 10.1093/bmb/ldt008] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Disorders of sex development (DSD) consist of a wide range of disorders and are commoner in those with an XY karyotype. In over half of these cases who have a 46,XY karyotype and who are raised as boys, the underlying aetiology remains unclear. AREAS OF AGREEMENT Identification of the underlying genetic abnormality may predict long-term outcome. However, genetic abnormalities that are associated with XY DSD manifest themselves with a wide range of phenotype. To understand the aetiology as well as the phenotypic variation, there is a need to harness the advanced genetic technology that is now available. AREAS OF CONTROVERSY The point at which genetic analysis should be undertaken in the course of investigations is unclear. In addition, there is little agreement on the most effective approach for genetic analysis that will be of clinical benefit to the patient. AREAS TIMELY FOR DEVELOPING RESEARCH There is a need to understand and improve the clinical utility of genetic analysis in the clinical setting of the patient with a suspected DSD. This will be even more important when parallel gene sequencing identifies variations in multiple genes.
Collapse
Affiliation(s)
- S F Ahmed
- School of Medicine, University of Glasgow, Royal Hospital for Sick Children, Yorkhill, Glasgow, UK.
| | | | | | | |
Collapse
|
22
|
Zhou L, Charkraborty T, Yu X, Wu L, Liu G, Mohapatra S, Wang D, Nagahama Y. R-spondins are involved in the ovarian differentiation in a teleost, medaka (Oryzias latipes). BMC DEVELOPMENTAL BIOLOGY 2012; 12:36. [PMID: 23217106 PMCID: PMC3542121 DOI: 10.1186/1471-213x-12-36] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 11/13/2012] [Indexed: 02/04/2023]
Abstract
Background In mammals, R-spondin (Rspo), an activator of the Wnt/β-catenin signaling pathway, has been shown to be involved in ovarian differentiation. However, the role of the Rspo/Wnt/β-catenin signaling pathway in fish gonads is still unknown. Results In the present study, full-length cDNAs of Rspo1, 2 and 3 were cloned from the gonads of medaka (Oryzias latipes). The deduced amino acid sequences of mRspo1-3 were shown to have a similar structural organization. Phylogenetic analysis showed that Rspo1, 2 and 3 were specifically clustered into three distinct clads. Tissue distribution revealed that three Rspo genes were abundantly expressed in the brain and ovary. Real-time PCR analysis around hatching (S33-5dah) demonstrated that three Rspo genes were specifically enhanced in female gonads from S38. In situ hybridization (ISH) analysis demonstrated that three Rspo genes were expressed in the germ cell in ovary, but not in testis. Fluorescence multi-color ISH showed that Rspo1 was expressed in both somatic cells and germ cells at 10dah. Exposure to ethinylestradiol (EE2) in XY individuals for one week dramatically enhanced the expression of three Rspo genes both at 0dah and in adulthood. Conclusions These results suggest that the Rspo-activating signaling pathway is involved in the ovarian differentiation and maintenance in medaka.
Collapse
Affiliation(s)
- Linyan Zhou
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), School of Life Science, Southwest University, Chongqing, PR China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Rosario R, Araki H, Print CG, Shelling AN. The transcriptional targets of mutant FOXL2 in granulosa cell tumours. PLoS One 2012; 7:e46270. [PMID: 23029457 PMCID: PMC3460904 DOI: 10.1371/journal.pone.0046270] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 08/28/2012] [Indexed: 11/18/2022] Open
Abstract
Background Despite their distinct biology, granulosa cell tumours (GCTs) are treated the same as other ovarian tumours. Intriguingly, a recurring somatic mutation in the transcription factor Forkhead Box L2 (FOXL2) 402C>G has been found in nearly all GCTs examined. This investigation aims to identify the pathogenicity of mutant FOXL2 by studying its altered transcriptional targets. Methods The expression of mutant FOXL2 was reduced in the GCT cell line KGN, and wildtype and mutant FOXL2 were overexpressed in the GCT cell line COV434. Total RNA was hybridised to Affymetrix U133 Plus 2 microarrays. Comparisons were made between the transcriptomes of control cells and cells altered by FOXL2 knockdown and overexpression, to detect potential transcriptional targets of mutant FOXL2. Results The overexpression of wildtype and mutant FOXL2 in COV434, and the silencing of mutant FOXL2 expression in KGN, has shown that mutant FOXL2 is able to differentially regulate the expression of many genes, including two well known FOXL2 targets, StAR and CYP19A. We have shown that many of the genes regulated by mutant FOXL2 are clustered into functional annotations of cell death, proliferation, and tumourigenesis. Furthermore, TGF-β signalling was found to be enriched when using the gene annotation tools GATHER and GeneSetDB. This enrichment was still significant after performing a robust permutation analysis. Conclusion Given that many of the transcriptional targets of mutant FOXL2 are known TGF-β signalling genes, we suggest that deregulation of this key antiproliferative pathway is one way mutant FOXL2 contributes to the pathogenesis of adult-type GCTs. We believe this pathway should be a target for future therapeutic interventions, if outcomes for women with GCTs are to improve.
Collapse
Affiliation(s)
- Roseanne Rosario
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand.
| | | | | | | |
Collapse
|
24
|
Kobayashi Y, Nagahama Y, Nakamura M. Diversity and plasticity of sex determination and differentiation in fishes. Sex Dev 2012; 7:115-25. [PMID: 22948719 DOI: 10.1159/000342009] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Among vertebrates, fishes show an exceptional range of reproductive strategies regarding the expression of their sexuality. Fish sexualities were categorized into gonochorism, synchronous/sequential hermaphrodite, or unisexual reproduction. In gonochoristic fishes, sex is determined genetically or by environmental factors. After sex determination, the gonads are differentiated into ovary or testis, with the sex remaining fixed for the entire life cycle. In contrast, some sequential hermaphrodite fishes can change their sex from male to female (protandrous), female to male (protogynous), or serially (bi-directional sex change) in their life cycle. In many cases, sex change is cued by social factors such as the disappearance of a male or female from a group. This unique diversity in fishes provides an ideal animal model to investigate sex determination and differentiation in vertebrates. This review first discusses genetic-orientated sex determination mechanisms. Then, we address the gonadal sex differentiation process in a gonochoristic fish, using an example of the Nile tilapia. Finally, we discuss various types of sex change that occur in hermaphrodite fishes.
Collapse
Affiliation(s)
- Y Kobayashi
- Tropical Biosphere Research Center, Sesoko Station, University of the Ryukyus, Motobu, Japan.
| | | | | |
Collapse
|
25
|
Abstract
During embryonic development, ovarian somatic cells embark on a course that is separate from male somatic cells and from indifferent precursor cells. While the former aspect of ovarian development is well known, the latter has not received much attention until recently. This review attempts to integrate the most recent work regarding the differentiation of ovarian somatic cells. The discussion of the parallel development of the testis is limited to the key differences only. Similarly, germ cell development will be introduced only inasmuch as it becomes necessary to draw attention to a particular aspect of the somatic component differentiation. Finally, while postnatal ovarian development and folliculogenesis undoubtedly provide the ultimate morphological and functional fitness tests for the ovarian somatic cells, postnatal phenotypes will be only referred to when they have already been connected to genes that are expressed during embryogenesis.
Collapse
Affiliation(s)
- S G Tevosian
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Fla. 32601, USA.
| |
Collapse
|
26
|
Hayrabedyan S, Todorova K, Pashova S, Mollova M, Fernández N. Sertoli Cell Quiescence - New Insights. Am J Reprod Immunol 2012; 68:451-5. [DOI: 10.1111/j.1600-0897.2012.01137.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 03/13/2012] [Indexed: 12/26/2022] Open
Affiliation(s)
- Soren Hayrabedyan
- Institute of Biology and Immunology of Reproduction; BAS; Sofia; Bulgaria
| | | | - Shina Pashova
- Institute of Biology and Immunology of Reproduction; BAS; Sofia; Bulgaria
| | - Margarita Mollova
- Institute of Biology and Immunology of Reproduction; BAS; Sofia; Bulgaria
| | - Nelson Fernández
- School of Biological Sciences; University of Essex; Colchester; UK
| |
Collapse
|
27
|
|
28
|
Boyer A, Yeh JR, Zhang X, Paquet M, Gaudin A, Nagano MC, Boerboom D. CTNNB1 signaling in sertoli cells downregulates spermatogonial stem cell activity via WNT4. PLoS One 2012; 7:e29764. [PMID: 22253774 PMCID: PMC3257228 DOI: 10.1371/journal.pone.0029764] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 12/03/2011] [Indexed: 01/15/2023] Open
Abstract
Constitutive activation of the WNT signaling effector CTNNB1 (β-catenin) in the Sertoli cells of the Ctnnb1tm1Mmt/+;Amhr2tm3(cre)Bhr/+ mouse model results in progressive germ cell loss and sterility. In this study, we sought to determine if this phenotype could be due to a loss of spermatogonial stem cell (SSC) activity. Reciprocal SSC transplants between Ctnnb1tm1Mmt/+;Amhr2tm3(cre)Bhr/+ and wild-type mice showed that SSC activity is lost in Ctnnb1tm1Mmt/+;Amhr2tm3(cre)Bhr/+ testes over time, whereas the mutant testes could not support colonization by wild-type SSCs. Microarray analyses performed on cultured Sertoli cells showed that CTNNB1 induces the expression of genes associated with the female sex determination pathway, which was also found to occur in Ctnnb1tm1Mmt/+;Amhr2tm3(cre)Bhr/+ testes. One CTNNB1 target gene encoded the secreted signaling molecule WNT4. We therefore tested the effects of WNT4 on SSC-enriched germ cell cultures, and found that WNT4 induced cell death and reduced SSC activity without affecting cell cycle. Conversely, conditional inactivation of Wnt4 in the Ctnnb1tm1Mmt/+;Amhr2tm3(cre)Bhr/+ model rescued spermatogenesis and male fertility, indicating that WNT4 is the major effector downstream of CTNNB1 responsible for germ cell loss. Furthermore, WNT4 was found to signal via the CTNNB1 pathway in Sertoli cells, suggesting a self-reinforcing positive feedback loop. Collectively, these data indicate for the first time that ectopic activation of a signaling cascade in the stem cell niche depletes SSC activity through a paracrine factor. These findings may provide insight into the pathogenesis of male infertility, as well as embryonic gonadal development.
Collapse
Affiliation(s)
- Alexandre Boyer
- Animal Reproduction Research Centre (CRRA), Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Québec, Canada
| | - Jonathan R. Yeh
- Department of Obstetrics and Gynecology, McGill University, Montréal, Québec, Canada
| | - Xiangfan Zhang
- Department of Obstetrics and Gynecology, McGill University, Montréal, Québec, Canada
| | - Marilène Paquet
- Comparative Medicine and Animal Resources Centre, McGill University, Montréal, Québec, Canada
| | - Aurore Gaudin
- Animal Reproduction Research Centre (CRRA), Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Québec, Canada
| | - Makoto C. Nagano
- Department of Obstetrics and Gynecology, McGill University, Montréal, Québec, Canada
| | - Derek Boerboom
- Animal Reproduction Research Centre (CRRA), Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, Québec, Canada
- * E-mail:
| |
Collapse
|
29
|
Xi W, Wan HT, Zhao YG, Wong MH, Giesy JP, Wong CKC. Effects of perinatal exposure to bisphenol A and di(2-ethylhexyl)-phthalate on gonadal development of male mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2011; 19:2515-27. [PMID: 22828881 DOI: 10.1007/s11356-012-0827-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 02/13/2012] [Indexed: 04/15/2023]
Abstract
PURPOSE In this study, we investigated the effects of maternal transfer of bisphenol A (BPA) and di(2-ethylhexyl) phthalate (DEHP) during gestational and weaning periods on gonadal development of male offspring. METHODS Pregnant CD-1 mice were administered by gavages in corn oil with 0.1, 1, or 10 mg/kg/day of BPA and DEHP from gestational days (GD1-21) to the weaning period (postnatal days (PND) 1-21). RESULTS Our data indicated that the exposure significantly reduced the male-to-female sex ratio and the sizes of the gonads of male pups as recorded at PND15. The testes of the perinatally exposed male pups were developed less and the expression levels of testicular anti-mullerian hormone, androgen receptor, cyclin A, and StAR were significantly lesser than the control male pups. The less developed testes were accompanied with significant reductions in the expression levels of Gnrh and Fsh at the hypothalamic-pituitary levels. The negative effects were found to be persistent in the sexually mature pups at PND42. CONCLUSION Our data reveal that the maternal transfer of BPA and DEHP may impose negative influence on the development and functions of the reproductive system of male pups.
Collapse
Affiliation(s)
- Wei Xi
- Croucher Institute of Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong, People's Republic of China
| | | | | | | | | | | |
Collapse
|
30
|
Cools M, Wolffenbuttel KP, Drop SLS, Oosterhuis JW, Looijenga LHJ. Gonadal development and tumor formation at the crossroads of male and female sex determination. Sex Dev 2011; 5:167-80. [PMID: 21791949 DOI: 10.1159/000329477] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2011] [Indexed: 01/19/2023] Open
Abstract
Malignant germ cell tumor (GCT) formation is a well-known complication in the management of patients with a disorder of sex development (DSD). DSDs are defined as congenital conditions in which development of chromosomal, gonadal, or anatomical sex is atypical. DSD patients in whom the karyotype - at least at the gonadal level - contains (a part of) the Y chromosome are at increased risk for neoplastic transformation of germ cells, leading to the development of the so-called 'type II germ cell tumors'. However, tumor risk in the various forms of DSD varies considerably between the different diagnostic groups. This contribution integrates our actual knowledge on the pathophysiology of tumor development in DSDs, recent findings on gonadal (mal)development in DSD patients, and possible correlations between the patient's phenotype and his/her risk for germ cell tumor development.
Collapse
Affiliation(s)
- M Cools
- Division of Pediatric Endocrinology, Department of Pediatrics, University Hospital Ghent and Ghent University, Belgium. martine.cools @ ugent.be
| | | | | | | | | |
Collapse
|
31
|
Yeung BH, Wan HT, Law AY, Wong CK. Endocrine disrupting chemicals: Multiple effects on testicular signaling and spermatogenesis. SPERMATOGENESIS 2011; 1:231-239. [PMID: 22319671 PMCID: PMC3271665 DOI: 10.4161/spmg.1.3.18019] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 09/04/2011] [Accepted: 09/05/2011] [Indexed: 02/01/2023]
Abstract
In the past 200 years, an enormous number of synthetic chemicals with diverse structural features have been produced for industrial, medical and domestic purposes. These chemicals, originally thought to have little or no biological toxicity, are widely used in our daily lives as well as are commonly present in foods. It was not until the first World Wildlife Federation Wingspread Conference held in 1994 were concerns about the endocrine disrupting (ED) effects of these chemicals articulated. The potential hazardous effects of endocrine disrupting chemicals (EDCs) on human health and ecological well-being are one of the global concerns that affect the health and propagation of human beings. Considerable numbers of studies indicated that endocrine disruption is linked to "the developmental basis of adult disease," highlighting the significant effects of EDC exposure on a developing organism, leading to the propensity of an individual to develop a disease or dysfunction in later life. In this review, we intend to provide environmental, epidemiological and experimental data to associate pollutant exposure with reproductive disorders, in particular on the development and function of the male reproductive system. Possible effects of pollutant exposure on the processes of embryonic development, like sex determination and masculinization are described. In addition, the effects of pollutant exposure on hypothalamus-pituitary-gonadal axis, testicular signaling, steroidogenesis and spermatogenesis are also discussed.
Collapse
Affiliation(s)
- Bonnie Hy Yeung
- Croucher Institute of Environmental Sciences; Department of Biology; Hong Kong Baptist University; Hong Kong
| | | | | | | |
Collapse
|
32
|
Nilsson EE, Schindler R, Savenkova MI, Skinner MK. Inhibitory actions of Anti-Müllerian Hormone (AMH) on ovarian primordial follicle assembly. PLoS One 2011; 6:e20087. [PMID: 21637711 PMCID: PMC3103528 DOI: 10.1371/journal.pone.0020087] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 04/17/2011] [Indexed: 11/18/2022] Open
Abstract
The current study was designed to investigate the actions of Anti-Müllerian Hormone (AMH) on primordial follicle assembly. Ovarian primordial follicles develop from the breakdown of oocyte nests during fetal development for the human and immediately after birth in rodents. AMH was found to inhibit primordial follicle assembly and decrease the initial primordial follicle pool size in a rat ovarian organ culture. The AMH expression was found to be primarily in the stromal tissue of the ovaries at this period of development, suggesting a stromal-epithelial cell interaction for primordial follicle assembly. AMH was found to promote alterations in the ovarian transcriptome during primordial follicle assembly with over 200 genes with altered expression. A gene network was identified suggesting a potential central role for the Fgf2/Nudt6 antisense transcript in the follicle assembly process. A number of signal transduction pathways are regulated by AMH actions on the ovarian transcriptome, in particular the transforming growth factor – beta (TGFß) signaling process. AMH is the first hormone/protein shown to have an inhibitory action on primordial follicle assembly. Due to the critical role of the primordial follicle pool size for female reproduction, elucidation of factors, such as AMH, that regulate the assembly process will provide insights into potential therapeutics to manipulate the pool size and female reproduction.
Collapse
Affiliation(s)
- Eric E. Nilsson
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, Washington, United States of America
| | - Ryan Schindler
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, Washington, United States of America
| | - Marina I. Savenkova
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, Washington, United States of America
| | - Michael K. Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, Washington, United States of America
- * E-mail:
| |
Collapse
|
33
|
Park S, Zeidan K, Shin JS, Taketo T. SRY upregulation of SOX9 is inefficient and delayed, allowing ovarian differentiation, in the B6.Y(TIR) gonad. Differentiation 2011; 82:18-27. [PMID: 21592645 DOI: 10.1016/j.diff.2011.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 04/22/2011] [Accepted: 04/27/2011] [Indexed: 11/18/2022]
Abstract
SRY on the Y-chromosome acts as a transcription factor to initiate testicular differentiation in mammals. Sox9 is a SRY target gene, upregulated immediately after Sry expression, and plays a key role in testicular differentiation. In the present study, we examined the expression of SRY and SOX9 proteins in the B6.Y(TIR) gonad, which undergoes partial or complete sex reversal. The results show that the ontogeny of SRY expression in the B6.Y(TIR) gonad was comparable with that in the B6.XY gonad. On the other hand, while SOX9 expression immediately followed SRY expression in the B6.XY gonad, it was considerably delayed compared to SRY expression in the B6.Y(TIR) gonad or SOX9 expression in the B6.XY gonad. Although SOX9 expression reached the entire gonad at a time point, it was downregulated and became restricted to the central area in which testis cords were organized. MIS, a marker of Sertoli cells, appeared only in well-organized testis cords. We speculate that the SRY protein from the Y(TIR)-chromosome is inefficient in upregulating the Sox9 gene on the B6 background, allowing the initiation of ovarian differentiation.
Collapse
Affiliation(s)
- Stephanie Park
- Department of Biology, McGill University, Royal Victoria Hospital, 687 Pine Avenue West, Montreal, Quebec, Canada H3A 1A1
| | | | | | | |
Collapse
|
34
|
Farhat A, Philibert P, Sultan C, Poulat F, Boizet-Bonhoure B. Hematopoietic-Prostaglandin D2 synthase through PGD2 production is involved in the adult ovarian physiology. J Ovarian Res 2011; 4:3. [PMID: 21352547 PMCID: PMC3050850 DOI: 10.1186/1757-2215-4-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 02/25/2011] [Indexed: 02/01/2023] Open
Abstract
Background The prostaglandin D2 (PGD2) pathway is involved in numerous biological processes and while it has been identified as a partner of the embryonic sex determining male cascade, the roles it plays in ovarian function remain largely unknown. PGD2 is secreted by two prostaglandin D synthases (Pgds); the male-specific lipocalin (L)-Pgds and the hematopoietic (H)-Pgds. Methods To study the expression of the Pgds in the adult ovary, in situ hybridization were performed. Then, to evaluate the role of H-Pgds produced PGD2 in the ovarian physiology, adult female mice were treated with HQL-79, a specific inhibitor of H-Pgds enzymatic activity. The effects on expression of the gonadotrophin receptors FshR and LhR, steroidogenic genes Cyp11A1, StAR and on circulating progesterone and estradiol, were observed. Results We report the localization of H-Pgds mRNA in the granulosa cells from the primary to pre-ovulatory follicles. We provide evidence of the role of H-Pgds-produced PGD2 signaling in the FSH signaling through increased FshR and LhR receptor expression. This leads to the activation of steroidogenic Cyp11A1 and StAR gene expression leading to progesterone secretion, independently on other prostanoid-synthetizing mechanisms. We also identify a role whereby H-Pgds-produced PGD2 is involved in the regulation of follicular growth through inhibition of granulosa cell proliferation in the growing follicles. Conclusions Together, these results show PGD2 signaling to interfere with FSH action within granulosa cells, thus identifying an important and unappreciated role for PGD2 signaling in modulating the balance of proliferation, differentiation and steroidogenic activity of granulosa cells.
Collapse
Affiliation(s)
- Andalib Farhat
- Institut de Génétique Humaine, Department of Genetic and Development, CNRS UPR1142, 141, rue de la Cardonille, 34396 Montpellier CEDEX5, France.
| | | | | | | | | |
Collapse
|
35
|
Genetic Disorders of Sex Differentiation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 707:91-9. [DOI: 10.1007/978-1-4419-8002-1_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
36
|
Kobayashi Y, Horiguchi R, Nozu R, Nakamura M. Expression and localization of forkhead transcriptional factor 2 (Foxl2) in the gonads of protogynous wrasse, Halichoeres trimaculatus. Biol Sex Differ 2010; 1:3. [PMID: 21208465 PMCID: PMC3010103 DOI: 10.1186/2042-6410-1-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 11/04/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Three-spot wrasse, Halichoeres trimaculatus, is a marine protogynous hermaphrodite fish. Individuals mature either as initial phase (IP) males or females. Appropriate social cues induce the sex change from IP female to terminal phase (TP) male. However, the molecular mechanisms behind such a sex change remain largely unknown. Recently, the forkhead transcription factor 2 (Foxl2) was identified as an essential regulator of vertebrate ovarian development/function/phenotype. Inspired by this information, we characterized the expression patterns of Foxl2 in the protogynous wrasse assuming Foxl2 as the female-specific marker in this species. METHODS First, we clonedFoxl2 cDNA from ovary by reverse transcription polymerase chain reaction (RT-PCR) followed by rapid amplification of cDNA ends (RACE). Next, we analysed expression pattern of Foxl2 messenger RNA (mRNA) and protein in gonads of different sexual phases by real time quantitative PCR assay and flour fluorescence immunohistochemical method, respectively. Additionally, we studied the changes in Foxl2 expression pattern during aromatase inhibitor (AI)-induced sex change. RESULTS The amino acid sequence (306 AA) of wrasse Foxl2, especially the forkhead domain, shows high identity with that of other reported teleost Foxl2s. Quite unexpectedly, no sexual dimorphism was observable between the testes and ovary in the expression pattern of Foxl2. In female phase fish, signals for Foxl2 protein were detectable in the granulosa cells, but not the theca cells. Transcript levels of Foxl2 in the testes of IP and TP males were identical to that in the ovaries of females and, further, Foxl2 protein was found to be localized in the interstitial cells including tubules and Leydig cells. Treatment with AI induced sex change in male gonads and an up-regulation was seen in the expression of Foxl2 in these gonads. CONCLUSIONS Unlike in other vertebrates, including teleosts, Foxl2 may have a different role in the naturally sex changing fishes.
Collapse
Affiliation(s)
- Yasuhisa Kobayashi
- Sesoko Station, Tropical Biosphere Research Center, University of the Ryukyus, 3422 Sesoko, Motobu 905-0227, Okinawa, Japan
- Solution-Oriented Research for Science and Technology (SORST), Kawaguchi, Saitama, Japan
| | - Ryo Horiguchi
- Sesoko Station, Tropical Biosphere Research Center, University of the Ryukyus, 3422 Sesoko, Motobu 905-0227, Okinawa, Japan
- Solution-Oriented Research for Science and Technology (SORST), Kawaguchi, Saitama, Japan
| | - Ryo Nozu
- Sesoko Station, Tropical Biosphere Research Center, University of the Ryukyus, 3422 Sesoko, Motobu 905-0227, Okinawa, Japan
| | - Masaru Nakamura
- Sesoko Station, Tropical Biosphere Research Center, University of the Ryukyus, 3422 Sesoko, Motobu 905-0227, Okinawa, Japan
- Solution-Oriented Research for Science and Technology (SORST), Kawaguchi, Saitama, Japan
| |
Collapse
|
37
|
Bashamboo A, Ledig S, Wieacker P, Achermann J, McElreavey K. New technologies for the identification of novel genetic markers of disorders of sex development (DSD). Sex Dev 2010; 4:213-24. [PMID: 20820110 PMCID: PMC2948663 DOI: 10.1159/000314917] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Although the genetic basis of human sexual determination and differentiation has advanced considerably in recent years, the fact remains that in most subjects with disorders of sex development (DSD) the underlying genetic cause is unknown. Where pathogenic mutations have been identified, the phenotype can be highly variable, even within families, suggesting that other genetic variants are influencing the expression of the phenotype. This situation is likely to change, as more powerful and affordable tools become widely available for detailed genetic analyses. Here, we describe recent advances in comparative genomic hybridisation, sequencing by hybridisation and next generation sequencing, and we describe how these technologies will have an impact on our understanding of the genetic causes of DSD.
Collapse
Affiliation(s)
- A. Bashamboo
- Human Developmental Genetics Unit, Institut Pasteur, Paris, France
| | - S. Ledig
- Institute of Human Genetics, Westfalian-Wilhelms University Münster, Münster, Germany
| | - P. Wieacker
- Institute of Human Genetics, Westfalian-Wilhelms University Münster, Münster, Germany
| | - J. Achermann
- The Developmental Endocrinology Research Group, Clinical and Molecular Genetics Unit, UCL Institute of Child Health, London, UK
| | - K. McElreavey
- Human Developmental Genetics Unit, Institut Pasteur, Paris, France
| |
Collapse
|