1
|
Skjold V, Afanasyev S, Burgerhout E, Sveen L, Rørvik KA, Mota VFCN, Dessen JE, Krasnov A. Endocrine and Transcriptome Changes Associated with Testicular Growth and Differentiation in Atlantic Salmon ( Salmo salar L.). Curr Issues Mol Biol 2024; 46:5337-5351. [PMID: 38920991 PMCID: PMC11202266 DOI: 10.3390/cimb46060319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024] Open
Abstract
Sexual maturation of Atlantic salmon males is marked by dramatic endocrine changes and rapid growth of the testes, resulting in an increase in the gonad somatic index (GSI). We examined the association of gonadal growth with serum sex steroids, as well as pituitary and testicular gene expression levels, which were assessed with a DNA oligonucleotide microarray. The testes transcriptome was stable in males with a GSI < 0.08% despite the large difference between the smallest and the largest gonads. Fish with a GSI ≥ 0.23% had 7-17 times higher serum levels of five male steroids and a 2-fold increase in progesterone, without a change in cortisol and related steroids. The pituitary transcriptome showed an upregulation of the hormone-coding genes that control reproduction and behavior, and structural rearrangement was indicated by the genes involved in synaptic transmission and the differentiation of neurons. The observed changes in the abundance of testicular transcripts were caused by the regulation of transcription and/or disproportional growth, with a greater increase in the germinative compartment. As these factors could not be separated, the transcriptome results are presented as higher or lower specific activities (HSA and LSA). LSA was observed in 4268 genes, including many genes involved in various immune responses and developmental processes. LSA also included genes with roles in female reproduction, germinal cell maintenance and gonad development, responses to endocrine and neural regulation, and the biosynthesis of sex steroids. Two functional groups prevailed among HSA: structure and activity of the cilia (95 genes) and meiosis (34 genes). The puberty of A. salmon testis is marked by the predominance of spermatogenesis, which displaces other processes; masculinization; and the weakening of external regulation. Results confirmed the known roles of many genes involved in reproduction and pointed to uncharacterized genes that deserve attention as possible regulators of sexual maturation.
Collapse
Affiliation(s)
- Vetle Skjold
- The Norwegian Institute of Aquaculture, Nofima, 9291 Tromsø, Norway; (V.S.); (E.B.); (L.S.); (K.-A.R.); (J.-E.D.)
- Department of Mechanical Engineering and Technology Management, Norwegian University of Life Sciences, 1433 Ås, Norway;
| | - Sergey Afanasyev
- Sechenov Institute of Evolutionary Physiology and Biochemistry, 194223 Saint Petersburg, Russia;
| | - Erik Burgerhout
- The Norwegian Institute of Aquaculture, Nofima, 9291 Tromsø, Norway; (V.S.); (E.B.); (L.S.); (K.-A.R.); (J.-E.D.)
| | - Lene Sveen
- The Norwegian Institute of Aquaculture, Nofima, 9291 Tromsø, Norway; (V.S.); (E.B.); (L.S.); (K.-A.R.); (J.-E.D.)
| | - Kjell-Arne Rørvik
- The Norwegian Institute of Aquaculture, Nofima, 9291 Tromsø, Norway; (V.S.); (E.B.); (L.S.); (K.-A.R.); (J.-E.D.)
- Department of Mechanical Engineering and Technology Management, Norwegian University of Life Sciences, 1433 Ås, Norway;
| | | | - Jens-Erik Dessen
- The Norwegian Institute of Aquaculture, Nofima, 9291 Tromsø, Norway; (V.S.); (E.B.); (L.S.); (K.-A.R.); (J.-E.D.)
| | - Aleksei Krasnov
- The Norwegian Institute of Aquaculture, Nofima, 9291 Tromsø, Norway; (V.S.); (E.B.); (L.S.); (K.-A.R.); (J.-E.D.)
| |
Collapse
|
2
|
Lærkeholm Müller M, Busch AS, Ljubicic ML, Upners EN, Fischer MB, Hagen CP, Albrethsen J, Frederiksen H, Juul A, Andersson AM. Urinary concentration of phthalates and bisphenol A during minipuberty is associated with reproductive hormone concentrations in infant boys. Int J Hyg Environ Health 2023; 250:114166. [PMID: 37058994 DOI: 10.1016/j.ijheh.2023.114166] [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: 11/16/2022] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 04/16/2023]
Abstract
BACKGROUND The transient postnatal activation of the hypothalamic-pituitary-gonadal hormone axis is termed minipuberty and considered an important developmental period, which is highly sensitive to endocrine disruption. Here, we explore exposure-outcome associations during minipuberty between concentrations of potentially endocrine disrupting chemicals (EDCs) in urine of infant boys and their serum reproductive hormone concentrations. METHODS In total, 36 boys participating in the COPENHAGEN Minipuberty Study had data available for both urine biomarkers of target endocrine disrupting chemicals and reproductive hormones in serum from samples collected on the same day. Serum concentrations of reproductive hormones were measured by immunoassays or by LC-MS/MS. Urinary concentrations of metabolites of 39 non-persisting chemicals, including phthalates and phenolic compounds, were measured by LC-MS/MS. Nineteen chemicals had concentrations above the limit of detection in ≥50% of children and were included in data analysis. Associations of urinary phthalate metabolite and phenol concentrations (in tertiles) with hormone outcomes (age- and sex-specific SD-scores) were analysed by linear regression. Primarily, we focused on the EU regulated phthalates; butylbenzyl phthalate (BBzP), di-iso-butyl phthalate (DiBP), di-n-butyl phthalate (DnBP), and di-(2-ethylhexyl) phthalate (DEHP) as well as bisphenol A (BPA). Urinary metabolites of DiBP, DnBP and DEHP were summed and expressed as ∑DiBPm, ∑DnBPm and ∑DEHPm. RESULTS Compared to boys in the lowest ∑DnBPm tertile, urinary concentration of ∑DnBPm was associated with concurrent higher luteinizing hormone (LH) and anti-Müllerian hormone (AMH) SD-scores as well as lower testosterone/LH ratio in boys in the middle ∑DnBPm tertile (estimates (CI 95%) 0.79 (0.04; 1.54), 0.91 (0.13; 1.68), and -0.88 (-1.58;-0.19), respectively). Further, higher insulin-like peptide 3 (INSL3) SD-scores and lower DHEAS SD-score in boys in the highest ∑DnBPm tertile (0.91 (0.12; 1.70) and -0.85 (-1.51;-0.18), respectively) were observed. In addition, boys in the middle and highest ∑DEHPm tertile had higher LH (1.07 (0.35; 1.79) and 0.71 (-0.01; 1.43), respectively) and in the highest ∑DEHPm tertile also higher AMH (0.85 (0.10; 1.61)) concentration SD-scores, respectively. Boys in the highest BPA tertile had significantly higher AMH and lower DHEAS concentration compared to boys in the lowest BPA tertile (1.28 (0.54; 2.02) and -0.73 (-1.45; -0.01)), respectively. DISCUSSION Our findings indicate that exposure to chemicals with known or suspected endocrine disrupting potential, especially the EU-regulated DnBP, DEHP and BPA, may modify male reproductive hormone concentrations in infant boys suggesting that minipuberty is a critical window sensitive to endocrine disruption.
Collapse
Affiliation(s)
- Matilde Lærkeholm Müller
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Alexander Siegfried Busch
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; University of Münster, Department of General Pediatrics, Münster, Germany
| | - Marie Lindhardt Ljubicic
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Emmie N Upners
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Margit B Fischer
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Casper P Hagen
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Jakob Albrethsen
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Hanne Frederiksen
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anna-Maria Andersson
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
3
|
Wang L, Wang Y, Li B, Zhang Y, Song S, Ding W, Xu D, Zhao Z. BMP6 regulates AMH expression via SMAD1/5/8 in goat ovarian granulosa cells. Theriogenology 2023; 197:167-176. [PMID: 36525856 DOI: 10.1016/j.theriogenology.2022.11.045] [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: 07/20/2022] [Revised: 11/23/2022] [Accepted: 11/27/2022] [Indexed: 12/02/2022]
Abstract
Anti-Müllerian hormone (AMH) is produced by ovarian granulosa cells (GCs)and plays a major role in inhibiting the recruitment of primordial follicles and reducing the sensitivity of growing follicles to follicle-stimulating hormone (FSH). Bone morphogenetic protein 6 (BMP6) has similar spatiotemporal expression to AMH during follicular development, suggesting that BMP6 may regulate AMH expression. However, the specific mechanism by which BMP6 regulates AMH expression remains unclear. The objectives of this study were to examine the molecular pathway by which BMP6 regulates AMH expression. The results showed that BMP6 promoted the secretion and expression of AMH in goat ovarian GCs. Mechanistically, BMP6 upregulated the expression of sex-determining region Y-box 9 (SOX9) and GATA-binding factor 4 (GATA4), which was associated with the transcriptional initiation of AMH. AMH expression was significantly decreased by GATA4 knockdown. Moreover, BMP6 treatment promoted the phosphorylation of SMAD1/5/8, whereas inhibiting the SMAD1/5/8 signaling pathway significantly abolished BMP6-induced upregulation of AMH and GATA4 expression. Interestingly, the activation of SMAD1/5/8 alone did not affect the expression of AMH or GATA4. The results suggested that BMP6 upregulated GATA4 through the SMAD1/5/8 signaling pathway, which in turn promoted AMH expression.
Collapse
Affiliation(s)
- Lei Wang
- College of Animal Science and Technology, Southwest University,Beibei, Chongqing, 400715, PR China
| | - Yukun Wang
- College of Animal Science and Technology, Southwest University,Beibei, Chongqing, 400715, PR China
| | - Bijun Li
- College of Animal Science and Technology, Southwest University,Beibei, Chongqing, 400715, PR China
| | - Yiyu Zhang
- College of Animal Science and Technology, Southwest University,Beibei, Chongqing, 400715, PR China
| | - Shuaifei Song
- College of Animal Science and Technology, Southwest University,Beibei, Chongqing, 400715, PR China
| | - Wenfei Ding
- College of Animal Science and Technology, Southwest University,Beibei, Chongqing, 400715, PR China
| | - Dejun Xu
- College of Animal Science and Technology, Southwest University,Beibei, Chongqing, 400715, PR China.
| | - Zhongquan Zhao
- College of Animal Science and Technology, Southwest University,Beibei, Chongqing, 400715, PR China.
| |
Collapse
|
4
|
Moses MM, Mullen RD, Idowu DI, Maye P, Jamin SP, Behringer RR. A transgenic bacterial artificial chromosome approach to identify regulatory regions that direct Amhr2 and Osterix expression in Müllerian duct mesenchyme. Front Cell Dev Biol 2022; 10:1006087. [PMID: 36313563 PMCID: PMC9597298 DOI: 10.3389/fcell.2022.1006087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
A transgenic mouse approach using bacterial artificial chromosomes (BAC) was used to identify regulatory regions that direct Müllerian duct expression for Amhr2 and Osterix (Osx, also known as Sp7). Amhr2 encodes the receptor that mediates anti-Müllerian hormone (AMH) signaling for Müllerian duct regression in male embryos. Amhr2 is expressed in the Müllerian duct mesenchyme of both male and female embryos. A ∼147-kb BAC clone containing the Amhr2 locus was used to generate transgenic mice. The transgene was able to rescue the block in Müllerian duct regression of Amhr2-null males, suggesting that the BAC clone contains regulatory sequences active in male embryos. Osx is expressed in the developing skeleton of male and female embryos but is also an AMH-induced gene that is expressed in the Müllerian duct mesenchyme exclusively in male embryos. Osx-Cre transgenic mice were previously generated using a ∼204-kb BAC clone. Crosses of Osx-Cre mice to Cre-dependent lacZ reporter mice resulted in reporter expression in the developing skeleton and in the Müllerian duct mesenchyme of male but not female embryos. Osx-Cherry transgenic mice were previously generated using a 39-kb genomic region surrounding the Osx locus. Osx-Cherry embryos expressed red fluorescence in the developing skeleton and Müllerian duct mesenchyme of male but not female embryos. In addition, female Osx-Cherry embryos ectopically expressing human AMH from an Mt1-AMH transgene activated red fluorescence in the Müllerian duct mesenchyme. These results suggest that the 39-kb region used to generate Osx-Cherry contains male-specific Müllerian duct mesenchyme regulatory sequences that are responsive to AMH signaling. These BAC transgenic mouse approaches identify two distinct regions that direct Müllerian duct mesenchyme expression and contribute fundamental knowledge to define a gene regulatory network for sex differentiation.
Collapse
Affiliation(s)
- Malcolm M. Moses
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Graduate Program in Genetics and Epigenetics, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Rachel D. Mullen
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Daniel I. Idowu
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Peter Maye
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Soazik P. Jamin
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Université de Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), Rennes, France
| | - Richard R. Behringer
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Graduate Program in Genetics and Epigenetics, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
5
|
di Clemente N, Racine C, Pierre A, Taieb J. Anti-Müllerian Hormone in Female Reproduction. Endocr Rev 2021; 42:753-782. [PMID: 33851994 DOI: 10.1210/endrev/bnab012] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/26/2022]
Abstract
Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance, was shown to be synthesized by the ovary in the 1980s. This article reviews the main findings of the past 20 years on the regulation of the expression of AMH and its specific receptor AMHR2 by granulosa cells, the mechanism of action of AMH, the different roles it plays in the reproductive organs, its clinical utility, and its involvement in the principal pathological conditions affecting women. The findings in respect of regulation tell us that AMH and AMHR2 expression is mainly regulated by bone morphogenetic proteins, gonadotropins, and estrogens. It has now been established that AMH regulates the different steps of folliculogenesis and that it has neuroendocrine effects. On the other hand, the importance of serum AMH as a reliable marker of ovarian reserve and as a useful tool in the prediction of the polycystic ovary syndrome (PCOS) and primary ovarian failure has also been acknowledged. Last but not least, a large body of evidence points to the involvement of AMH in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Nathalie di Clemente
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Chrystèle Racine
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France.,Sorbonne Paris Cité, Paris-Diderot Université, Paris, France
| | - Alice Pierre
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| | - Joëlle Taieb
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| |
Collapse
|
6
|
Poulat F. Non-Coding Genome, Transcription Factors, and Sex Determination. Sex Dev 2021; 15:295-307. [PMID: 34727549 DOI: 10.1159/000519725] [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: 08/08/2021] [Accepted: 09/15/2021] [Indexed: 11/19/2022] Open
Abstract
In vertebrates, gonadal sex determination is the process by which transcription factors drive the choice between the testicular and ovarian identity of undifferentiated somatic progenitors through activation of 2 different transcriptional programs. Studies in animal models suggest that sex determination always involves sex-specific transcription factors that activate or repress sex-specific genes. These transcription factors control their target genes by recognizing their regulatory elements in the non-coding genome and their binding motifs within their DNA sequence. In the last 20 years, the development of genomic approaches that allow identifying all the genomic targets of a transcription factor in eukaryotic cells gave the opportunity to globally understand the function of the nuclear proteins that control complex genetic programs. Here, the major transcription factors involved in male and female vertebrate sex determination and the genomic profiling data of mouse gonads that contributed to deciphering their transcriptional regulation role will be reviewed.
Collapse
Affiliation(s)
- Francis Poulat
- Institute of Human Genetics, CNRS UMR9002 University of Montpellier, Montpellier, France
| |
Collapse
|
7
|
Song W, Xie Y, Sun M, Li X, Fitzpatrick CK, Vaux F, O'Malley KG, Zhang Q, Qi J, He Y. A duplicated amh is the master sex-determining gene for Sebastes rockfish in the Northwest Pacific. Open Biol 2021; 11:210063. [PMID: 34255977 PMCID: PMC8277470 DOI: 10.1098/rsob.210063] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Teleost fish are the most diverse group of vertebrates and provide opportunities to study the evolution of sex determination (SD) systems. Using genomic and functional analyses, we identified a male-specific duplication of anti-Müllerian hormone (amh) gene as the male master sex-determining (MSD) gene in Sebastes schlegelii. By resequencing 10 males and 10 females, we characterized a 5 kb-long fragment in HiC_Scaffold_12 as a male-specific region, which contained an amh gene (named amhy). We then demonstrated that amhy is a duplication of autosomal amh that was later translocated to the ancestral Y chromosome. amha and amhy shared high-nucleotide identity with the most significant difference being two insertions in intron 4 of amhy. Furthermore, amhy overexpression triggered female-to-male sex reversal in S. schlegelii, displaying its fundamental role in driving testis differentiation. We developed a PCR assay which successfully identified sexes in two species of northwest Pacific rockfish related to S. schlegelii. However, the PCR assay failed to distinguish the sexes in a separate clade of northeast Pacific rockfish. Our study provides new examples of amh as the MSD in fish and sheds light on the convergent evolution of amh duplication as the driving force of sex determination in different fish taxa.
Collapse
Affiliation(s)
- Weihao Song
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Yuheng Xie
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Minmin Sun
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Xuemei Li
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Cristín K Fitzpatrick
- State Fisheries Genomics Lab, Coastal Oregon Marine Experiment Station, Department of Fisheries and Wildlife, Hatfield Marine Science Center, Oregon State University, Newport, OR, USA
| | - Felix Vaux
- Department of Zoology, University of Otago, Dunedin, New Zealand
| | - Kathleen G O'Malley
- State Fisheries Genomics Lab, Coastal Oregon Marine Experiment Station, Department of Fisheries and Wildlife, Hatfield Marine Science Center, Oregon State University, Newport, OR, USA
| | - Quanqi Zhang
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Jie Qi
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, People's Republic of China
| | - Yan He
- MOE Key Laboratory of Molecular Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, People's Republic of China
| |
Collapse
|
8
|
Morohashi KI, Inoue M, Baba T. Coordination of Multiple Cellular Processes by NR5A1/Nr5a1. Endocrinol Metab (Seoul) 2020; 35:756-764. [PMID: 33397036 PMCID: PMC7803590 DOI: 10.3803/enm.2020.402] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 11/27/2020] [Indexed: 11/11/2022] Open
Abstract
The agenesis of the gonads and adrenal gland in revealed by knockout mouse studies strongly suggested a crucial role for Nr5a1 (SF-1 or Ad4BP) in organ development. In relation to these striking phenotypes, NR5A1/Nr5a1 has the potential to reprogram cells to steroidogenic cells, endow pluripotency, and regulate cell proliferation. However, due to limited knowledge regarding NR5A1 target genes, the mechanism by which NR5A1/Nr5a1 regulates these fundamental processes has remained unknown. Recently, newlyestablished technologies have enabled the identification of NR5A1 target genes related to multiple metabolic processes, as well as the aforementioned biological processes. Considering that active cellular processes are expected to be accompanied by active metabolism, NR5A1 may act as a key factor for processes such as cell differentiation, proliferation, and survival by coordinating these processes with cellular metabolism. A complete and definite picture of the cellular processes coordinated by NR5A1/Nr5a1 could be depicted by accumulating evidence of the potential target genes through whole genome studies.
Collapse
Affiliation(s)
- Ken-Ichirou Morohashi
- Division of Biology of Sex Differences, Graduate School of Medical Sciences, and Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| | - Miki Inoue
- Division of Biology of Sex Differences, Graduate School of Medical Sciences, and Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Baba
- Division of Biology of Sex Differences, Graduate School of Medical Sciences, and Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
9
|
Valeri C, Lovaisa MM, Racine C, Edelsztein NY, Riggio M, Giulianelli S, Venara M, Bedecarrás P, Ballerini MG, di Clemente N, Lamb CA, Schteingart HF, Rey RA. Molecular mechanisms underlying AMH elevation in hyperoestrogenic states in males. Sci Rep 2020; 10:15062. [PMID: 32934281 PMCID: PMC7492256 DOI: 10.1038/s41598-020-71675-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/06/2020] [Indexed: 02/08/2023] Open
Abstract
Anti-Müllerian hormone (AMH) is secreted by Sertoli cells of the testes from early fetal life until puberty, when it is downregulated by androgens. In conditions like complete androgen insensitivity syndrome (CAIS), AMH downregulation does not occur and AMH increases at puberty, due in part to follicle-stimulating hormone (FSH) effect. However, other conditions like Peutz-Jeghers syndrome (PJS), characterised by low FSH, also have increased AMH. Because both CAIS and PJS may present as hyperoestrogenic states, we tested the hypothesis that oestradiol (E2) upregulates AMH expression in peripubertal Sertoli cells and explored the molecular mechanisms potentially involved. The results showed that E2 is capable of inducing an upregulation of endogenous AMH and of the AMH promoter activity in the prepubertal Sertoli cell line SMAT1, signalling through ERα binding to a specific ERE sequence present on the hAMH promoter. A modest action was also mediated through the membrane oestrogen receptor GPER. Additionally, the existence of ERα expression in Sertoli cells in patients with CAIS was confirmed by immunohistochemistry. The evidence presented here provides biological plausibility to the hypothesis that testicular AMH production increases in clinical conditions in response to elevated oestrogen levels.
Collapse
Affiliation(s)
- Clara Valeri
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - María M Lovaisa
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Chrystèle Racine
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine (CRSA), 75012, Paris, France.,Institut Hospitalo-Universitaire ICAN, 75013, Paris, France.,Sorbonne Paris Cité, Paris-Diderot Université, 75013, Paris, France
| | - Nadia Y Edelsztein
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Marina Riggio
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), C1428ADN, Buenos Aires, Argentina
| | - Sebastián Giulianelli
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), C1428ADN, Buenos Aires, Argentina.,Instituto de Biología de Organismos Marinos, IBIOMAR-CCT (CENPAT-CONICET), U9120ACD, Puerto Madryn, Argentina
| | - Marcela Venara
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Patricia Bedecarrás
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - María G Ballerini
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Nathalie di Clemente
- Sorbonne Université, INSERM, Centre de Recherche Saint Antoine (CRSA), 75012, Paris, France.,Institut Hospitalo-Universitaire ICAN, 75013, Paris, France
| | - Caroline A Lamb
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), C1428ADN, Buenos Aires, Argentina
| | - Helena F Schteingart
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina. .,Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, C1121ABG, Buenos Aires, Argentina.
| |
Collapse
|
10
|
Melo LH, Melo RMC, Luz RK, Bazzoli N, Rizzo E. Expression of Vasa, Nanos2 and Sox9 during initial testicular development in Nile tilapia (Oreochromis niloticus) submitted to sex reversal. Reprod Fertil Dev 2020; 31:1637-1646. [PMID: 31097079 DOI: 10.1071/rd18488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 04/28/2019] [Indexed: 11/23/2022] Open
Abstract
Sexual differentiation and early gonadal development are critical events in vertebrate reproduction. In this study, the initial testis development and expression of the Vasa, Nanos2 and Sox9 proteins were examined in Nile tilapia Oreochromis niloticus submitted to induced sex reversal. To that end, 150O. niloticus larvae at 5 days post-hatching (dph) were kept in nurseries with no hormonal addition (control group) and 150 larvae were kept with feed containing 17α-methyltestosterone to induce male sex reversal (treated group). Morphological sexual differentiation of Nile tilapia occurred between 21 and 25 dph and sex reversal resulted in 94% males, whereas the control group presented 53% males. During sexual differentiation, gonocytes (Gon) were the predominant germ cells, which decreased and disappeared after that stage in both groups. Undifferentiated spermatogonia (Aund) were identified at 21 dph in the control group and at 23 dph in the treated group. Differentiated spermatogonia (Adiff) were found at 23 dph in both groups. Vasa and Nanos2 occurred in Gon, Aund and Adiff and there were no significant differences between groups. Vasa-labelled Adiff increased at 50 dph in both groups and Nanos2 presented a high proportion of labelled germ cells during sampling. Sertoli cells expressed Sox9 throughout the experiment and its expression was significantly greater during sexual differentiation in the control group. The results indicate that hormonal treatment did not alter initial testis development and expression of Vasa and Nanos2 in Nile tilapia, although lower expression of Sox9 and a delay in sexual differentiation was detected in the treated group.
Collapse
Affiliation(s)
- Luis H Melo
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, UFMG, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Rafael M C Melo
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, UFMG, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Ronald K Luz
- Laboratório de Aquacultura, Escola de Veterinária, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - Nilo Bazzoli
- Programa de Pós-Graduação em Biologia de Vertebrados, Pontifícia Universidade Católica de Minas Gerais, PUC Minas, Av. Dom José Gaspar 500, 30535-610 Belo Horizonte, Minas Gerais, Brazil
| | - Elizete Rizzo
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, UFMG, Av. Antônio Carlos 6627, 31270-901 Belo Horizonte, Minas Gerais, Brazil; and Corresponding author.
| |
Collapse
|
11
|
Rey RA. Biomarcadores de hipogonadismo masculino en la infancia y la adolescencia. ADVANCES IN LABORATORY MEDICINE 2020; 1:20190043. [PMCID: PMC10158747 DOI: 10.1515/almed-2019-0043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 01/19/2020] [Indexed: 06/28/2023]
Abstract
El eje hipotálamo-hipófiso-testicular es activo en la vida fetal y durante los primeros meses de la vida posnatal: la hipófisis secreta hormona luteinizante (LH) y folículo-estimulante (FSH), mientras que el testículo produce testosterona y factor insulino-símil 3 (INSL3) en las células de Leydig y hormona anti-Mülleriana (AMH) e inhibina B en las células de Sertoli. En la infancia, los niveles séricos de gonadotrofinas, testosterona y factor INSL3 disminuyen a valores prácticamente indetectables, pero los de AMH e inhibina B permanecen altos. En la pubertad, se reactivan las gonadotrofinas y la producción de testosterona e INSL3, aumenta la inhibina y disminuye la AMH, como signo de maduración de la célula de Sertoli. Sobre la base del conocimiento de la fisiología del desarrollo del eje, es posible utilizar clínicamente estos biomarcadores para interpretar la fisiopatología y diagnosticar las diferentes formas de hipogonadismo que pueden presentarse en la infancia y la adolescencia.
Collapse
Affiliation(s)
- Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET-FEI- División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo, 1330, C1425EFD, Buenos Aires, Argentina
| |
Collapse
|
12
|
Rey RA. Biomarkers of male hypogonadism in childhood and adolescence. ADVANCES IN LABORATORY MEDICINE 2020; 1:20200024. [PMID: 37363780 PMCID: PMC10159267 DOI: 10.1515/almed-2020-0024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 01/19/2020] [Indexed: 06/28/2023]
Abstract
Objectives The objective of this review was to characterize the use of biomarkers of male hypogonadism in childhood and adolescence. Contents The hypothalamic-pituitary-gonadal (HPG) axis is active during fetal life and over the first months of postnatal life. The pituitary gland secretes follicle stimulating hormone (FSH) and luteinizing hormone (LH), whereas the testes induce Leydig cells to produce testosterone and insulin-like factor 3 (INSL), and drive Sertoli cells to secrete anti-Müllerian hormone (AMH) and inhibin B. During childhood, serum levels of gonadotropins, testosterone and insulin-like 3 (INSL3) decline to undetectable levels, whereas levels of AMH and inhibin B remain high. During puberty, the production of gonadotropins, testosterone, and INSL3 is reactivated, inhibin B increases, and AMH decreases as a sign of Sertoli cell maturation. Summary and outlook Based on our knowledge of the developmental physiology of the HPG axis, these biomarkers can be used in clinical practice to interpret the physiopathology of hypogonadism. Additionally, these markers can have diagnostic value in different forms of hypogonadism that may appear during childhood and adolescence.
Collapse
Affiliation(s)
- Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET-FEI- División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD, Buenos Aires, Argentina
- Departamento de Histología, Biología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, C1121ABG, Buenos Aires, Argentina
| |
Collapse
|
13
|
Bizzarri C, Cappa M. Ontogeny of Hypothalamus-Pituitary Gonadal Axis and Minipuberty: An Ongoing Debate? Front Endocrinol (Lausanne) 2020; 11:187. [PMID: 32318025 PMCID: PMC7154076 DOI: 10.3389/fendo.2020.00187] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
The fetal hypothalamus-pituitary gonadal (HPG) axis begins to function during mid-gestation but its activity decreases during late pregnancy due to the suppressive effects of placental estrogens. Placental hormones drop immediately after birth, FSH and LH surge at around 1 week and peak between 1 and 3 months of life. The HPG axis is activated in both sexes, but a sexual dimorphism is evident with higher LH values in boys, while FSH prevails in girls. Both gonadotrophins decline in boys by around 6 months of age. In girls, LH declines at the same time as in boys, while FSH persists elevated up to 3 or 4 years of age. As a result of gonadotropin activation, testicular testosterone increases in males and ovarian estradiol rises in females. These events clinically translate into testicular and penile growth in boys, enlargement of uterus and breasts in girls. The functional impact of HPG axis activity in infancy on later reproductive function is uncertain. According to the perinatal programming theory, this period may represent an essential programming process. In boys, long-term testicular hormonal function and spermatogenesis seem to be, at least in part, regulated by minipuberty. On the contrary, the role of minipuberty in girls is still uncertain. Recently, androgen exposure during minipuberty has been correlated with later sex-typed behavior. Premature and/or SGA infants show significant differences in postnatal HPG axis activity in comparison to full-term infants and the consequences of these differences on later health and disease require further research. The sex-dimorphic HPG activation during mid-gestation is probably responsible for the body composition differences observed ad birth between boys and girls, with boys showing greater total body mass and lean mass, and a lower proportion of fat mass. Testosterone exposure during minipuberty further contributes to these differences and seems to be responsible for the significantly higher growth velocity observed in male infants. Lastly, minipuberty is a valuable "window of opportunity" for differential diagnosis of disorders of sex development and it represents the only time window before puberty when congenital hypogonadism can be diagnosed by the simple analysis of basal gonadotropin and gonadal hormone levels.
Collapse
|
14
|
Promoter Identification and Transcriptional Regulation of the Goose AMH Gene. Animals (Basel) 2019; 9:ani9100816. [PMID: 31623192 PMCID: PMC6826907 DOI: 10.3390/ani9100816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/28/2019] [Accepted: 10/10/2019] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Anti-Müllerian hormone (AMH) plays a vital role in the development of follicles. We found that the cloning nucleotide sequence of AMH was high homology in geese with other species. Several regulatory elements were identified and transcriptional factors were predicted in the AMH promoter sequence. Through a double-luciferase reporter assay, potential regulatory relationship spanning from −637 to −87 bp were identified. In addition, the mRNA expression of AMH gradually decreased during the development of follicles in geese. In the Chinese hamster ovary (CHO) cell line, the luciferase activity significantly increased by co-expression of AMH and GATA-4. However, when the binding sites of GATA-4 to the promoter of AMH were mutated, the luciferase activity significantly decreased. These results indicated that the transcription of AMH was activated by GATA-4 to inhibit the development of follicles in geese. Abstract Anti-Müllerian hormone (AMH) is recognized as a reliable marker of ovarian reserve. However, the regulatory mechanism of goose AMH gene remains poorly understood. In the present study, both the full-length coding sequence (CDS) and promoter sequence of goose AMH have been cloned. Its CDS consisted of 2013 nucleotides encoding 670 amino acids and the amino acid sequence contained two structural domain: AMH-N and transforming growth factor beta (TGF-β) domain. The obtained promoter sequence spanned from the −2386 bp to its transcription start site (ATG). Core promoter regions and regulatory elements were identified as well as transcription factors were predicted in its promoter sequence. The luciferase activity was the highest spanning from the −331 to −1 bp by constructing deletion promoter reporter vectors. In CHO cells, the luciferase activity significantly increased by co-expression of AMH and GATA binding protein 4 (GATA-4), while that significantly decreased by mutating the binding sites of GATA-4 located in the −778 and −1477 bp. Results from quantitative real-time polymerase chain reaction (qPCR) indicated that levels of AMH mRNA in geese granulosa layers decreased gradually with the increasing follicular diameter. Taken together, it could be concluded that the transcriptional activity of AMH was activated by GATA-4 to inhibit the development of small follicles in goose.
Collapse
|
15
|
Edelsztein NY, Racine C, di Clemente N, Schteingart HF, Rey RA. Androgens downregulate anti-Müllerian hormone promoter activity in the Sertoli cell through the androgen receptor and intact steroidogenic factor 1 sites. Biol Reprod 2019; 99:1303-1312. [PMID: 29985989 DOI: 10.1093/biolre/ioy152] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/06/2018] [Indexed: 12/14/2022] Open
Abstract
Testicular anti-Müllerian hormone (AMH) production is inhibited by androgens around pubertal onset, as observed under normal physiological conditions and in patients with precocious puberty. In agreement, AMH downregulation is absent in patients with androgen insensitivity. The molecular mechanisms underlying the negative regulation of AMH by androgens remain unknown. Our aim was to elucidate the mechanisms through which androgens downregulate AMH expression in the testis. A direct negative effect of androgens on the transcriptional activity of the AMH promoter was found using luciferase reporter assays in the mouse prepubertal Sertoli cell line SMAT1. A strong inhibition of AMH promoter activity was seen in the presence of both testosterone and DHT and of the androgen receptor. By site-directed mutagenesis and chromatin immunoprecipitation assays, we showed that androgen-mediated inhibition involved the binding sites for steroidogenic factor 1 (SF1) present in the proximal promoter of the AMH gene. In this study, we describe for the first time the mechanism behind AMH inhibition by androgens, as seen in physiological and pathological conditions in males. Inhibition of AMH promoter activity by androgens could be due to protein-protein interactions between the ligand-bound androgen receptor and SF1 or by blockage of SF1 binding to its sites on the AMH promoter.
Collapse
Affiliation(s)
- Nadia Y Edelsztein
- Centro de Investigaciones Endocrinológicas "Dr César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Chrystèle Racine
- Sorbonne Universitté, INSERM, Centre de Recherche Saint Antoine (CRSA), IHU ICAN, 75012 Paris, France
| | - Nathalie di Clemente
- Sorbonne Universitté, INSERM, Centre de Recherche Saint Antoine (CRSA), IHU ICAN, 75012 Paris, France
| | - Helena F Schteingart
- Centro de Investigaciones Endocrinológicas "Dr César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina.,Departamento de Histología, Biología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| |
Collapse
|
16
|
Edelsztein NY, Rey RA. Importance of the Androgen Receptor Signaling in Gene Transactivation and Transrepression for Pubertal Maturation of the Testis. Cells 2019; 8:E861. [PMID: 31404977 PMCID: PMC6721648 DOI: 10.3390/cells8080861] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 12/27/2022] Open
Abstract
Androgens are key for pubertal development of the mammalian testis, a phenomenon that is tightly linked to Sertoli cell maturation. In this review, we discuss how androgen signaling affects Sertoli cell function and morphology by concomitantly inhibiting some processes and promoting others that contribute jointly to the completion of spermatogenesis. We focus on the molecular mechanisms that underlie anti-Müllerian hormone (AMH) inhibition by androgens at puberty, as well as on the role androgens have on Sertoli cell tight junction formation and maintenance and, consequently, on its effect on proper germ cell differentiation and meiotic onset during spermatogenesis.
Collapse
Affiliation(s)
- Nadia Y Edelsztein
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) - CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina.
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE) - CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires C1425EFD, Argentina.
- Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina.
| |
Collapse
|
17
|
Moses MM, Behringer RR. A gene regulatory network for Müllerian duct regression. ENVIRONMENTAL EPIGENETICS 2019; 5:dvz017. [PMID: 31579527 PMCID: PMC6760261 DOI: 10.1093/eep/dvz017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 05/03/2023]
Abstract
Mammalian embryos initially develop progenitor tissues for both male and female reproductive tract organs, known as the Wolffian ducts and the Müllerian ducts, respectively. Ultimately, each individual develops a single set of male or female reproductive tract organs. Therefore, an essential step for sex differentiation is the regression of one duct and growth and differentiation of the other duct. In males, this requires Müllerian duct regression and Wolffian duct growth and differentiation. Müllerian duct regression is induced by the expression of Amh, encoding anti-Müllerian hormone, from the fetal testes. Subsequently, receptor-mediated signal transduction in mesenchymal cells surrounding the Müllerian duct epithelium leads to duct elimination. The genes that induce Amh transcription and the downstream signaling that results from Amh activity form a pathway. However, the molecular details of this pathway are currently unknown. A set of essential genes for AMH pathway function has been identified. More recently, transcriptome analysis of male and female Müllerian duct mesenchyme at an initial stage of regression has identified new genes that may mediate elimination of the Müllerian system. The evidence taken together can be used to generate an initial gene regulatory network describing the Amh pathway for Müllerian duct regression. An Amh gene regulatory network will be a useful tool to study Müllerian duct regression, sex differentiation, and its relationship to environmental influences.
Collapse
Affiliation(s)
- Malcolm M Moses
- Department of Genetics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Richard R Behringer
- Department of Genetics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
- Correspondence address. Department of Genetics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA. Tel: +713-834-6327; Fax: +713-834-6339; E-mail:
| |
Collapse
|
18
|
Salonia A, Rastrelli G, Hackett G, Seminara SB, Huhtaniemi IT, Rey RA, Hellstrom WJG, Palmert MR, Corona G, Dohle GR, Khera M, Chan YM, Maggi M. Paediatric and adult-onset male hypogonadism. Nat Rev Dis Primers 2019; 5:38. [PMID: 31147553 PMCID: PMC6944317 DOI: 10.1038/s41572-019-0087-y] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The hypothalamic-pituitary-gonadal axis is of relevance in many processes related to the development, maturation and ageing of the male. Through this axis, a cascade of coordinated activities is carried out leading to sustained testicular endocrine function, with gonadal testosterone production, as well as exocrine function, with spermatogenesis. Conditions impairing the hypothalamic-pituitary-gonadal axis during paediatric or pubertal life may result in delayed puberty. Late-onset hypogonadism is a clinical condition in the ageing male combining low concentrations of circulating testosterone and specific symptoms associated with impaired hormone production. Testosterone therapy for congenital forms of hypogonadism must be lifelong, whereas testosterone treatment of late-onset hypogonadism remains a matter of debate because of unclear indications for replacement, uncertain efficacy and potential risks. This Primer focuses on a reappraisal of the physiological role of testosterone, with emphasis on the critical interpretation of the hypogonadal conditions throughout the lifespan of the male individual, with the exception of hypogonadal states resulting from congenital disorders of sex development.
Collapse
Affiliation(s)
- Andrea Salonia
- Division of Experimental Oncology, Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
| | - Giulia Rastrelli
- Sexual Medicine and Andrology Unit Department of Experimental Clinical and Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
| | - Geoffrey Hackett
- Department of Urology, University of Bedfordshire, Bedfordshire, UK
| | - Stephanie B Seminara
- Harvard Reproductive Sciences Center and Reproductive Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ilpo T Huhtaniemi
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, London, UK
- Department of Physiology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas 'Dr César Bergadá' (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños R. Gutiérrez, Buenos Aires, Argentina
| | - Wayne J G Hellstrom
- Department of Urology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Mark R Palmert
- Division of Endocrinology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Departments of Paediatrics and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Giovanni Corona
- Sexual Medicine and Andrology Unit Department of Experimental Clinical and Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
- Endocrinology Unit, Medical Department, Azienda Usl Bologna Maggiore-Bellaria Hospital, Bologna, Italy
| | - Gert R Dohle
- Department of Urology, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Mohit Khera
- Scott Department of Urology, Baylor College of Medicine, Houston, TX, USA
| | - Yee-Ming Chan
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Mario Maggi
- Sexual Medicine and Andrology Unit Department of Experimental Clinical and Biomedical Sciences 'Mario Serio', University of Florence, Florence, Italy
- Istituto Nazionale Biostrutture e Biosistemi (INBB), Rome, Italy
| |
Collapse
|
19
|
Schteingart HF, Picard JY, Valeri C, Marshall I, Treton D, di Clemente N, Rey RA, Josso N. A mutation inactivating the distal SF1 binding site on the human anti-Müllerian hormone promoter causes persistent Müllerian duct syndrome. Hum Mol Genet 2019; 28:3211-3218. [DOI: 10.1093/hmg/ddz147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/13/2019] [Accepted: 06/16/2019] [Indexed: 01/10/2023] Open
Abstract
AbstractThe persistent Müllerian duct syndrome (PMDS) is a 46,XY disorder of sexual development characterized by the persistence of Müllerian duct derivatives, uterus and tubes, in otherwise normally masculinized males. The condition, transmitted as a recessive autosomal trait, is usually due to mutations in either the anti-Müllerian hormone (AMH) gene or its main receptor. Many variants of these genes have been described, all targeting the coding sequences. We report the first case of PMDS due to a regulatory mutation. The AMH promoter contains two binding sites for steroidogenic factor 1 (SF1), one at −102 and the other at −228. Our patient carries a single base deletion at −225, significantly decreasing its capacity for binding SF1, as measured by the electrophoresis mobility shift assay. Furthermore, by linking the AMH promoter to the luciferase gene, we show that the transactivation capacity of the promoter is significantly decreased by the mutation, in contrast to the disruption of the −102 binding site. To explain the difference in impact we hypothesize that SF1 could partially overcome the lack of binding to the −102 binding site by interacting with a GATA4 molecule linked to a nearby response element. We show that disruption of both the −102 SF1 and the −84 GATA response elements significantly decreases the transactivation capacity of the promoter. In conclusion, we suggest that the distance between mutated SF1 sites and potentially rescuing GATA binding motifs might play a role in the development of PMDS.
Collapse
Affiliation(s)
- Helena F Schteingart
- Centro de Investigaciones Endocrinológicas ‘Dr. César Bergadá’ (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Jean-Yves Picard
- Inserm UMR_S938, Centre de Recherche Saint Antoine, Sorbonne Université, IHU ICAN, Paris, France
| | - Clara Valeri
- Centro de Investigaciones Endocrinológicas ‘Dr. César Bergadá’ (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Ian Marshall
- Division of Pediatric Endocrinology, Rutgers-Robert Wood Johnson Medical School, Child Health Institute of New Jersey, New Brunswick, NJ, USA
| | - Dominique Treton
- Inserm UMR_S938, Centre de Recherche Saint Antoine, Sorbonne Université, IHU ICAN, Paris, France
| | - Nathalie di Clemente
- Inserm UMR_S938, Centre de Recherche Saint Antoine, Sorbonne Université, IHU ICAN, Paris, France
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas ‘Dr. César Bergadá’ (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD Buenos Aires, Argentina
| | - Nathalie Josso
- Inserm UMR_S938, Centre de Recherche Saint Antoine, Sorbonne Université, IHU ICAN, Paris, France
| |
Collapse
|
20
|
Urrutia M, Grinspon RP, Rey RA. Comparing the role of anti-Müllerian hormone as a marker of FSH action in male and female fertility. Expert Rev Endocrinol Metab 2019; 14:203-214. [PMID: 30880521 DOI: 10.1080/17446651.2019.1590197] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/01/2019] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Originally limited to the assessment of disorders of sex development, anti-Müllerian hormone (AMH) measurement has more recently been extended to several conditions affecting the reproductive axis in males and females. Follicle-stimulating hormone (FSH) regulation of gonadal function has been extensively studied, but its role on AMH production has been explored only recently. AREAS COVERED We addressed the relationship between FSH action on the gonads and the usefulness of AMH as a marker in conditions affecting the reproductive axis. EXPERT OPINION Sertoli cells are the most active cell population in the prepubertal testis. Serum AMH is an excellent marker of FSH action on Sertoli cell proliferation and function in patients with hypogonadotropic hypogonadism. Low serum AMH is expected to predict low sperm production and prompts initial FSH treatment followed by human chorionic gonadotropin (hCG) or luteinizing hormone (LH) addition. Gonadotropin treatment may be more effective if installed to mimic the postnatal activation stage of the hypothalamic-pituitary-testicular axis. In females, AMH secretion by small antral follicles is stimulated by FSH. Elevated AMH indicates increased follicle numbers and should be considered as a potential contraindication of gonadotropin treatment in infertile patients due to an increased risk of developing ovarian hyperstimulation syndrome.
Collapse
Affiliation(s)
- Mariela Urrutia
- a Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología , Hospital de Niños Ricardo Gutiérrez , Buenos Aires , Argentina
| | - Romina P Grinspon
- a Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología , Hospital de Niños Ricardo Gutiérrez , Buenos Aires , Argentina
| | - Rodolfo A Rey
- a Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología , Hospital de Niños Ricardo Gutiérrez , Buenos Aires , Argentina
- b Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| |
Collapse
|
21
|
Bouchard MF, Bergeron F, Grenier Delaney J, Harvey LM, Viger RS. In Vivo Ablation of the Conserved GATA-Binding Motif in the Amh Promoter Impairs Amh Expression in the Male Mouse. Endocrinology 2019; 160:817-826. [PMID: 30759208 PMCID: PMC6426834 DOI: 10.1210/en.2019-00047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 02/08/2019] [Indexed: 12/23/2022]
Abstract
GATA4 is an essential transcriptional regulator required for gonadal development, differentiation, and function. In the developing testis, proposed GATA4-regulated genes include steroidogenic factor 1 (Nr5a1), SRY-related HMG box 9 (Sox9), and anti-Müllerian hormone (Amh). Although some of these genes have been validated as genuine GATA4 targets, it remains unclear whether GATA4 is a direct regulator of endogenous Amh transcription. We used a CRISPR/Cas9-based approach to specifically inactivate or delete the sole GATA-binding motif of the proximal mouse Amh promoter. AMH mRNA and protein levels were assessed at developmental time points corresponding to elevated AMH levels: fetal and neonate testes in males and adult ovaries in females. In males, loss of GATA binding to the Amh promoter significantly reduced Amh expression. Although the loss of GATA binding did not block the initiation of Amh transcription, AMH mRNA and protein levels failed to upregulate in the developing fetal and neonate testis. Interestingly, adult male mice presented no anatomical anomalies and had no evidence of retained Müllerian duct structures, suggesting that AMH levels, although markedly reduced, were sufficient to masculinize the male embryo. In contrast to males, GATA binding to the Amh promoter was dispensable for Amh expression in the adult ovary. These results provide conclusive evidence that in males, GATA4 is a positive modulator of Amh expression that works in concert with other key transcription factors to ensure that the Amh gene is sufficiently expressed in a correct spatiotemporal manner during fetal and prepubertal testis development.
Collapse
Affiliation(s)
- Marie France Bouchard
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
| | - Francis Bergeron
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
| | - Jasmine Grenier Delaney
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
| | - Louis-Mathieu Harvey
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
| | - Robert S Viger
- Reproduction, Mother and Child Health, Centre de Recherche du CHU de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle, Quebec, Quebec, Canada
- Department of Obstetrics, Gynecology, and Reproduction, Université Laval, Quebec, Quebec, Canada
- Correspondence: Robert S. Viger, PhD, Reproduction, Mother and Child Health, Room T3-67, Centre de Recherche du CHU de Québec–Université Laval, 2705 Laurier Boulevard, Quebec, Quebec G1V 4G2, Canada. E-mail:
| |
Collapse
|
22
|
Wagner-Mahler K, Kurzenne JY, Gastaud F, Hoflack M, Panaia Ferrari P, Berard E, Giuliano F, Karmous-Benailly H, Moceri P, Jouannelle C, Bourcier M, Robart E, Morel Y. Is interstitial 8p23 microdeletion responsible of 46,XY gonadal dysgenesis? One case report from birth to puberty. Mol Genet Genomic Med 2019; 7:e558. [PMID: 30690934 PMCID: PMC6418366 DOI: 10.1002/mgg3.558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/09/2018] [Accepted: 12/02/2018] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Chromosome 8p deletions are associated with a variety of conditions, including cardiac abnormalities, mental, behavioral problems with variable morphotype and genitourinary anomalies in boys. METHODS We describe the follow-up over almost 15 years of a boy who initially presented with perineal hypospadias with a micropenis and cryptorchidism with 46,XY DSD. RESULTS Imaging, pathology, and hormonal exploration suggested gonadal dysgenesis. Further genetic studies were deemed necessary during follow-up. The child's further development recommended further genetic analyses. High-resolution analysis showed an interstitial deletion on the short arm of a chromosome 8: 46,XY,del(8)(p23.1p23.1). We reviewed the literature and found 102 cases including 54 boys: 62.7% had mental problems, 50.9% a dysmorphic disorder, 55.9% cardiac anomalies, and 46.3% of the boys had genitourinary anomalies. Our patient's genital abnormalities can be explained by the haploinsufficiency of the genes, such as GATA4 (OMIM 600576) that are included in the deleted area. CONCLUSION This case of severe 46,XY DSD raises the question of the role played by 8p23 microdeletion in gonadal dysgenesis. Clinicians are encouraged to look for this anomaly on chromosome 8 in cases of unexplained gonadal dysgenesis even when few signs suggestive of this anomaly are present.
Collapse
Affiliation(s)
- Kathy Wagner-Mahler
- Département de Pédiatrie, Centre Hospitalier de Nice, Nice, France.,Hôpitaux Pédiatriques de Nice CHU Lenval, Nice, France
| | - Jean-Yves Kurzenne
- Département de Pédiatrie, Centre Hospitalier de Nice, Nice, France.,Hôpitaux Pédiatriques de Nice CHU Lenval, Nice, France
| | | | - Marie Hoflack
- Hôpitaux Pédiatriques de Nice CHU Lenval, Nice, France
| | | | - Etienne Berard
- Département de Pédiatrie, Centre Hospitalier de Nice, Nice, France
| | | | | | - Pamela Moceri
- Département de Cardiologie, Centre Hospitalier de Nice, Nice, France
| | | | | | - Elise Robart
- Hôpitaux Pédiatriques de Nice CHU Lenval, Nice, France
| | - Yves Morel
- Centre Hospitalier Universitaire de Lyon - HCL GH Est, Centre de Biologie et Pathologie Est, Bron, France
| |
Collapse
|
23
|
Delssin AR, Sandoval MT, Ortiz MA, Alvarez BB. Development and differentiation of the reproductive system of Tropidurus catalanensis (Squamata: Tropiduridae). J Morphol 2019; 280:244-258. [PMID: 30653714 DOI: 10.1002/jmor.20940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/06/2018] [Accepted: 12/10/2018] [Indexed: 01/13/2023]
Abstract
Development and differentiation of the reproductive system in lizards begin in the embryonic period, although the stage and time of their occurrence vary according to populations and species. In this study, the events of the development and differentiation of the reproductive system of males and females of Tropidurus catalanensis were characterized during the embryonic, neonatal, and juvenile periods. Embryos at Stages 27, 34, 37, 40, and 41, neonates and juveniles, from Corrientes, Argentina, were analyzed. At Stage 27, the genital ridge was not observed but primordial germ cells were recorded in the yolk sac as well as the mesenteric mesenchyme, indicating the beginning of germ cell migration. Gonadal differentiation commenced at Stage 34. In males from Stage 37, the testes possessed seminiferous cords with Sertoli cells and spermatogonia, while in hatchlings seminiferous tubules and interstitial tissue with mature Leydig cells were present. Spermatogenesis was observed in a specimen of 51.9 mm snout-vent length, corresponding to the minimum reproductive size. In females, from Stage 37 until hatching, the ovaries had a cavernous medulla and a cortex with somatic cells and abundant oogonia. The onset of meiosis and folliculogenesis occurred in the juvenile period.
Collapse
Affiliation(s)
- Andrea Raquel Delssin
- Departamento de Biología, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Embriología Animal. Av. Libertad 5470, CP 3400, Corrientes, Argentina.,Universidad Nacional del Nordeste-Consejo Nacional de Investigaciones Científicas y Tecnológicas (UNNE-CONICET), FaCENA (CP 3400), Corrientes, Argentina
| | - María Teresa Sandoval
- Departamento de Biología, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Embriología Animal. Av. Libertad 5470, CP 3400, Corrientes, Argentina
| | - Martin Alejandro Ortiz
- Departamento de Biología, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Embriología Animal. Av. Libertad 5470, CP 3400, Corrientes, Argentina.,Universidad Nacional del Nordeste-Consejo Nacional de Investigaciones Científicas y Tecnológicas (UNNE-CONICET), FaCENA (CP 3400), Corrientes, Argentina
| | - Blanca Beatriz Alvarez
- Departamento de Biología, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Embriología Animal. Av. Libertad 5470, CP 3400, Corrientes, Argentina
| |
Collapse
|
24
|
Ovarian activity regulation by anti-Müllerian hormone in early stages of human female life, an overview. ANTHROPOLOGICAL REVIEW 2018. [DOI: 10.2478/anre-2018-0026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The present study aimed at describing the anti-Müllerian hormone (AMH), with special focus on molecular background for ovarian activity, in particular the role AMH plays in sex determination and gonadogenesis process in early stages of prenatal life and folliculogenesis in postnatal life. It is a review of the literature currently indexed and abstracted in MEDLINE, SCOPUS and Google Scholars. The process of sex determination and gonad differentiation occurring during embryogenesis was discussed along with underlying molecular mechanisms. In the postnatal life the impact of AMH on the process of folliculogenesis was described. Clinical use of recent findings was shown as well. Genetic studies and molecular analyses have demonstrated that AMH is highly conservative, indicating its significance in reproductive process on the background of evolutionary processes.
Collapse
|
25
|
Grinspon RP, Urrutia M, Rey RA. Male Central Hypogonadism in Paediatrics - the Relevance of Follicle-stimulating Hormone and Sertoli Cell Markers. EUROPEAN ENDOCRINOLOGY 2018; 14:67-71. [PMID: 30349597 PMCID: PMC6182919 DOI: 10.17925/ee.2018.14.2.67] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/02/2018] [Indexed: 11/24/2022]
Abstract
The definition of male hypogonadism, used in adult endocrinology, is not fully applicable to paediatrics. A clear understanding of the developmental physiology of the hypothalamic-pituitary-testicular axis is essential for the comprehension of the pathogenesis of hypogonadal states in boys and for the establishment of adequate definitions and classifications in paediatric ages. This is particularly true for central hypogonadism, usually called hypogonadotropic in adults. Because childhood is a period characterised by a physiological state of low gonadotropin and testosterone production, these markers of hypogonadism, typically used in adult endocrinology, are uninformative in the child. This review is focused on the physiological importance of prepubertal Sertoli cell markers - anti-Müllerian hormone (AMH) and inhibin B - and of the intratesticular actions of follicle-stimulating hormone (FSH) and testosterone during early infancy and the first stages of pubertal development. We discuss the role of FSH in regulating the proliferation of Sertoli cells - the main determinant of prepubertal testicular volume - and the secretion of AMH and inhibin B. We also address how intratesticular testosterone concentrations have different effects on the seminiferous tubule function in early infancy and during pubertal development.
Collapse
Affiliation(s)
- Romina P Grinspon
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), National Scientific and Technical Research Council (CONICET) - Fundación de Endocrinología Infantil (FEI) - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Mariela Urrutia
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), National Scientific and Technical Research Council (CONICET) - Fundación de Endocrinología Infantil (FEI) - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), National Scientific and Technical Research Council (CONICET) - Fundación de Endocrinología Infantil (FEI) - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina.,Departamento de Histología, Biología Celular, Embriologia y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
26
|
Grinspon RP, Gottlieb S, Bedecarrás P, Rey RA. Anti-Müllerian Hormone and Testicular Function in Prepubertal Boys With Cryptorchidism. Front Endocrinol (Lausanne) 2018; 9:182. [PMID: 29922225 PMCID: PMC5996917 DOI: 10.3389/fendo.2018.00182] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/04/2018] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The functional capacity of the testes in prepubertal boys with cryptorchidism before treatment has received very little attention. The assessment of testicular function at diagnosis could be helpful in the understanding of the pathophysiology of cryptorchidism and in the evaluation of the effect of treatment. Anti-Müllerian hormone is a well-accepted Sertoli cell biomarker to evaluate testicular function during childhood without the need for stimulation tests. OBJECTIVE The aim of the study was to assess testicular function in prepubertal children with cryptorchidism before orchiopexy, by determining serum anti-Müllerian hormone (AMH). We also evaluated serum gonadotropins and testosterone and looked for associations between testicular function and the clinical characteristics of cryptorchidism. MATERIALS AND METHODS We performed a retrospective, cross-sectional, analytical study at a tertiary pediatric public hospital. All clinical charts of patients admitted at the outpatient clinic, and recorded in our database with the diagnosis of cryptorchidism, were eligible. The main outcome measure of the study was the serum concentration of AMH. Secondary outcome measures were serum LH, FSH, and testosterone. For comparison, serum hormone levels from a normal population of 179 apparently normal prepubertal boys were used. RESULTS Out of 1,557 patients eligible in our database, 186 with bilateral and 124 with unilateral cryptorchidism were selected using a randomization software. Median AMH standard deviation score was below 0 in both the bilaterally and the unilaterally cryptorchid groups, indicating that testicular function was overall decreased in patients with cryptorchidism. Serum AMH was significantly lower in boys with bilateral cryptorchidism as compared with controls and unilaterally cryptorchid patients between 6 months and 1.9 years and between 2 and 8.9 years of age. Serum AMH below the normal range reflected testicular dysfunction in 9.5-36.5% of patients according to the age group in bilaterally cryptorchid boys and 6.3-16.7% in unilaterally cryptorchid boys. FSH was elevated in 8.1% and LH in 9.1% of boys with bilateral cryptorchidism, most of whom were anorchid. In patients with present testes, gonadotropins were only mildly elevated in less than 5% of the cases. Basal testosterone was mildly decreased in patients younger than 6 months old, and uninformative during childhood. CONCLUSION Prepubertal boys with cryptorchidism, especially those with bilaterally undescended gonads, have decreased AMH production. Although serum AMH may fall within the normal range, there is a considerable prevalence of testicular dysfunction during childhood in this frequent condition.
Collapse
Affiliation(s)
- Romina P. Grinspon
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Silvia Gottlieb
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Patricia Bedecarrás
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET – FEI – División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
- Departamento de Histología, Biología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- *Correspondence: Rodolfo A. Rey,
| |
Collapse
|
27
|
Recabarren SE, Recabarren M, Sandoval D, Carrasco A, Padmanabhan V, Rey R, Richter HG, Perez-Marin CC, Sir-Petermann T, Rojas-Garcia PP. Puberty arises with testicular alterations and defective AMH expression in rams prenatally exposed to testosterone. Domest Anim Endocrinol 2017; 61:100-107. [PMID: 28783504 DOI: 10.1016/j.domaniend.2017.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 06/15/2017] [Accepted: 06/16/2017] [Indexed: 11/20/2022]
Abstract
The male gonadal tissue can be a sensitive target to the reprogramming effects of testosterone (T) during prenatal development. We have demonstrated that male lambs born to dams receiving T during pregnancy-a model system to the polycystic ovary syndrome (PCOS)-show a decreased number of germ cells early in life, and when adult, a reduced amount of sperm and ejaculate volume. These findings are a key to put attention to the male offspring of women bearing PCOS, as they are exposed to increased levels of androgen during pregnancy which can reprogram their reproductive outcome. A possible origin of these defects can be a disruption in the expression of the anti-Müllerian hormone (AMH), due to its critical role in gonadal function at many postnatal stages. Therefore, we addressed the impact of prenatal T excess on the expression of AMH and factors related to its expression like AP2, SOX9, FSHR, and AR in the testicular tissue through real-time PCR during the peripubertal age. We also analyzed the testicular morphology and quantified the number of Sertoli cells and germ cells to evaluate any further defect in the testicle. Experiments were performed in rams at 24 wk of age, hence, prior puberty. The experimental animals (T-males) consisted of rams born to mothers receiving 30 mg testosterone twice a wk from Day 30 to 90 of pregnancy and then increased to 40 mg until Day 120 of pregnancy. The control males (C-males) were born to mothers receiving the vehicle of the hormone. We found a significant increase in the expression of the mRNA of AMH and SOX9, but not of the AP2, FHSR nor AR, in the T-males. Moreover, T-males showed a dramatic decrease in the number of germ cells, together with a decrease in the weight of their testicles. The findings of the present study show that before puberty, T-males are manifesting clear signs of disruption in the gonadal functions probably due to an alteration in the expression pattern of the AMH gene. The precise way by which T reprograms the expression of AMH gene remains to be established.
Collapse
Affiliation(s)
- S E Recabarren
- Laboratory of Animal Physiology and Endocrinology (FISENLAB), Faculty of Veterinary Sciences, University of Concepción, Chillán, Chile
| | - M Recabarren
- Laboratory of Animal Physiology and Endocrinology (FISENLAB), Faculty of Veterinary Sciences, University of Concepción, Chillán, Chile
| | - D Sandoval
- Laboratory of Animal Physiology and Endocrinology (FISENLAB), Faculty of Veterinary Sciences, University of Concepción, Chillán, Chile
| | - A Carrasco
- Laboratory of Animal Physiology and Endocrinology (FISENLAB), Faculty of Veterinary Sciences, University of Concepción, Chillán, Chile
| | - V Padmanabhan
- Departments of Pediatrics and the Reproductive Sciences Program, University of Michigan, Ann Arbor, Michigan, USA
| | - R Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina; Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - H G Richter
- Laboratory of Developmental Chronobiology (LDC), Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - C C Perez-Marin
- Department of Animal Medicine and Surgery, Faculty of Veterinary Medicine, University of Cordoba, Cordoba, Spain
| | - T Sir-Petermann
- Laboratory of Endocrinology and Metabolism, Department of Internal Medicine, Western Faculty of Medicine, University of Chile, Santiago, Chile
| | - P P Rojas-Garcia
- Laboratory of Animal Physiology and Endocrinology (FISENLAB), Faculty of Veterinary Sciences, University of Concepción, Chillán, Chile.
| |
Collapse
|
28
|
Convissar S, Armouti M, Fierro MA, Winston NJ, Scoccia H, Zamah AM, Stocco C. Regulation of AMH by oocyte-specific growth factors in human primary cumulus cells. Reproduction 2017; 154:745-753. [PMID: 28874516 DOI: 10.1530/rep-17-0421] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/15/2017] [Accepted: 09/05/2017] [Indexed: 12/21/2022]
Abstract
The regulation of AMH production by follicular cells is poorly understood. The purpose of this study was to determine the role of the oocyte-secreted factors, growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15), on AMH production in primary human cumulus cells. Cumulus cells from IVF patients were cultured with a combination of GDF9, BMP15, recombinant FSH and specific signaling inhibitors. Stimulation with GDF9 or BMP15 separately had no significant effect on AMH mRNA levels. In contrast, simultaneous stimulation with GDF9 and BMP15 (G + B) resulted in a significant increase in AMH mRNA expression. Increasing concentration of G + B (0.6, 2.5, 5 and 10 ng/mL) stimulated AMH in a dose-dependent manner, showing a maximal effect at 5 ng/mL. Western blot analyses revealed an average 16-fold increase in AMH protein levels in cells treated with G + B when compared to controls. FSH co-treatment decreased the stimulation of AMH expression by G + B. The stimulatory effect of G + B on the expression of AMH was significantly decreased by inhibitors of the SMAD2/3 signaling pathway. These findings show for the first time that AMH production is regulated by oocyte-secreted factors in primary human cumulus cells. Moreover, our novel findings establish that the combination of GDF9 + BMP15 potently stimulates AMH expression.
Collapse
Affiliation(s)
- Scott Convissar
- Department of Physiology and BiophysicsThe University of Illinois at Chicago College of Medicine, Chicago, Illinois, USA
| | - Marah Armouti
- Department of Physiology and BiophysicsThe University of Illinois at Chicago College of Medicine, Chicago, Illinois, USA
| | - Michelle A Fierro
- Division of Reproductive Endocrinology and InfertilityDepartment of Obstetrics and Gynecology, The University of Illinois at Chicago College of Medicine, Chicago, Illinois, USA
| | - Nicola J Winston
- Division of Reproductive Endocrinology and InfertilityDepartment of Obstetrics and Gynecology, The University of Illinois at Chicago College of Medicine, Chicago, Illinois, USA
| | - Humberto Scoccia
- Division of Reproductive Endocrinology and InfertilityDepartment of Obstetrics and Gynecology, The University of Illinois at Chicago College of Medicine, Chicago, Illinois, USA
| | - A Musa Zamah
- Division of Reproductive Endocrinology and InfertilityDepartment of Obstetrics and Gynecology, The University of Illinois at Chicago College of Medicine, Chicago, Illinois, USA
| | - Carlos Stocco
- Department of Physiology and BiophysicsThe University of Illinois at Chicago College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
29
|
Otake S, Park MK. Expressional changes of AMH signaling system in the quail testis induced by photoperiod. Reproduction 2016; 152:575-589. [DOI: 10.1530/rep-16-0175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 08/31/2016] [Indexed: 12/29/2022]
Abstract
Gonadal sex differentiation proceeds by the interplay of various genes including the transcription factors and secretory factors in a complex network. The sex-differentiating genes are expressed not only during early sex differentiation but also throughout the gonadal development and even in the adult gonads. In addition, the evidence that they actually function in the adult gonads have been accumulated from the studies using the conditional knockout mice. However, many previous studies were focused on one single gene though those genes function in a network. In this study, the expressions of various sex-differentiating genes were analyzed simultaneously in the adult testis of the Japanese quail (Coturnix japonica), whose testicular functions are dramatically changed by altering the photoperiod, to elucidate the roles of them in the adult gonad. Anti-Müllerian hormone (AMH) was significantly upregulated in the regressed testis induced by the short-day condition. The expressions of the transcription factors that promoteAMHexpression in mammals (SF1,SOX9,WT1andGATA4) were also increased in the regressed testis. Moreover, AMH receptor (AMHR2) showed similar expression pattern to its ligand. We also analyzed the expressions of other transforming growth factor beta (TGFB) superfamily members and their receptors. The expressions of the ligands and receptors of TGFB family, and follistatin and betaglycan in addition to inhibin subunits were increased in the regressed testis. These results suggest that AMH is involved in the adult testicular functions of the Japanese quail together with other TGFB superfamily members.
Collapse
|
30
|
Edelsztein NY, Grinspon RP, Schteingart HF, Rey RA. Anti-Müllerian hormone as a marker of steroid and gonadotropin action in the testis of children and adolescents with disorders of the gonadal axis. INTERNATIONAL JOURNAL OF PEDIATRIC ENDOCRINOLOGY 2016; 2016:20. [PMID: 27799946 PMCID: PMC5084469 DOI: 10.1186/s13633-016-0038-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 10/12/2016] [Indexed: 12/17/2022]
Abstract
In pediatric patients, basal testosterone and gonadotropin levels may be uninformative in the assessment of testicular function. Measurement of serum anti-Müllerian hormone (AMH) has become increasingly widespread since it provides information about the activity of the male gonad without the need for dynamic tests, and also reflects the action of FSH and androgens within the testis. AMH is secreted in high amounts by Sertoli cells from fetal life until the onset of puberty. Basal AMH expression is not dependent on gonadotropins or sex steroids; however, FSH further increases and testosterone inhibits AMH production. During puberty, testosterone induces Sertoli cell maturation, and prevails over FSH on AMH regulation. Therefore, AMH production decreases. Serum AMH is undetectable in patients with congenital or acquired anorchidism, or with complete gonadal dysgenesis. Low circulating levels of AMH may reflect primary testicular dysfunction, e.g. in certain patients with cryptorchidism, monorchidism, partial gonadal dysgenesis, or central hypogonadism. AMH is low in boys with precocious puberty, but it increases to prepubertal levels after successful treatment. Conversely, serum AMH remains at high, prepubertal levels in boys with constitutional delay of puberty. Serum AMH measurements are useful, together with testosterone determination, in the diagnosis of patients with ambiguous genitalia: both are low in patients with gonadal dysgenesis, including ovotesticular disorders of sex development, testosterone is low but AMH is in the normal male range or higher in patients with disorders of androgen synthesis, and both hormones are normal or high in patients with androgen insensitivity. Finally, elevation of serum AMH above normal male prepubertal levels may be indicative of rare cases of sex-cord stromal tumors or Sertoli cell-limited disturbance in the McCune Albright syndrome.
Collapse
Affiliation(s)
- Nadia Y Edelsztein
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina ; Departamento de Ecología, Genética y Evolución, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Romina P Grinspon
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Helena F Schteingart
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina ; Departamento de Biología Celular, Histología, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
31
|
Pashaiasl M, Ebrahimi M, Ebrahimie E. Identification of the key regulating genes of diminished ovarian reserve (DOR) by network and gene ontology analysis. Mol Biol Rep 2016; 43:923-37. [PMID: 27324248 DOI: 10.1007/s11033-016-4025-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 05/30/2016] [Indexed: 11/30/2022]
Abstract
Diminished ovarian reserve (DOR) is one of the reasons for infertility that not only affects both older and young women. Ovarian reserve assessment can be used as a new prognostic tool for infertility treatment decision making. Here, up- and down-regulated gene expression profiles of granulosa cells were analysed to generate a putative interaction map of the involved genes. In addition, gene ontology (GO) analysis was used to get insight intol the biological processes and molecular functions of involved proteins in DOR. Eleven up-regulated genes and nine down-regulated genes were identified and assessed by constructing interaction networks based on their biological processes. PTGS2, CTGF, LHCGR, CITED, SOCS2, STAR and FSTL3 were the key nodes in the up-regulated networks, while the IGF2, AMH, GREM, and FOXC1 proteins were key in the down-regulated networks. MIRN101-1, MIRN153-1 and MIRN194-1 inhibited the expression of SOCS2, while CSH1 and BMP2 positively regulated IGF1 and IGF2. Ossification, ovarian follicle development, vasculogenesis, sequence-specific DNA binding transcription factor activity, and golgi apparatus are the major differential groups between up-regulated and down-regulated genes in DOR. Meta-analysis of publicly available transcriptomic data highlighted the high coexpression of CTGF, connective tissue growth factor, with the other key regulators of DOR. CTGF is involved in organ senescence and focal adhesion pathway according to GO analysis. These findings provide a comprehensive system biology based insight into the aetiology of DOR through network and gene ontology analyses.
Collapse
Affiliation(s)
- Maryam Pashaiasl
- Women's Reproductive Health Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran. .,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mansour Ebrahimi
- Bioinformatics Research Group and Department of Biology, Qom University, Qom, Iran
| | - Esmaeil Ebrahimie
- Division of Information Technology, Engineering and the Environment, School of Information Technology and Mathematical Sciences, University of South Australia, Adelaide, Australia.,Institute of Biotechnology, Shiraz University, Shiraz, Iran.,Department of Genetics and Evolution, School of Biological Sciences, The University of Adelaide, Adelaide, Australia.,School of Biological Sciences, Faculty of Science and Engineering, Flinders University, Adelaide, Australia
| |
Collapse
|
32
|
Grinspon RP, Habib C, Bedecarrás P, Gottlieb S, Rey RA. Compensatory function of the remaining testis is dissociated in boys and adolescents with monorchidism. Eur J Endocrinol 2016; 174:399-407. [PMID: 26671976 DOI: 10.1530/eje-15-0938] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/15/2015] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Compensatory hypertrophy has been classically described in patients with monorchidism. However, it remains unclear whether there is a functional compensatory activity of the different cell populations. Our aim was to assess the functional capacity of the solitary testis in monorchid males from infancy through puberty in order to determine whether the remaining gonad is capable of compensating the functional activity of Sertoli and Leydig cells of the absent gonad. DESIGN In a retrospective, cross-sectional, analytical study performed at a tertiary paediatric public hospital, we included 89 boys with monorchidism and 358 healthy controls, aged 6 months-18 years. Testicular volume and circulating levels of reproductive hormones were compared between patients with monorchidism and normal boys. Serum anti-Müllerian hormone (AMH) and FSH were used as biomarkers of the functional mass of prepubertal Sertoli cells, whereas serum testosterone and LH were used as biomarkers of Leydig cells. RESULTS In the vast majority of the cases, the testicular volume of monorchid boys was smaller than the sum of the volume of both testes of healthy controls. Serum AMH was lower and FSH was higher in patients with monorchidism than in controls aged <3 and >13 years. Serum testosterone and LH did not differ significantly between patients and controls. CONCLUSION In boys and adolescents with monorchidism, there is a dissociated capacity of the remaining testis to compensate for the absence of the other gonad: while Leydig cell function is largely compensated, Sertoli cell proliferation and function was lower than in controls.
Collapse
Affiliation(s)
- Romina P Grinspon
- Centro de Investigaciones Endocrinológicas 'Dr. César Bergadá' (CEDIE)CONICET - FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD Buenos Aires, Argentina andDepartamento de HistologíaBiología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carolina Habib
- Centro de Investigaciones Endocrinológicas 'Dr. César Bergadá' (CEDIE)CONICET - FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD Buenos Aires, Argentina andDepartamento de HistologíaBiología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Patricia Bedecarrás
- Centro de Investigaciones Endocrinológicas 'Dr. César Bergadá' (CEDIE)CONICET - FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD Buenos Aires, Argentina andDepartamento de HistologíaBiología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Silvia Gottlieb
- Centro de Investigaciones Endocrinológicas 'Dr. César Bergadá' (CEDIE)CONICET - FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD Buenos Aires, Argentina andDepartamento de HistologíaBiología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rodolfo A Rey
- Centro de Investigaciones Endocrinológicas 'Dr. César Bergadá' (CEDIE)CONICET - FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD Buenos Aires, Argentina andDepartamento de HistologíaBiología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina Centro de Investigaciones Endocrinológicas 'Dr. César Bergadá' (CEDIE)CONICET - FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, C1425EFD Buenos Aires, Argentina andDepartamento de HistologíaBiología Celular, Embriología y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
33
|
|
34
|
Pfennig F, Standke A, Gutzeit HO. The role of Amh signaling in teleost fish--Multiple functions not restricted to the gonads. Gen Comp Endocrinol 2015; 223:87-107. [PMID: 26428616 DOI: 10.1016/j.ygcen.2015.09.025] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 09/24/2015] [Accepted: 09/25/2015] [Indexed: 12/16/2022]
Abstract
This review summarizes the important role of Anti-Müllerian hormone (Amh) during gonad development in fishes. This Tgfβ-domain bearing hormone was named after one of its known functions, the induction of the regression of Müllerian ducts in male mammalian embryos. Later in development it is involved in male and female gonad differentiation and extragonadal expression has been reported in mammals as well. Teleosts lack Müllerian ducts, but they have amh orthologous genes. amh expression is reported from 21 fish species and possible regulatory interactions with further factors like sex steroids and gonadotropic hormones are discussed. The gonadotropin Fsh inhibits amh expression in all fish species studied. Sex steroids show no consistent influence on amh expression. Amh is produced in male Sertoli cells and female granulosa cells and inhibits germ cell proliferation and differentiation as well as steroidogenesis in both sexes. Therefore, Amh might be a central player in gonad development and a target of gonadotropic Fsh. Furthermore, there is evidence that an Amh-type II receptor is involved in germ cell regulation. Amh and its corresponding type II receptor are also present in brain and pituitary, at least in some teleosts, indicating additional roles of Amh effects in the brain-pituitary-gonadal axis. Unraveling Amh signaling is important in stem cell research and for reproduction as well as for aquaculture and in environmental science.
Collapse
Affiliation(s)
- Frank Pfennig
- Institut für Zoologie, TU Dresden, D-01062 Dresden, Germany.
| | - Andrea Standke
- Institut für Zoologie, TU Dresden, D-01062 Dresden, Germany
| | | |
Collapse
|
35
|
Bernard V, Bouvattier C, Christin-Maitre S. [Therapeutic issues concerning male fertility]. ANNALES D'ENDOCRINOLOGIE 2015; 75 Suppl 1:S13-20. [PMID: 25617918 DOI: 10.1016/s0003-4266(14)70023-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Men reproductive health has long been ignored although it is responsible for 50% of couple's infertility. However, in recent years, the understanding of endocrine physiology underlying testis development and spermatogenesis has enabled the development of new therapeutic strategies. Some concern the management of male infertility. Others are dealing with finding an effective male contraceptive. In this review, we first present the management of infertility, in patients with congenital hypogonadotropic hypogonadism. We then describe the major improvements for Klinefelter patient's infertility. Finally, we review the different hormonal and non-hormonal methods for male contraception, currently in development. Efficacy and safety of the some non-hormonal methods remain to be demonstrated so far in humans.
Collapse
Affiliation(s)
- V Bernard
- Unité INSERM 693, 63 rue Gabriel-Péri, 94276 Le Kremlin-Bicêtre cedex, France.
| | - C Bouvattier
- Service d'endocrinologie et diabétologie pédiatrique, CHU de Bicêtre, Le Kremlin-Bicêtre, France
| | - S Christin-Maitre
- Service d'endocrinologie et maladies de la reproduction, Hôpital Saint-Antoine, Paris, France
| |
Collapse
|
36
|
Müllerian inhibiting substance/anti-Müllerian hormone: A novel treatment for gynecologic tumors. Obstet Gynecol Sci 2014; 57:343-57. [PMID: 25264524 PMCID: PMC4175594 DOI: 10.5468/ogs.2014.57.5.343] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 05/15/2014] [Accepted: 05/15/2014] [Indexed: 01/02/2023] Open
Abstract
Müllerian inhibiting substance (MIS), also called anti-Müllerian hormone (AMH), is a member of the transforming growth factor-β super-family of growth and differentiation response modifiers. It is produced in immature Sertoli cells in male embryos and binds to MIS/AMH receptors in primordial Müllerian ducts to cause regression of female reproductive structures that are the precursors to the fallopian tubes, the surface epithelium of the ovaries, the uterus, the cervix, and the upper third of the vagina. Because most gynecologic tumors originate from Müllerian duct-derived tissues, and since MIS/AMH causes regression of the Müllerian duct in male embryos, it is expected to inhibit the growth of gynecologic tumors. Purified recombinant human MIS/AMH causes growth inhibition of epithelial ovarian cancer cells and cell lines in vitro and in vitro via MIS receptor-mediated mechanism. Furthermore, several lines of evidence suggest that MIS/AMH inhibits proliferation in tissues and cell lines of other MIS/AMH receptor-expressing gynecologic tumors such as cervical, endometrial, breast, and in endometriosis as well. These findings indicate that bioactive MIS/AMH recombinant protein should be tested in patients against tumors expressing the MIS/AMH receptor complex, perhaps beginning with ovarian cancer because it has the worst prognosis. The molecular tools to identify MIS/AMH receptor expressing ovarian and other cancers are in place, thus, it is possible to select patients for treatment. An MIS/AMH ELISA exists to follow administered doses of MIS/AMH, as well. Clinical trials await the production of sufficient supplies of qualified recombinant human MIS/AMH for this purpose.
Collapse
|
37
|
Yuan C, Wu T, Zhang Y, Gao J, Yang Y, Qin F, Liu S, Zheng Y, Wang Z. Responsiveness of four gender-specific genes, figla, foxl2, scp3 and sox9a to 17α-ethinylestradiol in adult rare minnow Gobiocypris rarus. Gen Comp Endocrinol 2014; 200:44-53. [PMID: 24631545 DOI: 10.1016/j.ygcen.2014.02.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 02/17/2014] [Accepted: 02/20/2014] [Indexed: 11/16/2022]
Abstract
Proteins encoded by figla, foxl2, scp3 and sox9a play important roles in gonad differentiation and reproduction. In the present study, we aimed to determine the responsiveness of figla, foxl2, scp3 and sox9a to 17α-ethinylestradiol (EE2) in the gonads of adult Gobiocypris rarus. Full-length cDNAs of figla, scp3 and sox9a were cloned and characterized by RT-PCR and RACE methods. Expression patterns in adult tissues were investigated. Results indicated that figla was predominantly expressed in adult ovaries and scp3 was restrictively expressed in the male testes and sox9a was principally expressed in the brains of both genders and the testes of males. Gene expression profiles of figla, foxl2, scp3 and sox9a were analyzed in the gonads of adult G. rarus exposed to EE2 at 1, 5, 25, and 125ng/L for 3 and 6days. Three-day EE2 treatment at 1-125ng/L all caused a significant increase of figla transcript in testes and foxl2 transcript in ovaries. However, six-day EE2 exposure at 1-125ng/L repressed figla and scp3 transcript in testes and foxl2 transcript in ovaries. The present study indicates that the testicular transcripts of figla and scp3 in males and the ovarian foxl2 transcript in females have high responsiveness to EE2 and they can be used as sensitive molecular biomarkers for early warning to monitor the environmental estrogenic chemicals in fresh water environment. The present study also suggests that the effective EE2 dosage for feminization in male G. rarus might be at least 25ng/L.
Collapse
Affiliation(s)
- Cong Yuan
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Tingting Wu
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Yingying Zhang
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Jiancao Gao
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Yanping Yang
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Fang Qin
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Shaozhen Liu
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Yao Zheng
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China
| | - Zaizhao Wang
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling, Shaanxi 712100, China.
| |
Collapse
|
38
|
Abstract
The ontogeny of the hypothalamic-pituitary-gonadal axis is particularly characterised by incomplete functional maturation in utero and during early postnatal life, followed by functional regression and partial quiescence during childhood, and subsequently by final complete maturation during puberty. This review addresses the distinctive features of testis developmental physiology--especially in the seminiferous tubule compartment--which explain the differences observed in testicular function and its disorders between the early postnatal activation period--which many authors call "mini-puberty"--and canonical puberty.
Collapse
|
39
|
Cutting AD, Ayers K, Davidson N, Oshlack A, Doran T, Sinclair AH, Tizard M, Smith CA. Identification, expression, and regulation of anti-Müllerian hormone type-II receptor in the embryonic chicken gonad. Biol Reprod 2014; 90:106. [PMID: 24621923 DOI: 10.1095/biolreprod.113.116491] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Anti-Müllerian hormone (AMH) signaling is required for proper development of the urogenital system in vertebrates. In male mammals, AMH is responsible for regressing the Müllerian ducts, which otherwise develop into the fallopian tubes, oviducts, and upper vagina of the female reproductive tract. This role is highly conserved across higher vertebrates. However, AMH is required for testis development in fish species that lack Müllerian ducts, implying that AMH signaling has broader roles in other vertebrates. AMH signals through two serine/threonine kinase receptors. The primary AMH receptor, AMH receptor type-II (AMHR2), recruits the type I receptor, which transduces the signal intracellularly. To enhance our understanding of AMH signaling and the potential role of AMH in gonadal sex differentiation, we cloned chicken AMHR2 cDNA and examined its expression profile during gonadal sex differentiation. AMHR2 is expressed in the gonads and Müllerian ducts of both sexes but is more strongly expressed in males after the onset of gonadal sex differentiation. In the testes, the AMHR2 protein colocalizes with AMH, within Sertoli cells of the testis cords. AMHR2 protein expression is up-regulated in female embryos treated with the estrogen synthesis inhibitor fadrozole. Conversely, knockdown of the key testis gene DMRT1 leads to disruption of AMHR2 expression in the developing seminiferous cords of males. These results indicate that AMHR2 is developmentally regulated during testicular differentiation in the chicken embryo. AMH signaling may be important for gonadal differentiation in addition to Müllerian duct regression in birds.
Collapse
Affiliation(s)
- Andrew D Cutting
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia Commonwealth Scientific and Industrial Research Organisation (CSIRO) Food and Health Science, Australian Animal Health Laboratory, Geelong, Victoria, Australia Poultry Cooperative Research Centre, Armidale, New South Wales, Australia
| | - Katie Ayers
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia Poultry Cooperative Research Centre, Armidale, New South Wales, Australia
| | - Nadia Davidson
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Alicia Oshlack
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Tim Doran
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Food and Health Science, Australian Animal Health Laboratory, Geelong, Victoria, Australia Poultry Cooperative Research Centre, Armidale, New South Wales, Australia
| | - Andrew H Sinclair
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia Poultry Cooperative Research Centre, Armidale, New South Wales, Australia
| | - Mark Tizard
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Food and Health Science, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Craig A Smith
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Victoria, Australia Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia Department of Zoology, The University of Melbourne, Melbourne, Victoria, Australia Poultry Cooperative Research Centre, Armidale, New South Wales, Australia
| |
Collapse
|
40
|
Grinspon RP, Loreti N, Braslavsky D, Valeri C, Schteingart H, Ballerini MG, Bedecarrás P, Ambao V, Gottlieb S, Ropelato MG, Bergadá I, Campo SM, Rey RA. Spreading the clinical window for diagnosing fetal-onset hypogonadism in boys. Front Endocrinol (Lausanne) 2014; 5:51. [PMID: 24847309 PMCID: PMC4019849 DOI: 10.3389/fendo.2014.00051] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 03/27/2014] [Indexed: 11/25/2022] Open
Abstract
In early fetal development, the testis secretes - independent of pituitary gonadotropins - androgens and anti-Müllerian hormone (AMH) that are essential for male sex differentiation. In the second half of fetal life, the hypothalamic-pituitary axis gains control of testicular hormone secretion. Follicle-stimulating hormone (FSH) controls Sertoli cell proliferation, responsible for testis volume increase and AMH and inhibin B secretion, whereas luteinizing hormone (LH) regulates Leydig cell androgen and INSL3 secretion, involved in the growth and trophism of male external genitalia and in testis descent. This differential regulation of testicular function between early and late fetal periods underlies the distinct clinical presentations of fetal-onset hypogonadism in the newborn male: primary hypogonadism results in ambiguous or female genitalia when early fetal-onset, whereas it becomes clinically undistinguishable from central hypogonadism when established later in fetal life. The assessment of the hypothalamic-pituitary-gonadal axis in male has classically relied on the measurement of gonadotropin and testosterone levels in serum. These hormone levels normally decline 3-6 months after birth, thus constraining the clinical evaluation window for diagnosing male hypogonadism. The advent of new markers of gonadal function has spread this clinical window beyond the first 6 months of life. In this review, we discuss the advantages and limitations of old and new markers used for the functional assessment of the hypothalamic-pituitary-testicular axis in boys suspected of fetal-onset hypogonadism.
Collapse
Affiliation(s)
- Romina P. Grinspon
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Nazareth Loreti
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Débora Braslavsky
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Clara Valeri
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Helena Schteingart
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - María Gabriela Ballerini
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Patricia Bedecarrás
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Verónica Ambao
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Silvia Gottlieb
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - María Gabriela Ropelato
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Ignacio Bergadá
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Stella M. Campo
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
- *Correspondence: Rodolfo A. Rey, Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET, FEI, División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Gallo 1330, Buenos Aires C1425EFD, Argentina e-mail:
| |
Collapse
|
41
|
Chen SX, Bogerd J, Schoonen NE, Martijn J, de Waal PP, Schulz RW. A progestin (17α,20β-dihydroxy-4-pregnen-3-one) stimulates early stages of spermatogenesis in zebrafish. Gen Comp Endocrinol 2013; 185:1-9. [PMID: 23360837 DOI: 10.1016/j.ygcen.2013.01.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 12/12/2012] [Accepted: 01/12/2013] [Indexed: 10/27/2022]
Abstract
Recently, evidence has been provided for multiple regulatory functions of progestins during the late mitotic and meiotic phases of spermatogenesis in teleost fish. For example, our previous studies suggested that 17α,20β-dihydroxy-4-pregnen-3-one (DHP), potentially via Sertoli cells that express the progesterone receptor (pgr) gene, can contribute to the regulation of zebrafish spermatogenesis. To further our understanding of the function of DHP at early spermatogenetic stages, we investigated in the present study the expression of genes reflecting Sertoli cell function and spermatogenic development in adult zebrafish testis after DHP treatment in tissue culture. Moreover, using an in vivo model of estrogen-mediated down-regulation of androgen production to interrupt adult spermatogenesis, we studied the effects of DHP on estrogen-interrupted spermatogenesis. In this model, DHP treatment doubled the testis weight, and all differentiating germ cell types, such as type B spermatogonia and primary spermatocytes, were abundantly present and incorporated the DNA-synthesis marker (BrdU). Accordingly, transcript levels of germ cell marker genes were up-regulated. Moreover, transcripts of two Sertoli cell-derived genes anti-müllerian hormone (amh) and gonadal soma-derived growth factor (gsdf) were up-regulated, as were three genes of the insulin-like growth factor signaling system, insulin-like growth factor 2b (igf2b), insulin-like growth factor 3 (igf3) and insulin-like growth factor 1b receptor (igf1rb). We further analyzed the relationship between these genes and DHP treatment using a primary zebrafish testis tissue culture system. In the presence of DHP, only igf1rb mRNA levels showed a significant increase among the somatic genes tested, and germ cell marker transcripts were again up-regulated. Taken together, our results show that DHP treatment induced the proliferation of early spermatogonia, their differentiation into late spermatogonia and spermatocytes as well as expression of marker genes for these germ cell stages. DHP-mediated stimulation of spermatogenesis and hence growth of spermatogenic cysts and the associated increase in Sertoli cell number may in part explain the elevated expression of Sertoli cell genes, but our data also suggest an up-regulation of the activity of the Igf signaling system.
Collapse
Affiliation(s)
- Shi X Chen
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen 361005, PR China
| | | | | | | | | | | |
Collapse
|
42
|
Monniaux D, Drouilhet L, Rico C, Estienne A, Jarrier P, Touzé JL, Sapa J, Phocas F, Dupont J, Dalbiès-Tran R, Fabre S. Regulation of anti-Müllerian hormone production in domestic animals. Reprod Fertil Dev 2013; 25:1-16. [DOI: 10.1071/rd12270] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In mammals, anti-Müllerian hormone (AMH) expression is detected in the granulosa cells of all growing follicles and is highest in healthy small antral follicles, which contribute most significantly to AMH endocrine levels. AMH is a reliable endocrine marker of this population of gonadotrophin-responsive follicles in ruminants and, over the longer term, plasma AMH concentrations are characteristic of individual animals. In the cow, plasma AMH concentrations follow specific dynamic profiles throughout the prepubertal period, the oestrous cycle and the change from gestation to the post partum period, with the alterations most likely reflecting numerical changes in the population of high AMH-producing follicles. In granulosa cells, bone morphogenetic proteins (BMP) enhance AMH gene expression and AMH synthesis, with these effects antagonised by FSH. BMP could both support follicular growth and contribute significantly to the induction and/or maintenance of AMH expression in small growing follicles. AMH expression decreases sharply in large follicles when they become oestrogenic, suggesting a role for FSH and/or oestradiol in these changes, but the underlying mechanisms remain hypothetical. A better understanding of the factors and mechanisms regulating AMH production is needed to propose new strategies for managing the reserve of primordial and small growing follicles, as well as for improving embryo production.
Collapse
|
43
|
Josso N, Rey RA, Picard JY. Anti-müllerian hormone: a valuable addition to the toolbox of the pediatric endocrinologist. Int J Endocrinol 2013; 2013:674105. [PMID: 24382961 PMCID: PMC3870610 DOI: 10.1155/2013/674105] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 10/07/2013] [Indexed: 01/18/2023] Open
Abstract
Anti-Müllerian hormone (AMH), secreted by immature Sertoli cells, provokes the regression of male fetal Müllerian ducts. FSH stimulates AMH production; during puberty, AMH is downregulated by intratesticular testosterone and meiotic germ cells. In boys, AMH determination is useful in the clinical setting. Serum AMH, which is low in infants with congenital central hypogonadism, increases with FSH treatment. AMH is also low in patients with primary hypogonadism, for instance in Down syndrome, from early postnatal life and in Klinefelter syndrome from midpuberty. In boys with nonpalpable gonads, AMH determination, without the need for a stimulation test, is useful to distinguish between bilaterally abdominal gonads and anorchism. In patients with disorders of sex development (DSD), serum AMH determination helps as a first line test to orientate the etiologic diagnosis: low AMH is indicative of dysgenetic DSD whereas normal AMH is suggestive of androgen synthesis or action defects. Finally, in patients with persistent Müllerian duct syndrome (PMDS), undetectable serum AMH drives the genetic search to mutations in the AMH gene, whereas normal or high AMH is indicative of an end organ defect due to AMH receptor gene defects.
Collapse
Affiliation(s)
- Nathalie Josso
- INSERM U782, Université Paris-Sud, UMR-S0782, 92140 Clamart, France
- *Nathalie Josso:
| | - Rodolfo A. Rey
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE), CONICET-FEI-División de Endocrinología, Hospital de Niños “R. Gutiérrez”, C1425EFD Buenos Aires, Argentina
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, C1121ABG Buenos Aires, Argentina
| | - Jean-Yves Picard
- INSERM U782, Université Paris-Sud, UMR-S0782, 92140 Clamart, France
| |
Collapse
|
44
|
Pradhan A, Khalaf H, Ochsner SA, Sreenivasan R, Koskinen J, Karlsson M, Karlsson J, McKenna NJ, Orbán L, Olsson PE. Activation of NF-κB protein prevents the transition from juvenile ovary to testis and promotes ovarian development in zebrafish. J Biol Chem 2012; 287:37926-38. [PMID: 22988238 PMCID: PMC3488064 DOI: 10.1074/jbc.m112.386284] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Testis differentiation in zebrafish involves juvenile ovary to testis transformation initiated by an apoptotic wave. The molecular regulation of this transformation process is not fully understood. NF-κB is activated at an early stage of development and has been shown to interact with steroidogenic factor-1 in mammals, leading to the suppression of anti-Müllerian hormone (Amh) gene expression. Because steroidogenic factor-1 and Amh are important for proper testis development, NF-κB-mediated induction of anti-apoptotic genes could, therefore, also play a role in zebrafish gonad differentiation. The aim of this study was to examine the potential role of NF-κB in zebrafish gonad differentiation. Exposure of juvenile zebrafish to heat-killed Escherichia coli activated the NF-κB pathways and resulted in an increased ratio of females from 30 to 85%. Microarray and quantitative real-time-PCR analysis of gonads showed elevated expression of NF-κB-regulated genes. To confirm the involvement of NF-κB-induced anti-apoptotic effects, zebrafish were treated with sodium deoxycholate, a known inducer of NF-κB or NF-κB activation inhibitor (NAI). Sodium deoxycholate treatment mimicked the effect of heat-killed bacteria and resulted in an increased proportion of females from 25 to 45%, whereas the inhibition of NF-κB using NAI resulted in a decrease in females from 45 to 20%. This study provides proof for an essential role of NF-κB in gonadal differentiation of zebrafish and represents an important step toward the complete understanding of the complicated process of sex differentiation in this species and possibly other cyprinid teleosts as well.
Collapse
Affiliation(s)
- Ajay Pradhan
- Department of Biology, Örebro Life Science Center, School of Science and Technology, Örebro University, SE-701 82 Örebro, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Grinspon RP, Rey RA. New perspectives in the diagnosis of pediatric male hypogonadism: the importance of AMH as a Sertoli cell marker. ACTA ACUST UNITED AC 2011; 55:512-9. [DOI: 10.1590/s0004-27302011000800003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 10/16/2011] [Indexed: 12/11/2022]
Abstract
Sertoli cells are the most active cell population in the testis during infancy and childhood. In these periods of life, hypogonadism can only be evidenced without stimulation tests, if Sertoli cell function is assessed. AMH is a useful marker of prepubertal Sertoli cell activity and number. Serum AMH is high from fetal life until mid-puberty. Testicular AMH production increases in response to FSH and is potently inhibited by androgens. Serum AMH is undetectable in anorchidic patients. In primary or central hypogonadism affecting the whole gonad and established in fetal life or childhood, serum AMH is low. Conversely, when hypogonadism affects only Leydig cells (e.g. LHβ mutations, LH/CG receptor or steroidogenic enzyme defects), serum AMH is normal or high. In pubertal males with central hypogonadism, AMH is low for Tanner stage (reflecting lack of FSH stimulus), but high for the age (indicating lack of testosterone inhibitory effect). Treatment with FSH provokes an increase in serum AMH, whereas hCG administration increases testosterone levels, which downregulate AMH. In conclusion, assessment of serum AMH is helpful to evaluate gonadal function, without the need for stimulation tests, and guides etiological diagnosis of pediatric male hypogonadism. Furthermore, serum AMH is an excellent marker of FSH and androgen action on the testis.
Collapse
Affiliation(s)
| | - Rodolfo A. Rey
- Hospital de Niños Ricardo Gutiérrez, Argentina; University of Buenos Aires, Argentina
| |
Collapse
|
46
|
Lasala C, Schteingart HF, Arouche N, Bedecarrás P, Grinspon RP, Picard JY, Josso N, di Clemente N, Rey RA. SOX9 and SF1 are involved in cyclic AMP-mediated upregulation of anti-Mullerian gene expression in the testicular prepubertal Sertoli cell line SMAT1. Am J Physiol Endocrinol Metab 2011; 301:E539-47. [PMID: 21693691 DOI: 10.1152/ajpendo.00187.2011] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In Sertoli cells, anti-Müllerian hormone (AMH) expression is upregulated by FSH via cyclic AMP (cAMP), although no classical cAMP response elements exist in the AMH promoter. The response to cAMP involves NF-κB and AP2; however, targeted mutagenesis of their binding sites in the AMH promoter do not completely abolish the response. In this work we assessed whether SOX9, SF1, GATA4, and AP1 might represent alternative pathways involved in cAMP-mediated AMH upregulation, using real-time RT-PCR (qPCR), targeted mutagenesis, luciferase assays, and immunocytochemistry in the Sertoli cell line SMAT1. We also explored the signaling cascades potentially involved. In qPCR experiments, Amh, Sox9, Sf1, and Gata4 mRNA levels increased after SMAT1 cells were incubated with cAMP. Blocking PKA abolished the effect of cAMP on Sox9, Sf1, and Gata4 expression, inhibiting PI3K/PKB impaired the effect on Sf1 and Gata4, and reducing MEK1/2 and p38 MAPK activities curtailed Gata4 increase. SOX9 and SF1 translocated to the nucleus after incubation with cAMP. Mutations of the SOX9 or SF1 sites, but not of GAT4 or AP1 sites, precluded the response of a 3,063-bp AMH promoter to cAMP. In conclusion, in the Sertoli cell line SMAT1 cAMP upregulates SOX9, SF1, and GATA4 expression and induces SOX9 and SF1 nuclear translocation mainly through PKA, although other kinases may also participate. SOX9 and SF1 binding to the AMH promoter is essential to increase the activity of the AMH promoter in response to cAMP.
Collapse
Affiliation(s)
- Celina Lasala
- Centro de Investigaciones Endocrinológicas, Hospital de Niños R. Gutiérrez, Gallo, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Grinspon RP, Bedecarrás P, Ballerini MG, Iñiguez G, Rocha A, Mantovani Rodrigues Resende EA, Brito VN, Milani C, Figueroa Gacitúa V, Chiesa A, Keselman A, Gottlieb S, Borges MF, Ropelato MG, Picard JY, Codner E, Rey RA. Early onset of primary hypogonadism revealed by serum anti-Müllerian hormone determination during infancy and childhood in trisomy 21. ACTA ACUST UNITED AC 2011; 34:e487-98. [PMID: 21831236 DOI: 10.1111/j.1365-2605.2011.01210.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Male patients with an extra sex chromosome or autosome are expected to present primary hypogonadism at puberty owing to meiotic germ-cell failure. Scarce information is available on trisomy 21, a frequent autosomal aneuploidy. Our objective was to assess whether trisomy 21 presents with pubertal-onset, germ-cell specific, primary hypogonadism in males, or whether the hypogonadism is established earlier and affects other testicular cell populations. We assessed the functional status of the pituitary-testicular axis, especially Sertoli cell function, in 117 boys with trisomy 21 (ages: 2months-20year). To compare with an adequate control population, we established reference levels for serum anti-Müllerian hormone (AMH) in 421 normal males, from birth to adulthood, using a recently developed ultrasensitive assay. In trisomy 21, AMH was lower than normal, indicating Sertoli cell dysfunction, from early infancy, independently of the existence of cryptorchidism. The overall prevalence rate of AMH below the 3rd percentile was 64.3% in infants with trisomy 21. Follicle-stimulating hormone was elevated in patients <6months and after pubertal onset. Testosterone was within the normal range, but luteinizing hormone was elevated in most patients <6months and after pubertal onset, indicating a mild Leydig cell dysfunction. We conclude that in trisomy 21, primary hypogonadism involves a combined dysfunction of Sertoli and Leydig cells, which can be observed independently of cryptorchidism soon after birth, thus prompting the search for new hypotheses to explain the pathophysiology of gonadal dysfunction in autosomal trisomy.
Collapse
Affiliation(s)
- R P Grinspon
- División de Endocrinología, Centro de Investigaciones Endocrinológicas (CEDIE-CONICET), Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hwang SJ, Suh MJ, Yoon JH, Kim MR, Ryu KS, Nam SW, Donahoe PK, Maclaughlin DT, Kim JH. Identification of characteristic molecular signature of Müllerian inhibiting substance in human HPV-related cervical cancer cells. Int J Oncol 2011; 39:811-20. [PMID: 21573503 DOI: 10.3892/ijo.2011.1042] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 04/11/2011] [Indexed: 12/30/2022] Open
Abstract
Müllerian inhibiting substance (MIS), also known as anti-Müllerian hormone (AMH), is a member of the transforming growth factor-β (TGF-β) superfamily that plays an important role in the mesenchymal-epithelial interaction, cell growth and proliferation, extracellular matrix production and tissue remodeling. Previously, we demonstrated that MIS suppressed ovarian cancer cell growth and suggested large-scale genetic elements that could be responsible for anti-neoplastic effects of MIS on ovarian cancer cells. In this study, we demonstrated the expression of MIS type II receptor (MISRII) in the human papillomavirus (HPV)-16-related cervical cancer cell lines CaSki and SiHa, and a non-HPV-related cervical cancer cell line, C33A. We also showed that MIS inhibited growth of cervical cancer cells, and induced cellular apoptosis of C33A. In addition, we identified a characteristic molecular signature of MIS in CaSki cells by using whole genome expression analysis. Of the 1,690 genes that showed significant expression changes by MIS, 21 genes were related to cell cycle; 13 genes to apoptosis; and 52 genes to the cancer pathway. On performing a search for cell cycle pathways in the KEGG pathway database, several gene expressions at the G1/S checkpoint were found. In particular, the expression of p16 and p107 increased and that of E2F2 and E2F3 decreased at an early stage, whereas the expression of E2F4 and E2F5 decreased at a later stage after MIS treatment. These data suggest that MIS produces activity against HPV16-related cervical cancers in vitro, and MIS may also be an effective targeted therapy for HPV16-related cervical cancer. Genetic data obtained here could be useful in determining the treatment strategy of MISR-expressing cervical tumors in the future.
Collapse
Affiliation(s)
- Seong Jin Hwang
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Rico C, Médigue C, Fabre S, Jarrier P, Bontoux M, Clément F, Monniaux D. Regulation of Anti-Müllerian Hormone Production in the Cow: A Multiscale Study at Endocrine, Ovarian, Follicular, and Granulosa Cell Levels1. Biol Reprod 2011; 84:560-71. [DOI: 10.1095/biolreprod.110.088187] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
50
|
Urushitani H, Katsu Y, Miyagawa S, Kohno S, Ohta Y, Guillette LJ, Iguchi T. Molecular cloning of anti-Müllerian hormone from the American alligator, Alligator mississippiensis. Mol Cell Endocrinol 2011; 333:190-9. [PMID: 21187121 DOI: 10.1016/j.mce.2010.12.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 12/12/2010] [Accepted: 12/21/2010] [Indexed: 11/26/2022]
Abstract
Anti-Müllerian hormone (AMH) plays an important role in male sex differentiation in vertebrates. AMH produced by Sertoli cells of the fetal testis induces regression of the Müllerian duct in mammalian species. In alligators, sexual differentiation is controlled by the temperature during egg incubation, termed temperature-dependent sex determination (TSD). The TSD mechanism inducing sex differentiation is thought to be unique and different from that of genetic sex determination as no gene such as the SRY of mammals has been identified. However, many of the genes associated with gonadal differentiation in mammals also are expressed in the developing gonads of species exhibiting TSD. To clarify the molecular mechanisms associated with gonad formation during the temperature-sensitive period (TSP), we have cloned the full length AMH gene in the alligator, and quantitatively compared mRNA expression patterns in the gonad-adrenal-mesonephros (GAM) complex isolated from alligator embryos incubated at male and female producing temperatures. The deduced amino acid sequence of the alligator AMH cDNA showed high identity (59-53%) to avian AMH genes. AMH mRNA expression was high in the GAM of male alligator embryos at stage 24 (immediately after sex determination) and hatchlings, but suppressed in the GAM of estrogen-exposed hatchlings incubated at the male-producing temperature. In the alligator AMH proximal promoter, a number of transcriptional factors (for SF-1. GATA, WT-1 and SOX9) binding elements were also identified and they exhibit a conserved pattern seen in other species. SOX9 up-regulates transcriptional activity through the amAMH promoter region. These results suggested that AMH and SOX9 play important roles in TSD of the American alligator.
Collapse
Affiliation(s)
- Hiroshi Urushitani
- Okazaki Institute for Integrative Bioscience, National Institute for Basic Biology, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | | | | | | | | | | | | |
Collapse
|