1
|
Radomsky T, Anderson RC, Millar RP, Newton CL. Restoring function to inactivating G protein-coupled receptor variants in the hypothalamic-pituitary-gonadal axis 1. J Neuroendocrinol 2024; 36:e13418. [PMID: 38852954 DOI: 10.1111/jne.13418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 03/30/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024]
Abstract
G protein-coupled receptors (GPCRs) are central to the functioning of the hypothalamic-pituitary-gonadal axis (HPG axis) and include the rhodopsin-like GPCR family members, neurokinin 3 receptor, kappa-opioid receptor, kisspeptin 1 receptor, gonadotropin-releasing hormone receptor, and the gonadotropin receptors, luteinizing hormone/choriogonadotropin receptor and follicle-stimulating hormone receptor. Unsurprisingly, inactivating variants of these receptors have been implicated in a spectrum of reproductive phenotypes, including failure to undergo puberty, and infertility. Clinical induction of puberty in patients harbouring such variants is possible, but restoration of fertility is not always a realisable outcome, particularly for those patients suffering from primary hypogonadism. Thus, novel pharmaceuticals and/or a fundamental change in approach to treating these patients are required. The increasing wealth of data describing the effects of coding-region genetic variants on GPCR function has highlighted that the majority appear to be dysfunctional as a result of misfolding of the encoded receptor protein, which, in turn, results in impaired receptor trafficking through the secretory pathway to the cell surface. As such, these intracellularly retained receptors may be amenable to 'rescue' using a pharmacological chaperone (PC)-based approach. PCs are small, cell permeant molecules hypothesised to interact with misfolded intracellularly retained proteins, stabilising their folding and promoting their trafficking through the secretory pathway. In support of the use of this approach as a viable therapeutic option, it has been observed that many rescued variant GPCRs retain at least a degree of functionality when 'rescued' to the cell surface. In this review, we examine the GPCR PC research landscape, focussing on the rescue of inactivating variant GPCRs with important roles in the HPG axis, and describe what is known regarding the mechanisms by which PCs restore trafficking and function. We also discuss some of the merits and obstacles associated with taking this approach forward into a clinical setting.
Collapse
Affiliation(s)
- Tarryn Radomsky
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ross C Anderson
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Robert P Millar
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Claire L Newton
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
2
|
Bhattacharya I, Dey S, Banerjee A. Revisiting the gonadotropic regulation of mammalian spermatogenesis: evolving lessons during the past decade. Front Endocrinol (Lausanne) 2023; 14:1110572. [PMID: 37124741 PMCID: PMC10140312 DOI: 10.3389/fendo.2023.1110572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Spermatogenesis is a multi-step process of male germ cell (Gc) division and differentiation which occurs in the seminiferous tubules of the testes under the regulation of gonadotropins - Follicle Stimulating Hormone (FSH) and Luteinising hormone (LH). It is a highly coordinated event regulated by the surrounding somatic testicular cells such as the Sertoli cells (Sc), Leydig cells (Lc), and Peritubular myoid cells (PTc). FSH targets Sc and supports the expansion and differentiation of pre-meiotic Gc, whereas, LH operates via Lc to produce Testosterone (T), the testicular androgen. T acts on all somatic cells e.g.- Lc, PTc and Sc, and promotes the blood-testis barrier (BTB) formation, completion of Gc meiosis, and spermiation. Studies with hypophysectomised or chemically ablated animal models and hypogonadal (hpg) mice supplemented with gonadotropins to genetically manipulated mouse models have revealed the selective and synergistic role(s) of hormones in regulating male fertility. We here have briefly summarized the present concept of hormonal control of spermatogenesis in rodents and primates. We also have highlighted some of the key critical questions yet to be answered in the field of male reproductive health which might have potential implications for infertility and contraceptive research in the future.
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology, School of Biological Science, Central University of Kerala, Kasaragod, Kerala, India
- *Correspondence: Arnab Banerjee, ; Indrashis Bhattacharya,
| | - Souvik Dey
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Arnab Banerjee
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS) Pilani, Goa, India
- *Correspondence: Arnab Banerjee, ; Indrashis Bhattacharya,
| |
Collapse
|
3
|
Rivero-Müller A, Huhtaniemi I. Genetic variants of gonadotrophins and their receptors: Impact on the diagnosis and management of the infertile patient. Best Pract Res Clin Endocrinol Metab 2022; 36:101596. [PMID: 34802912 DOI: 10.1016/j.beem.2021.101596] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This narrative review is concerned with genetic variants of the genes encoding gonadotrophin subunits and their receptors, as well as their implications into the diagnosis and treatment of infertility. We first review briefly the basics of molecular biology and biochemistry of gonadotrophin and gonadotrophin receptor structure and function, then describe the phenotypic effects of polymorphisms and mutations of these genes, followed by diagnostic aspects. We will then summarise the information that inactivating gonadotrophin receptor mutations have provided about the controversial topic of extragonadal gonadotrophin action. Finally, we will close with the current and future therapeutic approaches on patients with gonadotrophin and their receptor mutations.
Collapse
Affiliation(s)
- Adolfo Rivero-Müller
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, 20-093, Poland
| | - Ilpo Huhtaniemi
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK.
| |
Collapse
|
4
|
Abstract
The diagnostic suspicion of congenital central hypogonadism is based on clinical signs. Biochemical confirmation is challenging, especially after the postnatal activation stage of the hypothalamic-pituitary-testicular axis. Sertoli cell markers, like AMH and inhibin B, have become useful tools for the diagnosis of male central hypogonadism during childhood. Different mechanisms can participate in the aetiopathogenesis of central hypogonadism, leading to a deficiency in the production of gonadotrophins. Advances in genetic studies, mainly next generation sequencing techniques, have allowed the discovery of a large number of genes related to central hypogonadism. However, a causal variant is found in approximately half of the patients. Central hypogonadism has been classically described as a pathology with variable expressivity and incomplete penetrance. Currently, these characteristics are known to be partially explained by the presence of oligogenicity, that is the participation of variants in more than one gene in the aetiology of central hypogonadism in the same patient.
Collapse
Affiliation(s)
- Romina P Grinspon
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), CONICET - FEI - División de, Endocrinología, Hospital de Niños Ricardo Gutiérrez, C1425EFD, Buenos Aires, Argentina.
| |
Collapse
|
5
|
Bhartiya D, Patel H. An overview of FSH-FSHR biology and explaining the existing conundrums. J Ovarian Res 2021; 14:144. [PMID: 34717708 PMCID: PMC8557046 DOI: 10.1186/s13048-021-00880-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 09/12/2021] [Indexed: 12/23/2022] Open
Abstract
FSH was first identified in 1930 and is central to mammalian reproduction. It is indeed intriguing that despite being researched upon for about 90 years, there is still so much more to learn about FSH-FSHR biology. The purpose of this review is to provide an overview of current understanding of FSH-FSHR biology, to review published data on biological and clinical relevance of reported mutations, polymorphisms and alternately spliced isoforms of FSHR. Tissue-resident stem/progenitor cells in multiple adult tissues including ovaries, testes and uterus express FSHR and this observation results in a paradigm shift in the field. The results suggest a direct action of FSH on the stem cells in addition to their well-studied action on Granulosa and Sertoli cells in the ovaries and testes respectively. Present review further addresses various concerns raised in recent times by the scientific community regarding extragonadal expression of FSHR, especially in cancers affecting multiple organs. Similar population of primitive and pluripotent tissue-resident stem cells expressing FSHR exist in multiple adult tissues including bone marrow and reproductive tissues and help maintain homeostasis throughout life. Any dysfunction of these stem cells results in various pathologies and they also most likely get transformed into cancer stem cells and initiate cancer. This explains why multiple solid as well as liquid tumors express OCT-4 and FSHR. More research efforts need to be focused on alternately spliced FSHR isoforms.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Maharashtra, 400012, Mumbai, India. .,Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| | - Hiren Patel
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Maharashtra, 400012, Mumbai, India.,Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
6
|
Bohaczuk SC, Cassin J, Slaiwa TI, Thackray VG, Mellon PL. Distal Enhancer Potentiates Activin- and GnRH-Induced Transcription of FSHB. Endocrinology 2021; 162:6213400. [PMID: 33824966 PMCID: PMC8157479 DOI: 10.1210/endocr/bqab069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Indexed: 11/19/2022]
Abstract
FSH is critical for fertility. Transcription of FSHB, the gene encoding the beta subunit, is rate-limiting in FSH production and is regulated by both GnRH and activin. Activin signals through SMAD transcription factors. Although the mechanisms and importance of activin signaling in mouse Fshb transcription are well-established, activin regulation of human FSHB is less well understood. We previously reported a novel enhancer of FSHB that contains a fertility-associated single nucleotide polymorphism (rs10031006) and requires a region resembling a full (8 base-pair) SMAD binding element (SBE). Here, we investigated the role of the putative SBE within the enhancer in activin and GnRH regulation of FSHB. In mouse gonadotrope-derived LβT2 cells, the upstream enhancer potentiated activin induction of both the human and mouse FSHB proximal promoters and conferred activin responsiveness to a minimal promoter. Activin induction of the enhancer required the SBE and was blocked by the inhibitory SMAD7, confirming involvement of the classical SMAD signaling pathway. GnRH induction of FSHB was also potentiated by the enhancer and dependent on the SBE, consistent with known activin/GnRH synergy regulating FSHB transcription. In DNA pull-down, the enhancer SBE bound SMAD4, and chromatin immunoprecipitation demonstrated SMAD4 enrichment at the enhancer in native chromatin. Combined activin/GnRH treatment elevated levels of the active transcriptional histone marker, histone 3 lysine 27 acetylation, at the enhancer. Overall, this study indicates that the enhancer is directly targeted by activin signaling and identifies a novel, evolutionarily conserved mechanism by which activin and GnRH can regulate FSHB transcription.
Collapse
Affiliation(s)
- Stephanie C Bohaczuk
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California San Diego, La Jolla, California 92093, USA
| | - Jessica Cassin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California San Diego, La Jolla, California 92093, USA
| | - Theresa I Slaiwa
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California San Diego, La Jolla, California 92093, USA
| | - Varykina G Thackray
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California San Diego, La Jolla, California 92093, USA
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California San Diego, La Jolla, California 92093, USA
- Correspondence: Pamela L. Mellon, Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA. E-mail:
| |
Collapse
|
7
|
Bohaczuk SC, Thackray VG, Shen J, Skowronska-Krawczyk D, Mellon PL. FSHB Transcription is Regulated by a Novel 5' Distal Enhancer With a Fertility-Associated Single Nucleotide Polymorphism. Endocrinology 2021; 162:bqaa181. [PMID: 33009549 PMCID: PMC7846141 DOI: 10.1210/endocr/bqaa181] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Indexed: 12/17/2022]
Abstract
The pituitary gonadotropins, follicle-stimulating hormone (FSH) and luteinizing hormone, signal the gonads to regulate male and female fertility. FSH is critical for female fertility as it regulates oocyte maturation, ovulation, and hormone synthesis. Multiple genome-wide association studies (GWAS) link a 130 Kb locus at 11p14.1, which encompasses the FSH beta-subunit (FSHB) gene, with fertility-related traits that include polycystic ovary syndrome, age of natural menopause, and dizygotic twinning. The most statistically significant single nucleotide polymorphism from several GWAS studies (rs11031006) resides within a highly conserved 450 bp region 26 Kb upstream of the human FSHB gene. Given that sequence conservation suggests an important biological function, we hypothesized that the region could regulate FSHB transcription. In luciferase assays, the conserved region enhanced FSHB transcription and gel shifts identified a binding site for Steroidogenic factor 1 (SF1) contributing to its function. Analysis of mouse pituitary single-cell ATAC-seq demonstrated open chromatin at the conserved region exclusive to a gonadotrope cell-type cluster. Additionally, enhancer-associated histone markers were identified by immunoprecipitation of chromatin from mouse whole pituitary and an immortalized mouse gonadotrope-derived LβT2 cell line at the conserved region. Furthermore, we found that the rs11031006 minor allele upregulated FSHB transcription via increased SF1 binding to the enhancer. All together, these results identify a novel upstream regulator of FSHB transcription and indicate that rs11031006 can modulate FSH levels.
Collapse
Affiliation(s)
- Stephanie C Bohaczuk
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California San Diego, La Jolla, California
| | - Varykina G Thackray
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California San Diego, La Jolla, California
| | - Jia Shen
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, California
| | - Dorota Skowronska-Krawczyk
- Shiley Eye Institute, Viterbi Family Department of Ophthalmology, School of Medicine, University of California, San Diego, California
- Department of Physiology and Biophysics, Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, University of California Irvine, Irvine, California
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
8
|
Zhang J, Tang SY, Zhu XB, Li P, Lu JQ, Cong JS, Wang LB, Zhang F, Li Z. Whole exome sequencing and trio analysis to broaden the variant spectrum of genes in idiopathic hypogonadotropic hypogonadism. Asian J Androl 2021; 23:288-293. [PMID: 33208564 PMCID: PMC8152424 DOI: 10.4103/aja.aja_65_20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Dozens of genes are associated with idiopathic hypogonadotropic hypogonadism (IHH) and an oligogenic etiology has been suggested. However, the associated genes may account for only approximately 50% cases. In addition, a genomic systematic pedigree analysis is still lacking. Here, we conducted whole exome sequencing (WES) on 18 unrelated men affected by IHH and their corresponding parents. Notably, one reported and 10 novel variants in eight known IHH causative genes (AXL, CCDC141, CHD7, DMXL2, FGFR1, PNPLA6, POLR3A, and PROKR2), nine variants in nine recently reported candidate genes (DCAF17, DCC, EGF, IGSF10, NOTCH1, PDE3A, RELN, SLIT2, and TRAPPC9), and four variants in four novel candidate genes for IHH (CCDC88C, CDON, GADL1, and SPRED3) were identified in 77.8% (14/18) of IHH cases. Among them, eight (8/18, 44.4%) cases carried more than one variant in IHH-related genes, supporting the oligogenic model. Interestingly, we found that those variants tended to be maternally inherited (maternal with n = 17 vs paternal with n = 7; P = 0.028). Our further retrospective investigation of published reports replicated the maternal bias (maternal with n = 46 vs paternal with n = 28; P = 0.024). Our study extended a variant spectrum for IHH and provided thefirst evidence that women are probably more tolerant to variants of IHH-related genes than men.
Collapse
Affiliation(s)
- Jian Zhang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai 200011, China
| | - Shu-Yan Tang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai 200011, China
| | - Xiao-Bin Zhu
- Department of Andrology, Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Peng Li
- Department of Andrology, Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Jian-Qi Lu
- Department of Research Institute, Reproduction Medical Center, The first Hospital of Lanzhou University, Lanzhou 730000, China
| | - Jiang-Shan Cong
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai 200011, China
| | - Ling-Bo Wang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai 200011, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), School of Life Sciences, Fudan University, Shanghai 200011, China
| | - Zheng Li
- Department of Andrology, Center for Men's Health, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
| |
Collapse
|
9
|
Shah W, Khan R, Shah B, Khan A, Dil S, Liu W, Wen J, Jiang X. The Molecular Mechanism of Sex Hormones on Sertoli Cell Development and Proliferation. Front Endocrinol (Lausanne) 2021; 12:648141. [PMID: 34367061 PMCID: PMC8344352 DOI: 10.3389/fendo.2021.648141] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/17/2021] [Indexed: 12/30/2022] Open
Abstract
Sustaining and maintaining the intricate process of spermatogenesis is liable upon hormones and growth factors acting through endocrine and paracrine pathways. The Sertoli cells (SCs) are the major somatic cells present in the seminiferous tubules and are considered to be the main regulators of spermatogenesis. As each Sertoli cell supports a specific number of germ cells, thus, the final number of Sertoli cells determines the sperm production capacity. Similarly, sex hormones are also major regulators of spermatogenesis and they can determine the proliferation of Sertoli cells. In the present review, we have critically and comprehensively discussed the role of sex hormones and some other factors that are involved in Sertoli cell proliferation, differentiation and maturation. Furthermore, we have also presented a model of Sertoli cell development based upon the recent advancement in the field of reproduction. Hence, our review article provides a general overview regarding the sex hormonal pathways governing Sertoli cell proliferation and development.
Collapse
Affiliation(s)
| | - Ranjha Khan
- *Correspondence: Xiaohua Jiang, ; Ranjha Khan, ; Jie Wen,
| | | | | | | | | | - Jie Wen
- *Correspondence: Xiaohua Jiang, ; Ranjha Khan, ; Jie Wen,
| | - Xiaohua Jiang
- *Correspondence: Xiaohua Jiang, ; Ranjha Khan, ; Jie Wen,
| |
Collapse
|
10
|
Bang AK, Almstrup K, Nordkap L, Priskorn L, Petersen JH, Blomberg Jensen M, Krause M, Holmboe SA, Egeberg Palme DL, Winge SB, Joensen UN, Olesen IA, Hvidman HW, Juul A, Rajpert-De Meyts E, Jørgensen N. FSHB and FSHR gene variants exert mild modulatory effect on reproductive hormone levels and testis size but not on semen quality: A study of 2020 men from the general Danish population. Andrology 2020; 9:618-631. [PMID: 33236519 DOI: 10.1111/andr.12949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 11/01/2020] [Accepted: 11/20/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Spermatogenesis depends on stimulation by follicle-stimulating hormone (FSH) which binds to FSH receptors (FSHR) on testicular Sertoli cells. Three FSH-related single-nucleotide polymorphisms (SNPs), FSHB -211G>T (rs10835638), FSHR -29G>A (rs1394205) and FSHR 2039A>G (rs6166) affect FSH action, and have been suggested to affect testicular function, but the evidence is uncertain. OBJECTIVE To describe the associations between the three SNPs and testicular function in a large and well-characterised cohort of men from the general population. MATERIALS AND METHODS A cross-sectional study of 2020 Danish men unselected regarding testicular function. Outcome variables were semen parameters, reproductive hormones and testis size. Genotyping was done by competitive allele-specific quantitative PCR. Differences in genotype frequencies were tested by chi-square test and associations between genotypes and outcomes were assessed by multivariate linear regressions. RESULTS The SNPs affected serum FSH; carriers of the variant affecting FSH secretion (FSHB -211G>T) had lower FSH levels while carriers of variants affecting receptor expression (FSHR -29G>A) and receptor sensitivity (FSHR 2039A>G) had higher FSH levels. Carriers of FSHB -211G>T had lower calculated free testosterone/LH ratio. Although both FSHB -211G>T and FSHR 2039A>G were associated with smaller testis size, no clear association was detected in relation to any semen parameters, except a lower total number of morphologically normal spermatozoa in the heterozygous carriers of the FSHB -211G>T DISCUSSION AND CONCLUSION: The studied polymorphisms have only minor modulating influence on testis size and function in healthy men. We detected subtle effects of the three SNPs on FSH levels, but also effects of FSHB -211G>T on calculated free testosterone/LH ratio, compatible with altered Leydig cell function. Thus, the role of these FSH-related polymorphisms is complex and modest in men with normal testicular function, but the possible importance of FSH polymorphisms in men with impaired testicular function should be evaluated in future studies in more detail.
Collapse
Affiliation(s)
- Anne Kirstine Bang
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Almstrup
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Loa Nordkap
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Laerke Priskorn
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen Holm Petersen
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Martin Blomberg Jensen
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Marianna Krause
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Stine Agergaard Holmboe
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Dorte Louise Egeberg Palme
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sofia Boeg Winge
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ulla Nordström Joensen
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Urology, Rigshospitalet, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Inge Ahlmann Olesen
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | - Anders Juul
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Niels Jørgensen
- Department of Growth and Reproduction and International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Gohil A, Eugster EA. Delayed and Precocious Puberty: Genetic Underpinnings and Treatments. Endocrinol Metab Clin North Am 2020; 49:741-757. [PMID: 33153677 PMCID: PMC7705597 DOI: 10.1016/j.ecl.2020.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Delayed puberty may signify a common variation of normal development, or indicate the presence of a pathologic process. Constitutional delay of growth and puberty is a strongly familial type of developmental pattern and accounts for the vast majority of children who are "late bloomers." Individuals with sex chromosomal abnormalities frequently have hypergonadotropic hypogonadism. There are currently 4 known monogenic causes of central precocious puberty. The primary treatment goal in children with hypogonadism is to mimic normal pubertal progression, while the primary aims for the management of precocious puberty are preservation of height potential and prevention of further pubertal development.
Collapse
Affiliation(s)
- Anisha Gohil
- Division of Pediatric Endocrinology, Department of Pediatrics, Riley Hospital for Children at IU Health, Indiana University School of Medicine, 705 Riley Hospital Drive, Room 5960, Indianapolis, IN 46202, USA.
| | - Erica A Eugster
- Division of Pediatric Endocrinology, Department of Pediatrics, Riley Hospital for Children at IU Health, Indiana University School of Medicine, 705 Riley Hospital Drive, Room 5960, Indianapolis, IN 46202, USA
| |
Collapse
|
12
|
Ajayi AF, Akhigbe RE. The physiology of male reproduction: Impact of drugs and their abuse on male fertility. Andrologia 2020; 52:e13672. [DOI: 10.1111/and.13672] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 04/28/2020] [Accepted: 05/04/2020] [Indexed: 12/16/2022] Open
Affiliation(s)
- Ayodeji Folorunsho Ajayi
- Department of Physiology College of Medicine Ladoke Akintola University of Technology Ogbomoso Nigeria
| | - Roland Eghoghosoa Akhigbe
- Department of Physiology College of Medicine Ladoke Akintola University of Technology Ogbomoso Nigeria
- Reproductive Biology and Toxicology Research Laboratories Oasis of Grace Hospital Osogbo Nigeria
| |
Collapse
|
13
|
Bhattacharya I, Sen Sharma S, Majumdar SS. Pubertal orchestration of hormones and testis in primates. Mol Reprod Dev 2019; 86:1505-1530. [DOI: 10.1002/mrd.23246] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology & BiotechnologyHNB Garhwal University, Srinagar CampusSrinagar India
- Cellular Endocrinology LabNational Institute of ImmunologyNew Delhi India
| | - Souvik Sen Sharma
- Cellular Endocrinology LabNational Institute of ImmunologyNew Delhi India
| | - Subeer S. Majumdar
- Cellular Endocrinology LabNational Institute of ImmunologyNew Delhi India
- Gene and Protein Engineering LabNational Institute of Animal BiotechnologyHyderabad India
| |
Collapse
|
14
|
Hsueh AJ, He J. Gonadotropins and their receptors: coevolution, genetic variants, receptor imaging, and functional antagonists. Biol Reprod 2019; 99:3-12. [PMID: 29462242 DOI: 10.1093/biolre/ioy012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/06/2018] [Indexed: 12/29/2022] Open
Abstract
Gonadotropins belong to the family of dimeric glycoprotein hormones and regulate gonadal physiology mediated by G protein-coupled, seven-transmembrane receptors. These glycoprotein hormones are widely used in the clinic to promote ovarian follicle development and for treating some cases of male infertility. We traced the coevolution of dimeric gonadotropin hormones and their receptors, together with thyrotropin and its receptor. We updated recent findings on human genetic variants of these genes and their association with dizygotic twining, polycystic ovarian syndrome, primary ovarian insufficiency, male-limited precocious puberty, and infertility. In addition to the known physiological roles of gonadotropin-receptor signaling in gonadal tissues, we also discussed emerging understanding of extragonadal functions of gonadotropins in bones and adipose tissues, together with recent advances in in vivo imaging of gonadotropin receptors in live animals. Recent development of gonadotropin receptor agonists and antagonists were summarized with an emphasis on the development of functional antagonists for FSH receptors to alleviate osteoporosis and obesity associated with menopause.
Collapse
Affiliation(s)
- Aaron J Hsueh
- Program of Reproductive and Stem Cell Biology, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California, USA
| | - Jiahuan He
- Program of Reproductive and Stem Cell Biology, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
15
|
Rougier C, Hieronimus S, Panaïa-Ferrari P, Lahlou N, Paris F, Fenichel P. Isolated follicle-stimulating hormone (FSH) deficiency in two infertile men without FSH β gene mutation: Case report and literature review. ANNALES D'ENDOCRINOLOGIE 2019; 80:234-239. [PMID: 31439307 DOI: 10.1016/j.ando.2019.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/01/2019] [Accepted: 06/22/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Congenital FSH deficiency is an exceptional cause of male infertility most often attributed to FSH β gene mutations. The few published cases report azoospermia, severe testicular hypotrophy and normal testosterone levels associated with normal virilization. We report the exploration of two young men aged 26 and 27 years with severe sperm abnormalities, moderate testicular hypotrophy and isolated FSH deficiency. METHODS Several FSH, LH, total testosterone and inhibin B assays and FSH β gene sequencing were performed. RESULTS FSH was almost undetectable at baseline and poorly responsive to GnRH test, whereas LH was normal at baseline and increased after GnRH test. Testosterone levels were within the adult range, while inhibin B levels were upper-normal to high. No FSH β gene mutations were found. Exogenous FSH treatment was followed by spontaneous pregnancy in one case and required intra-cytoplasmic sperm injection (ICSI) in the other. CONCLUSIONS The paradoxical high levels of inhibin B reflect the presence of functional Sertoli cells and may explain the isolated FSH deficiency. An intra-gonadal factor stimulating inhibin B secretion is discussed.
Collapse
Affiliation(s)
- Charlotte Rougier
- Department of Endocrinology and Reproductive Medicine, University Hospital of Nice, 151, route de Saint-Antoine, 06200 Nice, France.
| | - Sylvie Hieronimus
- Department of Endocrinology and Reproductive Medicine, University Hospital of Nice, 151, route de Saint-Antoine, 06200 Nice, France
| | - Patricia Panaïa-Ferrari
- Department of Biochemistry and Hormonology, University Hospital of Nice, 151, route de Saint-Antoine, 06200 Nice, France
| | - Najiba Lahlou
- Department of Biological Endocrinology, CHU Cochin, AP-HP, 75014 Paris, France
| | - Françoise Paris
- Department of Hormonology and Pediatric Endocrinology, University Hospital of Montpellier, 34295 Montpellier, France
| | - Patrick Fenichel
- Department of Endocrinology and Reproductive Medicine, University Hospital of Nice, 151, route de Saint-Antoine, 06200 Nice, France
| |
Collapse
|
16
|
Misgar RA, Wani AI, Bankura B, Bashir MI, Roy A, Das M. FSH β-subunit mutations in two sisters: the first report from the Indian sub-continent and review of previous cases. Gynecol Endocrinol 2019; 35:290-293. [PMID: 30602350 DOI: 10.1080/09513590.2018.1529159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Isolated FSH deficiency due to mutations in the gene for β-subunit of FSH is an extremely rare autosomal recessive disease of which only eleven cases have been reported so far. The clinical features include absent breast development and primary amenorrhea in females and azoospermia with normal testosterone levels in males. In this study we report two Kashmiri sisters born to native Kashmiri consanguineous parents with failure of onset of puberty. Hormonal evaluation revealed undetectable serum FSH and estradiol and high LH. Genetic analysis of FSH β-gene identified one nonsense mutation (c.343C > T:p. Arg115Stop) in exon 3. The two sisters were homozygous for this nonsense mutation while the parents were heterozygous. Incorporation of a stop codon at 115 codon position is predicted to result in the formation of truncated FSH β protein, lacking 14 amino acid from the carboxy-terminus (p.Arg115Stop). Very recently, this same mutation was reported for the first time in a Chinese male. Ours is the first ever report of any FSH β-subunit mutation from the Indian sub-continent and this particular mutation in any female from anywhere in the world. We conclude and emphasize that this diagnosis should be considered in girls with delayed puberty and selective deficiency of FSH.
Collapse
Affiliation(s)
- Raiz Ahmad Misgar
- a Department of Endocrinology , Sher-i-Kashmir Institute of Medical Sciences , Srinagar , India
| | - Arshad Iqbal Wani
- a Department of Endocrinology , Sher-i-Kashmir Institute of Medical Sciences , Srinagar , India
| | | | - Mir Iftikhar Bashir
- a Department of Endocrinology , Sher-i-Kashmir Institute of Medical Sciences , Srinagar , India
| | | | - Madhusudan Das
- b Department of Zoology , University of Calcutta , Kolkata , India
| |
Collapse
|
17
|
Bhattacharya I, Basu S, Pradhan BS, Sarkar H, Nagarajan P, Majumdar SS. Testosterone augments FSH signaling by upregulating the expression and activity of FSH-Receptor in Pubertal Primate Sertoli cells. Mol Cell Endocrinol 2019; 482:70-80. [PMID: 30579957 DOI: 10.1016/j.mce.2018.12.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 12/16/2018] [Accepted: 12/18/2018] [Indexed: 11/30/2022]
Abstract
The synergistic actions of Testosterone (T) and FSH via testicular Sertoli cells (Sc) regulate male fertility. We have previously reported that the actions of these hormones (T and FSH) in infant monkey testes are restricted only to the expansion of Sc and spermatogonial cells. The robust differentiation of male Germ cells (Gc) occurs after pubertal maturation of testis. The present study was aimed to investigate the molecular basis of the synergy between T and FSH action in pubertal primate (Macaca mulatta) Sc. Using primary Sc culture, we here have demonstrated that T (but not FSH) downregulated AMH and Inhibin-β-B (INHBB) mRNAs in pubertal Sc. We also found that, prolonged stimulation of T in pubertal Sc significantly elevated the expression of genes involved in FSH signaling pathway like FSH-Receptor (FSHR), GNAS and RIC8B, and this was associated with a rise in cAMP production. T also augmented FSH induced expression of genes like SCF, GDNF, ABP and Transferrin (TF) in pubertal Sc. We therefore conclude that T acts in synergy with FSH signaling in pubertal Sc. Such a coordinated network of hormonal signaling in Sc may facilitate the timely onset of the first spermatogenic wave in pubertal primates and is responsible for quantitatively and qualitatively normal spermatogenesis.
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, 110067, India; Primate Research Center, National Institute of Immunology, New Delhi, India; Department of Zoology and Biotechnology, HNB Garhwal University, Srinagar Campus, Uttarakhand, India
| | - Sayon Basu
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Bhola Shankar Pradhan
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Hironmoy Sarkar
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, 110067, India; Department of Microbiology, Raiganj University, Raiganj, West Bengal, India
| | - Perumal Nagarajan
- Primate Research Center, National Institute of Immunology, New Delhi, India
| | - Subeer S Majumdar
- Cellular Endocrinology Laboratory, National Institute of Immunology, New Delhi, 110067, India; Primate Research Center, National Institute of Immunology, New Delhi, India; National Institute of Animal Biotechnology, Hyderabad, Telangana, India.
| |
Collapse
|
18
|
Conforti A, Vaiarelli A, Cimadomo D, Bagnulo F, Peluso S, Carbone L, Di Rella F, De Placido G, Ubaldi FM, Huhtaniemi I, Alviggi C. Pharmacogenetics of FSH Action in the Female. Front Endocrinol (Lausanne) 2019; 10:398. [PMID: 31293516 PMCID: PMC6606727 DOI: 10.3389/fendo.2019.00398] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/05/2019] [Indexed: 12/19/2022] Open
Abstract
The purpose of a pharmacogenomic approach is to tailor treatment on the basis of an individual human genotype. This strategy is becoming increasingly common in medicine, and important results have been obtained in oncologic and antimicrobial therapies. The rapid technological developments and availability of innovative methodologies have revealed the existence of numerous genotypes that can influence the action of medications and give rise to the idea that a true "individualized" approach could become in the future a reality in clinical practice. Moreover, compared to the past, genotype analyses are now more easily available at accessible cost. Concerning human reproduction, there is ample evidence that several variants of gonadotropins and their receptors influence female reproductive health and ovarian response to exogenous gonadotropins. In more detail, variants in genes of follicle-stimulating hormone β-chain (FSH-B) and its receptor (FSH-R) seem to be the most promising candidates for a pharmacogenomic approach to controlled ovarian stimulation in assisted reproductive technologies. In the present review, we summarize the evidence regarding FSH-B and FSH-R variants, with special reference to their impact on reproductive health and assisted reproductive technology treatments.
Collapse
Affiliation(s)
- Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
- *Correspondence: Alessandro Conforti
| | - Alberto Vaiarelli
- G.E.N.E.R.A. Centre for Reproductive Medicine, Clinica Valle Giulia, Rome, Italy
| | - Danilo Cimadomo
- G.E.N.E.R.A. Centre for Reproductive Medicine, Clinica Valle Giulia, Rome, Italy
| | - Francesca Bagnulo
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Stefania Peluso
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Luigi Carbone
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Francesca Di Rella
- Medical Oncology, Department of Senology, National Cancer Institute, IRCCS Fondazione G. Pascale, Naples, Italy
| | - Giuseppe De Placido
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Filippo Maria Ubaldi
- G.E.N.E.R.A. Centre for Reproductive Medicine, Clinica Valle Giulia, Rome, Italy
| | - Ilpo Huhtaniemi
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Carlo Alviggi
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale (IEOS) Consiglio Nazionale delle Ricerche, Naples, Italy
| |
Collapse
|
19
|
Szymańska K, Kałafut J, Rivero-Müller A. The gonadotropin system, lessons from animal models and clinical cases. ACTA ACUST UNITED AC 2018; 70:561-587. [PMID: 30264954 DOI: 10.23736/s0026-4784.18.04307-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review article centers upon family of gonadotropin hormones which consists of two pituitary hormones - follicle-stimulating hormone (FSH) and luteinizing hormone (LH) as well as one non-pituitary hormone - human chorionic gonadotropin (hCG) secreted by placenta, and their receptors. Gonadotropins play an essential role in proper sexual development, puberty, gametogenesis, maintenance of pregnancy and male sexual differentiation during the fetal development. They belong to the family of glycoprotein hormones thus they constitute heterodimeric proteins built of common α subunit and hormone-specific β-subunit. Hitherto, several mutations in genes encoding both gonadotropins and their receptors have been identified in humans. Their occurrence resulted in a number of different phenotypes including delayed puberty, primary amenorrhea, hermaphroditism, infertility and hypogonadism. In order to understand the effects of mutations on the phenotype observed in affected patients, detailed molecular studies are required to map the relationship between the structure and function of gonadotropins and their receptors. Nonetheless, in vitro assays are often insufficient to understand physiology. Therefore, several animal models have been developed to unravel the physiological roles of gonadotropins and their receptors.
Collapse
|
20
|
Maione L, Dwyer AA, Francou B, Guiochon-Mantel A, Binart N, Bouligand J, Young J. GENETICS IN ENDOCRINOLOGY: Genetic counseling for congenital hypogonadotropic hypogonadism and Kallmann syndrome: new challenges in the era of oligogenism and next-generation sequencing. Eur J Endocrinol 2018; 178:R55-R80. [PMID: 29330225 DOI: 10.1530/eje-17-0749] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 01/10/2018] [Indexed: 12/22/2022]
Abstract
Congenital hypogonadotropic hypogonadism (CHH) and Kallmann syndrome (KS) are rare, related diseases that prevent normal pubertal development and cause infertility in affected men and women. However, the infertility carries a good prognosis as increasing numbers of patients with CHH/KS are now able to have children through medically assisted procreation. These are genetic diseases that can be transmitted to patients' offspring. Importantly, patients and their families should be informed of this risk and given genetic counseling. CHH and KS are phenotypically and genetically heterogeneous diseases in which the risk of transmission largely depends on the gene(s) responsible(s). Inheritance may be classically Mendelian yet more complex; oligogenic modes of transmission have also been described. The prevalence of oligogenicity has risen dramatically since the advent of massively parallel next-generation sequencing (NGS) in which tens, hundreds or thousands of genes are sequenced at the same time. NGS is medically and economically more efficient and more rapid than traditional Sanger sequencing and is increasingly being used in medical practice. Thus, it seems plausible that oligogenic forms of CHH/KS will be increasingly identified making genetic counseling even more complex. In this context, the main challenge will be to differentiate true oligogenism from situations when several rare variants that do not have a clear phenotypic effect are identified by chance. This review aims to summarize the genetics of CHH/KS and to discuss the challenges of oligogenic transmission and also its role in incomplete penetrance and variable expressivity in a perspective of genetic counseling.
Collapse
Affiliation(s)
- Luigi Maione
- University of Paris-Sud, Paris-Sud Medical School, Le Kremlin-Bicêtre, France
- Department of Reproductive Endocrinology, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, France
- INSERM U1185, Le Kremlin-Bicêtre, France
| | - Andrew A Dwyer
- Boston College, William F. Connell School of Nursing, Chestnut Hill, Massachusetts, USA
| | - Bruno Francou
- University of Paris-Sud, Paris-Sud Medical School, Le Kremlin-Bicêtre, France
- INSERM U1185, Le Kremlin-Bicêtre, France
- Department of Molecular Genetics, Pharmacogenomics, and Hormonology, Le Kremlin-Bicêtre, France
| | - Anne Guiochon-Mantel
- University of Paris-Sud, Paris-Sud Medical School, Le Kremlin-Bicêtre, France
- INSERM U1185, Le Kremlin-Bicêtre, France
- Department of Molecular Genetics, Pharmacogenomics, and Hormonology, Le Kremlin-Bicêtre, France
| | - Nadine Binart
- University of Paris-Sud, Paris-Sud Medical School, Le Kremlin-Bicêtre, France
- INSERM U1185, Le Kremlin-Bicêtre, France
| | - Jérôme Bouligand
- University of Paris-Sud, Paris-Sud Medical School, Le Kremlin-Bicêtre, France
- INSERM U1185, Le Kremlin-Bicêtre, France
- Department of Molecular Genetics, Pharmacogenomics, and Hormonology, Le Kremlin-Bicêtre, France
| | - Jacques Young
- University of Paris-Sud, Paris-Sud Medical School, Le Kremlin-Bicêtre, France
- Department of Reproductive Endocrinology, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, France
- INSERM U1185, Le Kremlin-Bicêtre, France
| |
Collapse
|
21
|
Wang H, Chen L, Jiang Y, Gao S, Chen S, Zheng X, Liu Z, Zhao Y, Li H, Yu J, Wang F, Liu Y, Li C, Zhou X. Association of gene polymorphisms of estrogen receptor, follicle-stimulating hormone β and leptin with follicular cysts in Large White sows. Theriogenology 2017; 103:143-148. [DOI: 10.1016/j.theriogenology.2017.03.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 12/24/2022]
|
22
|
|
23
|
Abstract
PURPOSE OF REVIEW To describe pubertal testicular growth in humans, changes in testicular cell populations that result in testicular growth, and the role of testosterone and gonadotrophins follicle-stimulating hormone (FSH) and luteinizing hormone (LH) in testicular growth. When human data were not available, studies in nonhuman primates and/or rodents were used as surrogates. RECENT FINDINGS Testicular growth in puberty follows a sigmoidal growth curve, with a large variation in timing of testicular growth and adult testicular volume. Testicular growth early in puberty is due to increase in Sertoli cell number and length of seminiferous tubules, whereas the largest and fastest growth results from the increase in the diameter of the seminiferous tubules first due to spermatogonial proliferation and then due to the expansion of meiotic and haploid germ cells. FSH stimulates Sertoli cell and spermatogonial proliferation, whereas LH/testosterone is mandatory to complete spermatogenesis. However, FSH and LH/testosterone work in synergy and are both needed for normal spermatogenesis. SUMMARY Testicular growth during puberty is rapid, and mostly due to germ cell expansion and growth in seminiferous tubule diameter triggered by androgens. Pre-treatment with FSH before the induction of puberty may improve the treatment of hypogonadotropic hypogonadism, but remains to be proven.
Collapse
Affiliation(s)
- Jaakko J Koskenniemi
- Institute of Biomedicine, Department of Physiology, University of Turku, and Department of Paediatrics, Turku University Hospital, Turku, Finland
| | | | | |
Collapse
|
24
|
Zheng J, Mao J, Cui M, Liu Z, Wang X, Xiong S, Nie M, Wu X. Novel FSHβ mutation in a male patient with isolated FSH deficiency and infertility. Eur J Med Genet 2017; 60:335-339. [DOI: 10.1016/j.ejmg.2017.04.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 02/04/2017] [Accepted: 04/05/2017] [Indexed: 10/19/2022]
|
25
|
|
26
|
Laws KM, Drummond-Barbosa D. Control of Germline Stem Cell Lineages by Diet and Physiology. Results Probl Cell Differ 2017; 59:67-99. [PMID: 28247046 DOI: 10.1007/978-3-319-44820-6_3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tight coupling of reproduction to environmental factors and physiological status is key to long-term species survival. In particular, highly conserved pathways modulate germline stem cell lineages according to nutrient availability. This chapter focuses on recent in vivo studies in genetic model organisms that shed light on how diet-dependent signals control the proliferation, maintenance, and survival of adult germline stem cells and their progeny. These signaling pathways can operate intrinsically in the germ line, modulate the niche, or act through intermediate organs to influence stem cells and their differentiating progeny. In addition to illustrating the extent of dietary regulation of reproduction, findings from these studies have implications for fertility during aging or disease states.
Collapse
Affiliation(s)
- Kaitlin M Laws
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Daniela Drummond-Barbosa
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA. .,Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
| |
Collapse
|
27
|
Şimşek E, Montenegro LR, Binay C, Demiral M, Acıkalin MF, Latronico AC. Clinical and Hormonal Features of a Male Adolescent with Congenital Isolated Follicle-Stimulating Hormone Deficiency. Horm Res Paediatr 2016; 85:207-12. [PMID: 26625121 DOI: 10.1159/000442289] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/09/2015] [Indexed: 11/19/2022] Open
Abstract
AIM Our aim was to describe the clinical and genetic findings in an adolescent male with isolated follicle-stimulating hormone (FSH) deficiency and demonstrate the efficacy of recombinant human FSH (rhFSH) replacement in this case. METHODS A 14.5-year-old adolescent male was referred with normal pubertal development and small testes. Serum testosterone, FSH, and luteinising hormone (LH) were measured at baseline and after gonadotropin-releasing hormone (GnRH) stimulation. Testicular biopsy was performed, and rhFSH replacement was administered for 6 months. The patient's FSHβ gene was amplified and sequenced. RESULTS Basal and GnRH-stimulated FSH levels were undetectable, in contrast with increased LH levels under both conditions. Histopathological investigation of a testicular biopsy specimen revealed a reduced number of Sertoli cells, the absence of germ cells, Leydig cell hyperplasia, and a thickened basement membrane in seminiferous tubules. The testicular size changed from 1 ml at baseline to 6 ml after 6 months of rhFSH replacement. Sequencing of the FSHβ gene exon 3 revealed a new missense mutation (c.364T>C, resulting in p.Cys122Arg) in a homozygous state in the patient; both parents and a sister carried the same mutation in a heterozygous state. We also compared our case with all similar cases published previously. CONCLUSION We herein described an adolescent male with isolated FSH deficiency due to a novel FSHβ gene mutation associated with a prepubertal testes size and normal virilisation.
Collapse
Affiliation(s)
- Enver Şimşek
- Department of Paediatric Endocrinology, Eskisehir Osmangazi University School of Medicine, Eskisehir, Turkey
| | | | | | | | | | | |
Collapse
|
28
|
Ulloa-Aguirre A, Lira-Albarrán S. Clinical Applications of Gonadotropins in the Male. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 143:121-174. [PMID: 27697201 DOI: 10.1016/bs.pmbts.2016.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pituitary gonadotropins, luteinizing hormone (LH), and follicle-stimulating hormone (FSH) play a pivotal role in reproduction. The synthesis and secretion of gonadotropins are regulated by complex interactions among several endocrine, paracrine, and autocrine factors of diverse chemical structure. In men, LH regulates the synthesis of androgens by the Leydig cells, whereas FSH promotes Sertoli cell function and thereby influences spermatogenesis. Gonadotropins are complex molecules composed of two subunits, the α- and β-subunit, that are noncovalently associated. Gonadotropins are decorated with glycans that regulate several functions of the protein including folding, heterodimerization, stability, transport, conformational maturation, efficiency of heterodimer secretion, metabolic fate, interaction with their cognate receptor, and selective activation of signaling pathways. A number of congenital and acquired abnormalities lead to gonadotropin deficiency and hypogonadotropic hypogonadism, a condition amenable to treatment with exogenous gonadotropins. Several natural and recombinant preparations of gonadotropins are currently available for therapeutic purposes. The difference between natural and the currently available recombinant preparations (which are massively produced in Chinese hamster ovary cells for commercial purposes) mainly lies in the abundance of some of the carbohydrates that conform the complex glycans attached to the protein core. Whereas administration of exogenous gonadotropins in patients with isolated congenital hypogonadotropic hypogonadism is a well recognized therapeutic approach, their role in treating men with normogonadotropic idiopathic infertility is still controversial. This chapter concentrates on the main structural and functional features of the gonadotropin hormones and how basic concepts have been translated into the clinical arena to guide therapy for gonadotropin deficit in males.
Collapse
Affiliation(s)
- A Ulloa-Aguirre
- Research Support Network, Universidad Nacional Autónoma de México (UNAM)-National Institutes of Health, Mexico City, Mexico.
| | - S Lira-Albarrán
- Department of Reproductive Biology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
29
|
Zhang Z, Zhu B, Ge W. Genetic analysis of zebrafish gonadotropin (FSH and LH) functions by TALEN-mediated gene disruption. Mol Endocrinol 2016; 29:76-98. [PMID: 25396299 DOI: 10.1210/me.2014-1256] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Vertebrate reproduction is controlled by two gonadotropins (FSH and LH) from the pituitary. Despite numerous studies on FSH and LH in fish species, their functions in reproduction still remain poorly defined. This is partly due to the lack of powerful genetic approaches for functional studies in adult fish. This situation is now changing with the emergence of genome-editing technologies, especially Transcription Activator-Like Effector Nuclease (TALEN) and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR). In this study, we deleted the hormone-specific β-genes of both FSH and LH in the zebrafish using TALEN. This was followed by a phenotype analysis for key reproductive events, including gonadal differentiation, puberty onset, gametogenesis, final maturation, and fertility. FSH-deficient zebrafish (fshb(-/-)) were surprisingly fertile in both sexes; however, the development of both the ovary and testis was significantly delayed. In contrast, LH-deficient zebrafish (lhb(-/-)) showed normal gonadal growth, but the females failed to spawn and were therefore infertile. Using previtellogenic follicles as the marker, we observed a significant delay of puberty onset in the fshb mutant but not the lhb mutant females. Interestingly, FSH seemed to play a role in maintaining the female status because we repeatedly observed sexual reversal in the fshb mutant. Neither the fshb nor lhb mutation alone seemed to affect gonadal differentiation; however, the double mutation of the two genes led to all males, although the development of the testis was significantly delayed. In summary, our data confirmed some well-known functions of FSH and LH in fish while also providing evidence for novel functions, which would be difficult to reveal using traditional biochemical and physiological approaches.
Collapse
Affiliation(s)
- Zhiwei Zhang
- School of Life Sciences (Z.Z., B.Z., W.G.), The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China; and Faculty of Health Sciences (Z.Z., W.G.), University of Macau, Taipa, Macau, China
| | | | | |
Collapse
|
30
|
Supriya C, Akhila B, Pratap Reddy K, Girish BP, Sreenivasula Reddy P. Effects of maternal exposure to aflatoxin B1 during pregnancy on fertility output of dams and developmental, behavioral and reproductive consequences in female offspring using a rat model. Toxicol Mech Methods 2016; 26:202-10. [DOI: 10.3109/15376516.2016.1151967] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
31
|
Ruth KS, Beaumont RN, Tyrrell J, Jones SE, Tuke MA, Yaghootkar H, Wood AR, Freathy RM, Weedon MN, Frayling TM, Murray A. Genetic evidence that lower circulating FSH levels lengthen menstrual cycle, increase age at menopause and impact female reproductive health. Hum Reprod 2016; 31:473-81. [PMID: 26732621 PMCID: PMC4716809 DOI: 10.1093/humrep/dev318] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 11/25/2015] [Indexed: 12/22/2022] Open
Abstract
STUDY QUESTION How does a genetic variant in the FSHB promoter, known to alter FSH levels, impact female reproductive health? SUMMARY ANSWER The T allele of the FSHB promoter polymorphism (rs10835638; c.-211G>T) results in longer menstrual cycles and later menopause and, while having detrimental effects on fertility, is protective against endometriosis. WHAT IS KNOWN ALREADY The FSHB promoter polymorphism (rs10835638; c.-211G>T) affects levels of FSHB transcription and, as a result, circulating levels of FSH. FSH is required for normal fertility and genetic variants at the FSHB locus are associated with age at menopause and polycystic ovary syndrome (PCOS). STUDY DESIGN, SIZE, DURATION We used cross-sectional data from the UK Biobank to look at associations between the FSHB promoter polymorphism and reproductive traits, and performed a genome-wide association study (GWAS) for length of menstrual cycle. PARTICIPANTS/MATERIALS, SETTING, METHODS We included white British individuals aged 40-69 years in 2006-2010, in the May 2015 release of genetic data from UK Biobank. We tested the FSH-lowering T allele of the FSHB promoter polymorphism (rs10835638; c.-211G>T) for associations with 29, mainly female, reproductive phenotypes in up to 63 350 women and 56 608 men. We conducted a GWAS in 9534 individuals to identify genetic variants associated with length of menstrual cycle. MAIN RESULTS AND THE ROLE OF CHANCE The FSH-lowering T allele of the FSHB promoter polymorphism (rs10835638; MAF 0.16) was associated with longer menstrual cycles [0.16 SD (c. 1 day) per minor allele; 95% confidence interval (CI) 0.12-0.20; P = 6 × 10(-16)], later age at menopause (0.13 years per minor allele; 95% CI 0.04-0.22; P = 5.7 × 10(-3)), greater female nulliparity [odds ratio (OR) = 1.06; 95% CI 1.02-1.11; P = 4.8 × 10(-3)] and lower risk of endometriosis (OR = 0.79; 95% CI 0.69-0.90; P = 4.1 × 10(-4)). The FSH-lowering T allele was not associated with other female reproductive illnesses or conditions in our study and we did not replicate associations with male infertility or PCOS. In the GWAS for menstrual cycle length, only variants near the FSHB gene reached genome-wide significance (P < 5 × 10(-9)). LIMITATIONS, REASONS FOR CAUTION The data included might be affected by recall bias. Cycle length was not available for 25% of women still cycling (1% did not answer, 6% did not know and for 18% cycle length was recorded as 'irregular'). Women with a cycle length recorded were aged over 40 and were approaching menopause; however, we did not find evidence that this affected the results. Many of the groups with illnesses had relatively small sample sizes and so the study may have been under-powered to detect an effect. WIDER IMPLICATIONS OF THE FINDINGS We found a strong novel association between a genetic variant that lowers FSH levels and longer menstrual cycles, at a locus previously robustly associated with age at menopause. The variant was also associated with nulliparity and endometriosis risk. These findings should now be verified in a second independent group of patients. We conclude that lifetime differences in circulating levels of FSH between individuals can influence menstrual cycle length and a range of reproductive outcomes, including menopause timing, infertility, endometriosis and PCOS. STUDY FUNDING/COMPETING INTERESTS None. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Katherine S Ruth
- Genetics of Complex Traits, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter EX2 5DW, UK
| | - Robin N Beaumont
- Genetics of Complex Traits, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter EX2 5DW, UK
| | - Jessica Tyrrell
- Genetics of Complex Traits, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter EX2 5DW, UK
| | - Samuel E Jones
- Genetics of Complex Traits, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter EX2 5DW, UK
| | - Marcus A Tuke
- Genetics of Complex Traits, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter EX2 5DW, UK
| | - Hanieh Yaghootkar
- Genetics of Complex Traits, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter EX2 5DW, UK
| | - Andrew R Wood
- Genetics of Complex Traits, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter EX2 5DW, UK
| | - Rachel M Freathy
- Genetics of Complex Traits, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter EX2 5DW, UK
| | - Michael N Weedon
- Genetics of Complex Traits, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter EX2 5DW, UK
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter EX2 5DW, UK
| | - Anna Murray
- Genetics of Complex Traits, University of Exeter Medical School, RILD Level 3, Royal Devon and Exeter Hospital, Barrack Road, Exeter EX2 5DW, UK
| |
Collapse
|
32
|
Aherrahrou R, Aherrahrou Z, Erdmann J, Moumni M. Identification of a novel ovine LH-beta promoter region, which dramatically enhances its promoter activity. SPRINGERPLUS 2015; 4:466. [PMID: 26355566 PMCID: PMC4554545 DOI: 10.1186/s40064-015-1182-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 07/24/2015] [Indexed: 11/20/2022]
Abstract
The luteinizing hormone beta subunit (LH-beta) gene plays a critical role in reproduction. In order to characterize and analyze the promoter region of LH-beta in sheep, a genomic library was constructed in phage lambda gt 10 and screened. A novel region of 1,224 bp upstream from the targeted LH-beta gene was identified. Blasting this sequence showed a perfect homology for the first 721 bp sequence with an upstream ovine LH-beta sequence in the database. However, the remaining 5′-503 bp showed no sequence matching. DNA from Moroccan breeds was isolated and the whole region was amplified and confirmed by sequencing. To further confirm the promoter activity of this region, an in vitro analysis using a luciferase assay was carried out. An increase in the promoter activity of the whole region was demonstrated compared to the empty vector. More interestingly, the unpublished region significantly enhanced the promoter activity compared to the known region alone. To predict putative transcription factor binding-sites (TFBSs), an in silico analysis was performed using the TFSEARCH program. The region features many TFBSs and contains two palindrome sequences of 17- and 18-bp. Taken together, a novel region was identified and confirmed in sheep which contained a promoter activity rich with binding sites for a putative regulatory element as shown in silico.
Collapse
Affiliation(s)
- Redouane Aherrahrou
- Institute for Integrative and Experimental Genomics (IIEG), Luebeck University, Luebeck, Maria-Goeppert-Str. 1, 23562 Lübeck, Germany ; Department of Biology, Faculty of Sciences, Moulay Ismail University, Zitoune, BP 11201, 50000 Meknes, Morocco
| | - Zouhair Aherrahrou
- Institute for Integrative and Experimental Genomics (IIEG), Luebeck University, Luebeck, Maria-Goeppert-Str. 1, 23562 Lübeck, Germany
| | - Jeanette Erdmann
- Institute for Integrative and Experimental Genomics (IIEG), Luebeck University, Luebeck, Maria-Goeppert-Str. 1, 23562 Lübeck, Germany
| | - Mohieddine Moumni
- Department of Biology, Faculty of Sciences, Moulay Ismail University, Zitoune, BP 11201, 50000 Meknes, Morocco
| |
Collapse
|
33
|
Zacharin M. Pubertal induction in hypogonadism: Current approaches including use of gonadotrophins. Best Pract Res Clin Endocrinol Metab 2015; 29:367-83. [PMID: 26051297 DOI: 10.1016/j.beem.2015.01.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Primary disorders of the gonad or those secondary to abnormalities of the hypothalamic pituitary axis result in hypogonadism. The range of health problems of childhood and adolescence that affect this axis has increased, as most children now survive chronic illness, but many have persisting deficits in gonadal function as a result of their underlying condition or its treatment. An integrated approach to hormone replacement is needed to optimize adult hormonal and bone health, and to offer opportunities for fertility induction and preservation that were not considered possible in the past. Timing of presentation ranges from birth, with disorders of sexual development, through adolescent pubertal failure, to adult fertility problems. This review addresses diagnosis and management of hypogonadism and focuses on new management strategies to address current concerns with fertility preservation. These include Turner syndrome, and fertility presevation prior to childhood cancer treatment. New strategies for male hormone replacement therapy that may impinge upon future fertility are emphasized.
Collapse
Affiliation(s)
- Margaret Zacharin
- Endocrinologist, Dept of Endocrinology, Royal Children's Hospital, Parkville, Victoria 3052, Australia.
| |
Collapse
|
34
|
Glomerular hyperfiltration in hypogonadotropic hypogonadic patients: Overlooking a cache? Int Urol Nephrol 2015; 47:1099-103. [PMID: 25947333 DOI: 10.1007/s11255-015-0995-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 04/21/2015] [Indexed: 01/25/2023]
Abstract
PURPOSE To investigate renal function in idiopathic hypogonadotropic hypogonadic (IHH) patients by measuring glomerular filtration rate (GFR) using modification of diet in renal disease formula, and determine whether there is any relationship between GFR and testosterone levels. METHODS Thirty-three patients with IHH and 37 healthy control subjects participated in this study. RESULTS The IHH group showed statistically significant higher GFR and proteinuria with respect to the control group (163.1 ± 46.9 to 117.9 ± 30.5 mL/min, p < 0.001; 0.2 ± 0.1 to 0.08 ± 0.02 mg/dL, p = 0.041, respectively). Uric acid and creatinine levels were statistically lower than in the control group (4.6 ± 0.5-3.6 ± 0.9 mg/dL, p = 0.02; 0.7 ± 0.2 to 0.9 ± 0.2 mg/dL, p < 0.001, respectively). Hyperfiltration positively correlated with IHH in multivariate linear regression analyses (β = 0.591, p < 0.001). In addition, in the IHH group, we found that the GFR increased independently of body mass index and age. CONCLUSION Our study confirms that low testosterone in IHH patients is associated with glomerular hyperfiltration. Patients with IHH should be carefully monitored with respect to their GFR.
Collapse
|
35
|
Bhattacharya I, Basu S, Sarda K, Gautam M, Nagarajan P, Pradhan BS, Sarkar H, Devi YS, Majumdar SS. Low levels of Gαs and Ric8b in testicular sertoli cells may underlie restricted FSH action during infancy in primates. Endocrinology 2015; 156:1143-55. [PMID: 25549048 DOI: 10.1210/en.2014-1746] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
FSH acts via testicular Sertoli cells (Sc) bearing FSH receptor (FSH-R) for regulating male fertility. Despite an adult-like FSH milieu in infant boys and monkeys, spermatogenesis is not initiated until the onset of puberty. We used infant and pubertal monkey Sc to reveal the molecular basis underlying developmental differences of FSH-R signaling in them. Unlike pubertal Sc, increasing doses of FSH failed to augment cAMP production by infant Sc. The expression of Gαs subunit and Ric8b, which collectively activate adenylyl cyclase (AC) for augmenting cAMP production and gene transcription, were significantly low in infant Sc. However, forskolin, which acts directly on AC bypassing FSH-R, augmented cAMP production and gene transcription uniformly in both infant and pubertal Sc. FSH-induced Gαs mRNA expression was higher in pubertal Sc. However, Gαi-2 expression was down-regulated by FSH in pubertal Sc, unlike infant Sc. FSH failed, but forskolin or 8-Bromoadenosine 3',5'-cyclic monophosphate treatment to infant Sc significantly augmented the expression of transferrin, androgen binding protein, inhibin-β-B, stem cell factor, and glial-derived neurotropic factor, which are usually up-regulated by FSH in pubertal Sc during spermatogenic onset. This suggested that lack of FSH mediated down-regulation of Gαi-2 expression and limited expression of Gαs subunit as well as Ric8b may underlie limited FSH responsiveness of Sc during infancy. This study also divulged that intracellular signaling events downstream of FSH-R are in place and can be activated exogenously in infant Sc. Additionally, this information may help in the proper diagnosis and treatment of infertile individuals having abnormal G protein-coupled FSH-R.
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Cellular Endocrinology Laboratory (I.B., S.B., K.S., M.G., B.S.P., H.S., Y.S.D., S.S.M.) and Primate Research Centre (P.N., S.S.M.), National Institute of Immunology, New Delhi, India 110067
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Jonas KC, Oduwole OO, Peltoketo H, Rulli SB, Huhtaniemi IT. Mouse models of altered gonadotrophin action: insight into male reproductive disorders. Reproduction 2014; 148:R63-70. [DOI: 10.1530/rep-14-0302] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The advent of technologies to genetically manipulate the mouse genome has revolutionised research approaches, providing a unique platform to study the causality of reproductive disorders in vivo. With the relative ease of generating genetically modified (GM) mouse models, the last two decades have yielded multiple loss-of-function and gain-of-function mutation mouse models to explore the role of gonadotrophins and their receptors in reproductive pathologies. This work has provided key insights into the molecular mechanisms underlying reproductive disorders with altered gonadotrophin action, revealing the fundamental roles of these pituitary hormones and their receptors in the hypothalamic–pituitary–gonadal axis. This review will describe GM mouse models of gonadotrophins and their receptors with enhanced or diminished actions, specifically focusing on the male. We will discuss the mechanistic insights gained from these models into male reproductive disorders, and the relationship and understanding provided into male human reproductive disorders originating from altered gonadotrophin action.
Collapse
|
37
|
Shiraishi K. Hormonal therapy for non-obstructive azoospermia: basic and clinical perspectives. Reprod Med Biol 2014; 14:65-72. [PMID: 29259404 DOI: 10.1007/s12522-014-0193-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 09/02/2014] [Indexed: 11/29/2022] Open
Abstract
Microdissection testicular sperm extraction (micro-TESE) combined with intracytoplasmic sperm injection is a standard therapeutic option for patients with non-obstructive azoospermia (NOA). Hormonal treatment has been believed to be ineffective for NOA because of high gonadotropin levels; however, several studies have stimulated spermatogenesis before or after micro-TESE by using anti-estrogens, aromatase inhibitors, and gonadotropins. These results remain controversial; however, it is obvious that some of the patients showed a distinct improvement in sperm retrieval by micro-TESE, and sperm was observed in the ejaculates of a small number of NOA patients. One potential way to improve spermatogenesis is by optimizing the intratesticular testosterone (ITT) levels. ITT has been shown to be increased after hCG-based hormonal therapy. The androgen receptor that is located on Sertoli cells plays a major role in spermatogenesis, and other hormonal and non-hormonal factors may also be involved. Before establishing a new hormonal treatment protocol to stimulate spermatogenesis in NOA patients, further basic investigations regarding the pathophysiology of spermatogenic impairment are needed. Gaining a better understanding of this issue will allow us to tailor a specific treatment for each patient.
Collapse
Affiliation(s)
- Koji Shiraishi
- Department of Urology Yamaguchi University School of Medicine 755-8505 Ube Yamaguchi Japan
| |
Collapse
|
38
|
ur Rehman K, Shahid K, Humayun H. Hypogonadotropic hypogonadism: new identification of testicular blood flow and varicocele after treatment with gonadotropins. Fertil Steril 2014; 102:700-704.e1. [PMID: 24934486 DOI: 10.1016/j.fertnstert.2014.05.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 05/17/2014] [Accepted: 05/19/2014] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To investigate testicular changes in patients with hypogonadotropic hypogonadism (HH) after treatment with gonadotropins. DESIGN Patients with HH were investigated and followed before and after treatment. SETTING Urology and andrology clinic of a teaching hospital. PATIENT(S) Consecutive male patients with diagnosed HH. INTERVENTION(S) All patients were treated with gonadotropins during the study period and later. MAIN OUTCOME MEASURE(S) The hormonal status and scrotal color Doppler ultrasound (CDUS) of patients was recorded before and after treatment. RESULT(S) Twenty-six patients with HH (ages 18-43 years) were followed for 8-29 months. After treatment, serum T and secondary sex characters improved in all and spermatogenesis developed in 61.5% of patients. Before treatment, testicular (intraparenchymal blood flow) was undetectable in all and barely detectable in three patients. This improved significantly to 4.53±5.44 and 4.27±4.97 cm/second, respectively, after treatment. Subcapsular arterial flow and testicular size also improved significantly. Similarly, after treatment, transverse epididymal diameter (TED) increased significantly. At baseline, no patient had detectable varicocele on CDUS. After treatment, varicocele was demonstrable in 23% of patients. This finding was further evaluated retrospectively from our 76 HH patient files. None of them had varicocele before treatment, but after treatment 19.73% were found to have varicocele. CONCLUSION(S) Patients with HH responded to gonadotropins by improvement in testicular blood flow and increase in TED. In some patients, varicocele was found to develop after treatment.
Collapse
Affiliation(s)
- Khaleeq ur Rehman
- Fatima Memorial Hospital, College of Medicine and Dentistry, Lahore, Pakistan.
| | - Khubaib Shahid
- Fatima Memorial Hospital, College of Medicine and Dentistry, Lahore, Pakistan
| | - Hina Humayun
- Fatima Memorial Hospital, College of Medicine and Dentistry, Lahore, Pakistan
| |
Collapse
|
39
|
Trabado S, Lamothe S, Maione L, Bouvattier C, Sarfati J, Brailly-Tabard S, Young J. Congenital hypogonadotropic hypogonadism and Kallmann syndrome as models for studying hormonal regulation of human testicular endocrine functions. ANNALES D'ENDOCRINOLOGIE 2014; 75:79-87. [PMID: 24815726 DOI: 10.1016/j.ando.2014.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Men with Kallmann syndrome (KS) and those with congenital isolated hypogonadotropic hypogonadism with normal olfaction share a chronic, usually profound deficit, in FSH and LH, the two pituitary gonadotropins. Many studies indicate that this gonadotropin deficiency is already present during fetal life, thus explaining the micropenis, cryptorchidism and marked testicular hypotrophy already present at birth. In addition, neonatal activation of gonadotropin secretion is compromised in boys with severe CHH/Kallmann, preventing the first phase of postnatal testicular activation. Finally, CHH is characterized by the persistence, in the vast majority of cases, of gonadotropin deficiency at the time of puberty and during adulthood. This prevents the normal pubertal testicular reactivation required for physiological sex steroid and testicular peptide production, and for spermatogenesis. CHH/KS thus represents a pathological paradigm that can help to unravel, in vivo, the role of each gonadotropin in human testicular exocrine and endocrine functions at different stages of development. Recombinant gonadotropins with pure LH or FSH activity have been used to stimulate Leydig's cells and Sertoli's cells, respectively, and thereby to clarify their paracrine interaction in vivo. The effects of these pharmacological probes can be assessed by measuring the changes they provoke in circulating testicular hormone concentrations. This review discusses the impact of chronic gonadotropin deficiency on the endocrine functions of the interstitial compartment, which contains testosterone-, estradiol- and INSL3-secreting Leydig's cells. It also examines the regulation of inhibin B and anti-Mullerian hormone (AMH) secretion in the seminiferous tubules, and the insights provided by studies of human testicular stimulation with recombinant gonadotropins, used either individually or in combination.
Collapse
Affiliation(s)
- Séverine Trabado
- Inserm U693, université Paris-Sud, 94275 Le Kremlin-Bicêtre, France; Laboratoire d'hormonologie et génétique, hôpital Bicêtre, Assistance publique-Hôpitaux de Paris, France
| | - Sophie Lamothe
- Service d'endocrinologie et des maladies de la reproduction, Hôpital Bicêtre, Assistance publique-Hôpitaux de Paris, 94275 Le Kremlin-Bicêtre, France
| | - Luigi Maione
- Service d'endocrinologie et des maladies de la reproduction, Hôpital Bicêtre, Assistance publique-Hôpitaux de Paris, 94275 Le Kremlin-Bicêtre, France
| | - Claire Bouvattier
- Département d'endocrinologie pédiatrique, hôpital Bicêtre, Assistance publique-Hôpitaux de Paris, 94275 Le Kremlin-Bicêtre, France
| | - Julie Sarfati
- Service d'endocrinologie et des maladies de la reproduction, Hôpital Bicêtre, Assistance publique-Hôpitaux de Paris, 94275 Le Kremlin-Bicêtre, France
| | - Sylvie Brailly-Tabard
- Inserm U693, université Paris-Sud, 94275 Le Kremlin-Bicêtre, France; Laboratoire d'hormonologie et génétique, hôpital Bicêtre, Assistance publique-Hôpitaux de Paris, France
| | - Jacques Young
- Inserm U693, université Paris-Sud, 94275 Le Kremlin-Bicêtre, France; Service d'endocrinologie et des maladies de la reproduction, Hôpital Bicêtre, Assistance publique-Hôpitaux de Paris, 94275 Le Kremlin-Bicêtre, France.
| |
Collapse
|
40
|
O'Shaughnessy PJ. Hormonal control of germ cell development and spermatogenesis. Semin Cell Dev Biol 2014; 29:55-65. [DOI: 10.1016/j.semcdb.2014.02.010] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 01/27/2023]
|
41
|
Ramaswamy S, Weinbauer GF. Endocrine control of spermatogenesis: Role of FSH and LH/ testosterone. SPERMATOGENESIS 2014; 4:e996025. [PMID: 26413400 PMCID: PMC4581062 DOI: 10.1080/21565562.2014.996025] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 12/04/2014] [Indexed: 12/21/2022]
Abstract
Evaluation of testicular functions (production of sperm and androgens) is an important aspect of preclinical safety assessment and testicular toxicity is comparatively far more common than ovarian toxicity. This chapter focuses (1) on the histological sequelae of disturbed reproductive endocrinology in rat, dog and nonhuman primates and (2) provides a review of our current understanding of the roles of gonadotropins and androgens. The response of the rodent testis to endocrine disturbances is clearly different from that of dog and primates with different germ cell types and spermatogenic stages being affected initially and also that the end-stage spermatogenic involution is more pronounced in dog and primates compared to rodents. Luteinizing hormone (LH)/testosterone and follicle-stimulating hormone (FSH) are the pivotal endocrine factors controlling testicular functions. The relative importance of either hormone is somewhat different between rodents and primates. Generally, however, both LH/testosterone and FSH are necessary for quantitatively normal spermatogenesis, at least in non-seasonal species.
Collapse
Affiliation(s)
- Suresh Ramaswamy
- Center for Research in Reproductive Physiology (CRRP); Department of Obstetrics, Gynecology & Reproductive Sciences; University of Pittsburgh School of Medicine; Magee-Womens Research Institute; Pittsburgh, PA USA
| | | |
Collapse
|
42
|
Regulation of spermatogenesis: An evolutionary biologist's perspective. Semin Cell Dev Biol 2014; 29:2-16. [DOI: 10.1016/j.semcdb.2014.03.007] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/28/2014] [Accepted: 03/04/2014] [Indexed: 02/03/2023]
|
43
|
Cocco C, Brancia C, D'Amato F, Noli B. Pituitary gonadotropins and autoimmunity. Mol Cell Endocrinol 2014; 385:97-104. [PMID: 24153235 DOI: 10.1016/j.mce.2013.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 10/08/2013] [Accepted: 10/09/2013] [Indexed: 10/26/2022]
Abstract
Autoimmune disease occurs when the body produces an inappropriate immune response against its own tissues producing antibodies, called autoantibodies, reacting to specific antigens. Studies regarding the presence of an autoimmune process specifically involving gonadotropins date from over than 20 years ago, when antibodies to gonadotropic-secreting cells were found by immunofluorescence in sera from a group of patients affected by cryptorchidism. Later on, antibodies detected by the same technique, and directed to the same cells were also found at high titer in sera from patients affected by hypogonadotropic hypogonadism, Kallmann's syndrome, lymphocytic hypophysitis with isolated gonadotropin deficiency, as well as autoimmune polyendocrine syndrome. Concerning the autoimmune target/s within the gonadotropic cells, rarely autoantibodies were found labeling gonadotropins while in a large number of cases, auto-antigens remained to be identified. Since pituitary gonadotropins are fundamental for the sexual maturity and reproductive mechanisms, patients with infertility were largely investigated by enzyme-linked immunosorbent assay for the presence of circulating antibodies likely interfering with gonadotropin activity. In infertile women, autoantibodies to gonadotropins were found related to ovarian autoimmunity, ovarian disorders that cause infertility and also associated with in vitro fertilization treatments. In infertile men, autoantibodies to gonadotropins may alter the testicular spermatogenesis and cause apoptosis of the spermatogenic cells. In conclusion, circulating antibodies were found labeling gonadotropic cells and/or gonadotropins, and in both cases they could create dysfunctions in gonadotropin related mechanism. The intriguing question of what can cause the production of such autoantibodies is not clear yet.
Collapse
Affiliation(s)
- Cristina Cocco
- NEF-Laboratory, Department of Biomedical Science, University of Cagliari, 09042 Monserrato, Cagliari, Italy.
| | - Carla Brancia
- NEF-Laboratory, Department of Biomedical Science, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Filomena D'Amato
- NEF-Laboratory, Department of Biomedical Science, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| | - Barbara Noli
- NEF-Laboratory, Department of Biomedical Science, University of Cagliari, 09042 Monserrato, Cagliari, Italy
| |
Collapse
|
44
|
Li X, Mao Z, Wu M, Xia J. Rescuing infertility of Pick1 knockout mice by generating testis-specific transgenic mice via testicular infection. Sci Rep 2013; 3:2842. [PMID: 24100262 PMCID: PMC3792414 DOI: 10.1038/srep02842] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 09/02/2013] [Indexed: 12/16/2022] Open
Abstract
PICK1 (protein interacting with C-kinase 1) is a peripheral membrane protein with high expression in brain, testis, pancreas and other neuroendocrine tissues. Male Pick1 knockout mice are completely infertile, with a phenotype resembling the human disease globozoospermia. Since PICK1 is expressed in both testis and neuroendocrine tissues, infertility of Pick1 knockout mice may be due to either impaired neuroendocrine function or abnormal spermatogenesis. To distinguish these two possibilities, we restored PICK1's expression in the testis by seminiferous tubule microinjection of PICK1-containing lentivirus. By examining the testis-specific Pick1 transgenic mice, we found that PICK1's expression in testis rescued the spermatogenic abnormalities and male infertility in Pick1 knockout mice. Our results indicate that the infertility is caused by the lack of PICK1 in the testis rather than in other organs. In addition, we found that seminiferous tubule microinjection of lentivirus has a strong preference to produce testis-specific transgenic mice.
Collapse
Affiliation(s)
- Xiumao Li
- Division of Life Science, Division of Biomedical Engineering and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Zhuo Mao
- Division of Life Science, Division of Biomedical Engineering and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Min Wu
- Division of Life Science, Division of Biomedical Engineering and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Jun Xia
- Division of Life Science, Division of Biomedical Engineering and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| |
Collapse
|
45
|
Benson CA, Kurz TL, Thackray VG. A human FSHB promoter SNP associated with low FSH levels in men impairs LHX3 binding and basal FSHB transcription. Endocrinology 2013; 154:3016-21. [PMID: 23766128 PMCID: PMC3749480 DOI: 10.1210/en.2013-1294] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
FSH production is important for human gametogenesis. In addition to inactivating mutations in the FSHB gene, which result in infertility in both sexes, a G/T single-nucleotide polymorphism (SNP) at -211 relative to the transcription start site of the 5' untranslated region of FSHB has been reported to be associated with reduced serum FSH levels in men. In this study, we sought to identify the potential mechanism by which the -211 SNP reduces FSH levels. Although the SNP resides in a putative hormone response element, we showed that, unlike the murine gene, human FSHB was not induced by androgens or progestins in gonadotropes. On the other hand, we found that the LHX3 homeodomain transcription factor bound to an 11-bp element in the human FSHB promoter that includes the -211 nucleotide. Furthermore, we also demonstrated that LHX3 bound with greater affinity to the wild-type human FSHB promoter compared with the -211 G/T mutation and that LHX3 binding was more effectively competed with excess wild-type oligonucleotide than with the SNP. Finally, we showed that FSHB transcription was decreased in gonadotrope cells with the -211 G/T mutation compared with the wild-type FSHB promoter. Altogether, our results suggest that decreased serum FSH levels in men with the SNP likely result from reduced LHX3 binding and induction of FSHB transcription.
Collapse
Affiliation(s)
- Courtney A Benson
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | | | | |
Collapse
|
46
|
Ruvolo G, Roccheri MC, Brucculeri AM, Longobardi S, Cittadini E, Bosco L. Lower sperm DNA fragmentation after r-FSH administration in functional hypogonadotropic hypogonadism. J Assist Reprod Genet 2013; 30:497-503. [PMID: 23435529 DOI: 10.1007/s10815-013-9951-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 01/28/2013] [Indexed: 01/09/2023] Open
Abstract
PURPOSE An observational clinical and molecular study was designed to evaluate the effects of the administration of recombinant human FSH on sperm DNA fragmentation in men with a non-classical form of hypogonadotropic hypogonadism and idiopathic oligoasthenoteratozoospermia. METHODS In the study were included 53 men with a non-classical form of hypogonadotropic hypogonadism and idiopathic oligoasthenoteratozoospermia. In all patients, sperm DNA fragmentation index (DFI), assessed by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate (dUTP) in situ DNA nick end-labelling (TUNEL) assay, was evaluated before starting the treatment with 150 IU of recombinant human FSH, given three times a week for at least 3 months. Patients' semen analysis and DNA fragmentation index were re-evaluated after the 3-month treatment period. RESULTS After recombinant human FSH therapy, we did not find any differences in terms of sperm count, motility and morphology. The average DNA fragmentation index was significantly reduced (21.15 vs 15.2, p<0.05), but we found a significant reduction in patients with high basal DFI values (>15 %), while no significant variation occurred in the patients with DFI values ≤ 15 %. CONCLUSIONS Recombinant human FSH administration improves sperm DNA integrity in hypogonadotropic hypogonadism and idiopathic oligoasthenoteratozoospermia men with DNA fragmentation index value >15 % .
Collapse
Affiliation(s)
- Giovanni Ruvolo
- Centro di Biologia della Riproduzione, via V. Villareale 54, 90141, Palermo, Italy.
| | | | | | | | | | | |
Collapse
|
47
|
Tran S, Zhou X, Lafleur C, Calderon MJ, Ellsworth BS, Kimmins S, Boehm U, Treier M, Boerboom D, Bernard DJ. Impaired fertility and FSH synthesis in gonadotrope-specific Foxl2 knockout mice. Mol Endocrinol 2013; 27:407-21. [PMID: 23340250 DOI: 10.1210/me.2012-1286] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Impairments in pituitary FSH synthesis or action cause infertility. However, causes of FSH dysregulation are poorly described, in part because of our incomplete understanding of mechanisms controlling FSH synthesis. Previously, we discovered a critical role for forkhead protein L2 (FOXL2) in activin-stimulated FSH β-subunit (Fshb) transcription in immortalized cells in vitro. Here, we tested the hypothesis that FOXL2 is required for FSH synthesis in vivo. Using a Cre/lox approach, we selectively ablated Foxl2 in murine anterior pituitary gonadotrope cells. Conditional knockout (cKO) mice developed overtly normally but were subfertile in adulthood. Testis size and spermatogenesis were significantly impaired in cKO males. cKO females exhibited reduced ovarian weight and ovulated fewer oocytes in natural estrous cycles compared with controls. In contrast, ovaries of juvenile cKO females showed normal responses to exogenous gonadotropin stimulation. Both male and female cKO mice were FSH deficient, secondary to diminished pituitary Fshb mRNA production. Basal and activin-stimulated Fshb expression was similarly impaired in Foxl2 depleted primary pituitary cultures. Collectively, these data definitively establish FOXL2 as the first identified gonadotrope-restricted transcription factor required for selective FSH synthesis in vivo.
Collapse
Affiliation(s)
- Stella Tran
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Choi D. The consequences of mutations in the reproductive endocrine system. Dev Reprod 2012; 16:235-51. [PMID: 25949097 PMCID: PMC4282240 DOI: 10.12717/dr.2012.16.4.235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 12/01/2012] [Accepted: 12/10/2012] [Indexed: 01/03/2023]
Abstract
The reproductive activity in male mammals is well known to be regulated by the hypothalamus-pituitary- gonad axis. The hypothalamic neurons secreting gonadotropin releasing hormone (GnRH) govern the reproductive neuroendocrine system by integrating all the exogenous information impinging on themselves. The GnRH synthesized and released from the hypothalamus arrives at the anterior pituitary through the portal vessels, provoking the production of the gonadotropins(follicle-stimulating hormone (FSH) and luteinizing hormone (LH)) at the same time. The gonadotropins affect the gonads to promote spermatogenesis and to secret testosterone. Testosterone acts on the GnRH neurons by a feedback loop through the circulatory system, resulting in the balance of all the hormones by regulating reproductive activities. These hormones exert their effects by acting on their own receptors, which are included in the signal transduction pathways as well. Unexpected aberrants are arised during this course of action of each hormone. This review summarizes these abnormal phenomena, including various mutations of molecules and their actions related to the reproductive function.
Collapse
Affiliation(s)
- Donchan Choi
- Dept. of Life Science, College of Environmental Sciences, Yong-In University, Yongin 449-714, Korea
| |
Collapse
|
49
|
Siegel ET, Kim HG, Nishimoto HK, Layman LC. The molecular basis of impaired follicle-stimulating hormone action: evidence from human mutations and mouse models. Reprod Sci 2012. [PMID: 23184658 DOI: 10.1177/1933719112461184] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The pituitary gonadotropin follicle-stimulating hormone (FSH) interacts with its membrane-bound receptor to produce biologic effects. Traditional functions of FSH include follicular development and estradiol production in females, and the regulation of Sertoli cell action and spermatogenesis in males. Knockout mice for both the ligand (Fshb) and the receptor (Fshr) serve as models for FSH deficiency, while Fshb and Fshr transgenic mice manifest FSH excess. In addition, inactivating mutations of both human orthologs (FSHB and FSHR) have been characterized in a small number of patients, with phenotypic effects of the ligand disruption being more profound than those of its receptor. Activating human FSHR mutants have also been described in both sexes, leading to a phenotype of normal testis function (male) or spontaneous ovarian hyperstimulation syndrome (females). As determined from human and mouse models, FSH is essential for normal puberty and fertility in females, particularly for ovarian follicular development beyond the antral stage. In males, FSH is necessary for normal spermatogenesis, but there are differences in human and mouse models. The FSHB mutations in humans result in azoospermia; while FSHR mutations in humans and knockouts of both the ligand and the receptor in mice affect testicular function but do not result in absolute infertility. Available evidence also indicates that FSH may also be necessary for normal androgen synthesis in males and females.
Collapse
Affiliation(s)
- Eric T Siegel
- Department of Obstetrics & Gynecology, Section of Reproductive Endocrinology, Infertility, & Genetics, Medical College of Georgia, Georgia Health Sciences University Augusta, GA 30912, USA
| | | | | | | |
Collapse
|
50
|
Abstract
INTRODUCTION Three single-nucleotide polymorphisms (SNPs) in the leptin (LEP) or leptin receptor (LEPR) genes were assessed for their association with central precocious puberty (CPP). RESULTS The control group with the A/G SNP at LEPR 223 or A/G SNP at LEPR 109 exhibited significantly higher peak luteinizing hormone (LH) levels. The leptin level in the CPP group was significantly higher than that in the control group, but SNPs in either LEP or LEPR gene could not explain this observation. DISCUSSION In conclusion, SNPs at LEPR 223 and LEPR 109 were significantly associated with higher levels of LH in girls without CPP, but none of the genotypes at these SNPs were significantly associated with CPP. METHODS The SNP genotypes of LEP (polymorphism at promoter at nt-2548) and LEPR (223A/G, 109A/G) of 219 healthy girls and 249 girls diagnosed with CPP were compared. Allele frequencies in SNPs were compared with anthropometric measures, circulating leptin, hormones (estradiol, follicle-stimulating hormone, and LH), and lipid concentrations for CPP risk.
Collapse
|