1
|
Graziani A, Grande G, Scafa R, Selice R, Garolla A, Rocca MS, Vinanzi C, Ferlin A. Pharmacogenetics of follicle-stimulating hormone action in the male. Andrology 2025. [PMID: 40304702 DOI: 10.1111/andr.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/11/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
Male factor infertility (MFI) is involved in half of the cases of couple infertility. The follicle-stimulating hormone (FSH) therapy is considered efficient to improve semen parameters and pregnancy rate in patients with idiopathic MFI, following the lesson learned from hypogonadotropic hypogonadism. However, while in patients with hypogonadotropic hypogonadism FSH therapy, in combination with human chorionic gonadotropin (hCG), is a well-established treatment, in patients with MFI the effects of the FSH therapy are variable and unpredictable. The FSH therapy in MFI should be a personalized treatment, tailored on the characteristics of the male patient and the couple. The pivotal aspect is the accurate identification of patients who might benefit from such treatment (responders) from those who might not (nonresponders). To date, selection of patients to be treated is based on history, physical examination, semen analysis, and hormonal assessment. However, these parameters cannot adequately identify a priori responder patients. Furthermore, tailored management should include pharmacological adaptation (dosage and duration of the therapy), as happens during ovarian hyperstimulation in assisted reproductive technologies. In a fully personalized therapy, pharmacogenetic factors must be considered. In this paper, we describe the evidence dealing with the pharmacogenetics of the FSH therapy in MFI, presenting the physiological and physiopathological basis and the pharmacogenetics studies dealing with effects of polymorphisms in the beta-subunit of FSH (FSHB) and the FSH receptor (FSHR) gene. According to the evidence so far available, genetic evaluation of FSHB and FSHR is recommended only for research purposes, since the data are not conclusive and even contrasting. Furthermore, the evidence so far is derived from quite small studies with different endpoints considered and relatively few cases. Better studies that consider the combined effect of several FSHB and FSHR gene polymorphisms, together with clinical, biochemical, seminal and testicular cytology, are necessary to develop an algorithm that might predict the response to the FSH treatment.
Collapse
Affiliation(s)
| | - Giuseppe Grande
- Department of Systems Medicine, Unit of Andrology and Reproductive Medicine, University Hospital of Padova, Padova, Italy
| | - Raffele Scafa
- Department of Medicine, University of Padova, Padova, Italy
| | - Riccardo Selice
- Department of Systems Medicine, Unit of Andrology and Reproductive Medicine, University Hospital of Padova, Padova, Italy
| | - Andrea Garolla
- Department of Medicine, University of Padova, Padova, Italy
- Department of Systems Medicine, Unit of Andrology and Reproductive Medicine, University Hospital of Padova, Padova, Italy
| | - Maria Santa Rocca
- Department of Systems Medicine, Unit of Andrology and Reproductive Medicine, University Hospital of Padova, Padova, Italy
| | - Cinzia Vinanzi
- Department of Systems Medicine, Unit of Andrology and Reproductive Medicine, University Hospital of Padova, Padova, Italy
| | - Alberto Ferlin
- Department of Medicine, University of Padova, Padova, Italy
- Department of Systems Medicine, Unit of Andrology and Reproductive Medicine, University Hospital of Padova, Padova, Italy
| |
Collapse
|
2
|
Neves AR, Garcia S, Vuong LN, Blockeel C, Spits C, Polyzos NP. The Additive Effect of Combinations of FSH Receptor Gene Variants in Ovarian Response to Stimulation. Reprod Sci 2024; 31:3560-3568. [PMID: 39322797 DOI: 10.1007/s43032-024-01700-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
To analyze whether combinations of polymorphisms within FSHR gene influence ovarian response (OR) to stimulation. A multicenter prospective cohort study was conducted from 11/2016-06/2019 in Europe and Asia including predicted normo-responders under 38y. Patients underwent ovarian stimulation using fixed-dose 150 IU rFSH in a GnRH antagonist protocol. FSHR variants rs6165, rs6166 and rs1394205 were genotyped and combined in diplotypes. OR was compared following multivariable regression. rs6165/rs6166 genotype AG/AG exhibited more hypo-response (33.1% vs. 24%,adjOR 1.77 [95%CI 1.08-2.90]) and lower Follicle to Oocyte Index (FOI) compared with other diplotypes (EMD -11.72 [95%CI -20.89;-2.55]). Genotype GG/AA showed less hypo-response (19.1% vs. 31%, adjOR 0.48 [95%CI 0.24-0.96]), while AA/AA had higher FOI (EMD 20.04 [95%CI 4.51;35.56]). Concerning rs6165/rs1394205, less oocytes (EMD -1.99 [95%CI -3.57;-0.42]) and lower FOI (EMD -12.07 [95%CI -23.09;-1.05]) were retrieved with genotype AG/AG and higher FORT with genotype AA/AG (EMD 17.88 [95%CI 3.77;31.98]). Regarding rs6166/rs1394205, less hypo-response (16.3% vs. 29.5%,adjOR 0.42 [95%CI 0.19-0.97]), more oocytes (EMD 3.45 [95%CI 1.57;5.34]) and higher FOI (EMD 17.57 [95%CI 4.41;30.73) were found with genotype AA/GG. Genotype AA/AG presented higher FORT (EMD 13.47 [95%CI 2.51,24.42]), while more hypo-response (56.3% vs. 26.4%,adjOR 6.30 [95%CI 1.88;21.08]) and lower FOI (EMD -23.51 [95%CI -45.04;-1.97]) was reported with AG/AA. In accordance with our previous studies, FSHR polymorphisms have a statistically significant impact on OR, both individually and in association. However, only rs6166/rs1394205 genotype AA/GG seems to have a clinically significant effect, with a decrease in the prevalence of hypo-response, higher oocyte yield and increase in FOI.
Collapse
Affiliation(s)
- Ana Raquel Neves
- IVI-RMA Lisboa, 1-9ª, Av. Infante Dom Henrique 333 H, Lisbon, Portugal.
- Dexeus Fertility, Dexeus University Hospital, 08028, Barcelona, Spain.
- Autonomous University of Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain.
| | - Sandra Garcia
- Dexeus Fertility, Dexeus University Hospital, 08028, Barcelona, Spain
| | - Lan N Vuong
- University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
- IVFMD and HOPE Research Center, My Duc Hospital, Ho Chi Minh City, Vietnam
| | - Christophe Blockeel
- Brussels IVF, Centre for Reproductive Medicine, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, Brussels, Belgium
- Department of Obstetrics and Gynecology, University of Zagreb-School of Medicine, Šalata 3, Zagreb, Croatia
- Research Group Genetics Reproduction and Development, Vrije Universiteit Brussel, Brussels, Belgium
| | - Claudia Spits
- Research Group Genetics Reproduction and Development, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nikolaos P Polyzos
- Dexeus Fertility, Dexeus University Hospital, 08028, Barcelona, Spain
- Faculty of Medicine and Health Sciences, Ghent University (UZ Gent), 9000 Gent, Belgium
| |
Collapse
|
3
|
Search for Associations of FSHR, INHA, INHAB, PRL, TNP2 and SPEF2 Genes Polymorphisms with Semen Quality in Russian Holstein Bulls (Pilot Study). Animals (Basel) 2021; 11:ani11102882. [PMID: 34679903 PMCID: PMC8532936 DOI: 10.3390/ani11102882] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 11/16/2022] Open
Abstract
The aim of the study was to search for new mutations in the previously studied gene loci of follicle-stimulating hormone receptor (FSHR), inhibin α (INHA), inhibin β A (INHAB), prolactin (PRL), transition protein 2 (TNP2), and sperm flagella 2 (SPEF2) by sequencing, as well as the search for associations of previously identified mutations at these loci with fresh semen quality in Russian Holstein bulls. Phenotypic data from 189 bulls was collected. Data was analyzed for most bulls for three years of semen collection. The maximum value of each semen quality indicator (doublet ejaculate volume, sperm concentration, progressive motility and total number of spermatozoa) were selected. SNPs were identified in the FSHR, INHA, INHAB, TNP2, SPEF2 genes. The PRL gene did not have polymorphism. Significant (p < 0.05) associations of polymorphisms in the FSHR gene with double ejaculate volume, concentration and total number of spermatozoa were identified. Polymorphism in the INHA gene was significantly associated (p < 0.05) with sperm concentration. Polymorphism in the INHAB gene was significantly associated (p < 0.05) with doublet ejaculate volume and total number of spermatozoa. Polymorphisms in the TNP2 and SPEF2 genes did not have significant associations with semen quality. The SNPs studied in our pilot work may be considered as candidate genetic markers in the selection of bulls.
Collapse
|
4
|
Song D, Hong L, Zhang ZF, Xu JH, Zhang HQ, Huang XL, Du J. The FSHR G-29A variant is not associated with the ovarian response to exogenous FSH stimulation. Am J Reprod Immunol 2021; 86:e13500. [PMID: 34558137 DOI: 10.1111/aji.13500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/13/2021] [Accepted: 09/20/2021] [Indexed: 11/28/2022] Open
Abstract
A common genetic variant in the follicle stimulating hormone receptor gene (FSHR) 5'-untranslated region has been previously reported to influence FSHR gene expression. However, studies on the ovarian response to exogenous gonadotropin stimulation are limited. The aim of this study was to evaluate the association of variants at positions -29 of the FSHR gene with the ovarian response to exogenous FSH stimulation in Chinese women. The genotypes of the FSHR gene were assayed using the Sequenom MassARRAY system. Total RNA and protein was extracted from granulosa cells, and FSHR expression at the mRNA and protein levels was assessed using quantitative PCR and western blotting. Our data revealed that there was no association between the FSHR genotype at the -29 position and the outcome of controlled ovarian stimulation. The expression of FSHR, at both the mRNA and protein levels, was similar amongst the different FSHR genotypes assessed, but was significantly reduced in the low responders. These results indicate that the variants caused by mutations at position -29 are not associated with ovarian response, and the low ovarian response to gonadotropin stimulation may be caused by decreased FSHR expression.
Collapse
Affiliation(s)
- Di Song
- Naval Medical University, Changhai Hospital, Shanghai, China
| | - Ling Hong
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhao-Feng Zhang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Jian-Hua Xu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| | - Hui-Qin Zhang
- Naval Medical University, Changhai Hospital, Shanghai, China
| | - Xian-Liang Huang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China.,Shanghai Institute of Planned Parenthood Research Hospital, Shanghai, China
| | - Jing Du
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Fudan University, Shanghai, China
| |
Collapse
|
5
|
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: 0.4] [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
|
6
|
Cannarella R, Musso N, Condorelli RA, Musmeci M, Stefani S, Aversa A, La Vignera S, Calogero AE. The 2039 A/G FSH receptor gene polymorphism influences glucose metabolism in healthy men. Endocrine 2020; 70:629-634. [PMID: 32681384 PMCID: PMC7674314 DOI: 10.1007/s12020-020-02420-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/06/2020] [Indexed: 11/12/2022]
Abstract
OBJECTIVE To assess the role of c. 2039 A/G (p. Asp680Ser) (rs6166) and c. -29 G/A (rs1394205) follicle-stimulating hormone receptor (FSHR) gene single nucleotide polymorphisms (SNPs) in a cohort of healthy men. METHODS One-hundred twenty-seven healthy men underwent evaluation of the anthropometric parameters, assessment of metabolic and lipid profile, measurement FSH serum levels, and genotyping of both the aforementioned FSHR SNPs. Data grouped according to the FSHR rs6166 or rs1394205 genotypes underwent to statistical analysis. MAIN RESULTS The three groups of men for each FSHR SNP did not differ statistically significantly for body mass index and serum FSH levels. As for FSHR rs6166 SNP, glucose levels were significantly lower in men with the GG genotype compared with those with the AA genotype. Men with AG had lower insulin levels and HOMA index values compared with those carrying the genotype AA (p < 0.05). The GG group showed a negative correlation between serum FSH levels and insulin and between serum FSH levels and HOMA index (p < 0.05). In contrast, men grouped according to the FSHR rs1394205 genotype showed no significant difference in blood glucose, serum insulin levels, and HOMA index. The AG group showed a negative correlation between FSH insulin and between serum FSH levels and HOMA index (p < 0.05). CONCLUSIONS Men with the genotype GG of the FSHR rs6166 SNP have lower blood glucose levels than those with the AA genotype. Their FSH levels inversely correlated with insulin and HOMA index. In contrast, the genotype FSHR rs6166 A/G did not reveal any role of FSH on glucose metabolism in healthy men. The inverse relationship between FSH and insulin or HOMA index in the group with the genotype GG of the FSHR rs6166 SNP suggests a possible cross-talk between FSH and insulin.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Nicolò Musso
- Bio-nanotech Research and Innovation Tower (BRIT), University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Marco Musmeci
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Stefania Stefani
- Bio-nanotech Research and Innovation Tower (BRIT), University of Catania, Catania, Italy
| | - Antonio Aversa
- Department of Experimental and Clinical Medicine, "Magna Graecia" University, Catanzaro, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
| |
Collapse
|
7
|
Cannarella R, Musso N, Condorelli RA, Musmeci M, Stefani S, La Vignera S, Calogero AE. Combined Effects of the FSHR 2039 A/G and FSHR -29 G/A Polymorphisms on Male Reproductive Parameters. World J Mens Health 2020; 39:516-525. [PMID: 33151046 PMCID: PMC8255408 DOI: 10.5534/wjmh.200070] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/29/2020] [Accepted: 09/07/2020] [Indexed: 01/15/2023] Open
Abstract
PURPOSE The aim of this study was to evaluate the combined effect of FSHR 2039 A/G and FSHR -29 G/A single nucleotide polymorphisms (SNPs) on the male reproductive function in a cohort of Sicilian men. MATERIALS AND METHODS One-hundred thirty Sicilian men were enrolled and underwent blood withdrawal for hormone measurement and FSHR 2039 A/G and FSHR -29 G/A SNP genotyping, testicular volume evaluation by ultrasound scan, and semen analysis. A meta-analysis of the FSHR -29 G/A SNP, evaluated in a previous study of the Sicilian population was done. RESULTS No genotype of the FSHR 2039 A/G SNP correlated with serum follicle-stimulating hormone (FSH) levels, testicular volume, sperm concentration, and total sperm count. In contrast, normozoospermic men with FSHR -29 GG and FSHR -29 GA genotypes had significantly lower sperm concentrations compared to men with the FSHR -29 AA genotype. The other sperm parameters did not show any significant difference. The meta-analysis showed no significant difference in serum FSH levels, testicular volume, sperm concentration, and total sperm count between FSHR -29 GG and FSHR -29 AA in Sicilian men. No difference was found even when the two SNPs were evaluated in combination. However, this combination was present, as expected, only in a low proportion (3.8%) of the men studied. CONCLUSIONS The SNPs FSHR 2039 A/G and FSHR -29 G/A in combination did not seem to have any effect on male reproductive function in a cohort of Sicilian men. The effect of these SNPs has only been studied in granulosa cells so far. Further studies on their role in Sertoli cells are needed.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
| | - Nicolò Musso
- Department of Biomedical and Biotechnological Science, BIOMETEC, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Marco Musmeci
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Stefania Stefani
- Department of Biomedical and Biotechnological Science, BIOMETEC-Sect. of Microbiology, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
8
|
Simoni M, Brigante G, Rochira V, Santi D, Casarini L. Prospects for FSH Treatment of Male Infertility. J Clin Endocrinol Metab 2020; 105:5831300. [PMID: 32374828 DOI: 10.1210/clinem/dgaa243] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022]
Abstract
CONTEXT Despite the new opportunities provided by assisted reproductive technology (ART), male infertility treatment is far from being optimized. One possibility, based on pathophysiological evidence, is to stimulate spermatogenesis with gonadotropins. EVIDENCE ACQUISITION We conducted a comprehensive systematic PubMed literature review, up to January 2020, of studies evaluating the genetic basis of follicle-stimulating hormone (FSH) action, the role of FSH in spermatogenesis, and the effects of its administration in male infertility. Manuscripts evaluating the role of genetic polymorphisms and FSH administration in women undergoing ART were considered whenever relevant. EVIDENCE SYNTHESIS FSH treatment has been successfully used in hypogonadotropic hypogonadism, but with questionable results in idiopathic male infertility. A limitation of this approach is that treatment plans for male infertility have been borrowed from hypogonadism, without daring to overstimulate, as is done in women undergoing ART. FSH effectiveness depends not only on its serum levels, but also on individual genetic variants able to determine hormonal levels, activity, and receptor response. Single-nucleotide polymorphisms in the follicle-stimulating hormone subunit beta (FSHB) and follicle-stimulating hormone receptor (FSHR) genes have been described, with some of them affecting testicular volume and sperm output. The FSHR p.N680S and the FSHB -211G>T variants could be genetic markers to predict FSH response. CONCLUSIONS FSH may be helpful to increase sperm production in infertile men, even if the evidence to recommend the use of FSH in this setting is weak. Placebo-controlled clinical trials, considering the FSHB-FSHR haplotype, are needed to define the most effective dosage, the best treatment length, and the criteria to select candidate responder patients.
Collapse
Affiliation(s)
- Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Institut Français du Cheval et de l'Equitation (IFCE), Université de Tours, Nouzilly, France
| | - Giulia Brigante
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Vincenzo Rochira
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Daniele Santi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Livio Casarini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| |
Collapse
|
9
|
Follicle-Stimulating Hormone Treatment and Male Idiopathic Infertility: Effects on Sperm Parameters and Oxidative Stress Indices according to FSHR c. 2039 A/G and c. -29 G/A Genotypes. J Clin Med 2020; 9:jcm9061690. [PMID: 32498268 PMCID: PMC7356235 DOI: 10.3390/jcm9061690] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/23/2022] Open
Abstract
Scientific evidence shows that the administration of follicle-stimulating hormone (FSH) to infertile patients with normal serum FSH concentrations improves sperm parameters in oligozoospermic men. The aim of this study was to evaluate the effects of highly purified urofollitropin (hpFSH) on conventional and bio-functional sperm parameters and on oxidative stress indices in patients with idiopathic infertility. We also evaluated the response to hpFSH on these parameters in relationship to FSHR c. 2039 A/G and FSHR c. -29 G/A genotypes. A prospective longitudinal study was conducted on 42 patients with idiopathic male infertility, 23 of whom underwent to FSHR c. 2039 A/G and FSHR c. -29 G/A genotyping. Each patient was asked to collect two semen samples before and after administration of 150 IU hpFSH three times a week for 16 weeks. Patients were divided into responders or non-responders based on whether their total sperm count had at least doubled or was less than double at the end of treatment, respectively. Responders showed a significantly higher semen volume, sperm concentration, spermatids, and leukocytes. Non-responders had a significant decrease of the percentage of spermatozoa in early apoptosis after hpFSH administration. Oxidative stress indexes did not differ significantly after FSH administration in both groups. Conventional and bio-functional sperm parameters did not differ in patients with FSHR c. 2039 GG and AA genotypes, and FSHR c. -29 GG genotype both before and after FSH administration. The FSHR c. 2039 and FSHR -29 G/A genotypes and allelic distribution did not differ between responders and non-responders. FSH showed to be capable of ameliorating sperm parameters in about half patients treated, therefore it may be helpful in patients with idiopathic infertility.
Collapse
|
10
|
Sabbatini AR, Kararigas G. Estrogen-related mechanisms in sex differences of hypertension and target organ damage. Biol Sex Differ 2020; 11:31. [PMID: 32487164 PMCID: PMC7268741 DOI: 10.1186/s13293-020-00306-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
Hypertension (HTN) is a primary risk factor for cardiovascular (CV) events, target organ damage (TOD), premature death and disability worldwide. The pathophysiology of HTN is complex and influenced by many factors including biological sex. Studies show that the prevalence of HTN is higher among adults aged 60 and over, highlighting the increase of HTN after menopause in women. Estrogen (E2) plays an important role in the development of systemic HTN and TOD, exerting several modulatory effects. The influence of E2 leads to alterations in mechanisms regulating the sympathetic nervous system, renin-angiotensin-aldosterone system, body mass, oxidative stress, endothelial function and salt sensitivity; all associated with a crucial inflammatory state and influenced by genetic factors, ultimately resulting in cardiac, vascular and renal damage in HTN. In the present article, we discuss the role of E2 in mechanisms accounting for the development of HTN and TOD in a sex-specific manner. The identification of targets with therapeutic potential would contribute to the development of more efficient treatments according to individual needs.
Collapse
Affiliation(s)
| | - Georgios Kararigas
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
11
|
Alviggi C, Conforti A, Santi D, Esteves SC, Andersen CY, Humaidan P, Chiodini P, De Placido G, Simoni M. Clinical relevance of genetic variants of gonadotrophins and their receptors in controlled ovarian stimulation: a systematic review and meta-analysis. Hum Reprod Update 2019; 24:599-614. [PMID: 29924306 DOI: 10.1093/humupd/dmy019] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 05/12/2018] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Genotype has been implicated in the outcome of ovarian stimulation. The analysis of patient-specific genotypes might lead to an individualized pharmacogenomic approach to controlled ovarian stimulation (COS). However, the validity of such an approach remains to be established. OBJECTIVE AND RATIONALE To define the impact of specific genotype profiles of follicle-stimulating hormone, luteinizing hormone and their receptors (FSHR, LHR and LHCGR) on ovarian stimulation outcome. Specifically, our aim was to identify polymorphisms that could be useful in clinical practice, and those that need further clinical investigation. SEARCH METHODS A systematic review followed by a meta-analysis was performed according to the Cochrane Collaboration and Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines without time restriction. We searched the PubMed/MEDLINE, Cochrane Library, SCOPUS and EMBASE databases to identify all relevant studies published before January 2017. Only clinical trials published as full-text articles in peer-reviewed journals were included. The primary outcome was the number of oocytes retrieved. OUTCOMES Fifty-seven studies were assessed for eligibility, 33 of which were included in the qualitative and quantitative analyses. Data were independently extracted using quality indicators. COS outcomes related to seven polymorphisms (FSHR [rs6165], FSHR [rs6166], FSHR [rs1394205], LHB [rs1800447], LHB [rs1056917], LHCGR [rs2293275] and LHCGR [rs13405728]) were evaluated. More oocytes were retrieved from FSHR (rs6165) AA homozygotes (five studies, 677 patients, weighted mean difference [WMD]: 1.85, 95% CI: 0.85-2.85, P < 0.001; I2 = 0%) than from GG homozygotes and AG heterozygotes (four studies, 630 patients, WMD: 1.62, 95% CI: 0.28-2.95, P = 0.020; I2 = 56%). Moreover, stimulation duration was shorter in FSHR (rs6165) AA homozygotes than in AG carriers (three studies, 588 patients, WMD -0.48, 95% CI: -0.87 to -0.10, P = 0.010, I2 = 44%). A higher number of oocytes (21 studies, 2632 patients WMD: 0.84, 95% CI: 0.19 to 1.49, P = 0.01, I2 = 76%) and metaphase II oocytes (five studies, 608 patients, WMD: 1.03, 95% CI: 0.01-2.05, P = 0.050, I2 = 0%) was observed in AA than in GG homozygote carriers. FSH consumption was significantly lower in FSHR (rs1394205) GG homozygotes (three studies, 411 patients, WMD: -1294.61 IU, 95% CI: -593.08 to -1996.14 IU, P = 0.0003, I2 = 99%) and AG heterozygotes (three studies, 367 patients, WMD: -1014.36 IU, 95% CI: -364.11 to -1664.61 IU, P = 0.002, I2 = 99%) than in AA homozygotes. WIDER IMPLICATIONS These results support the clinical relevance of specific genotype profiles on reproductive outcome. Further studies are required to determine their application in a pharmacogenomic approach to ovarian stimulation.
Collapse
Affiliation(s)
- Carlo Alviggi
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Italy.,Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Italy
| | - Daniele Santi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, and Azienda Ospedaliera-Universitaria di Modena, Italy
| | - Sandro C Esteves
- Androfert, Andrology and Human Reproduction Clinic, and Department of Surgery (Division of Urology), University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Peter Humaidan
- Fertility Clinic, Skive Regional Hospital, Skive, Denmark, and Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Paolo Chiodini
- Medical Statistics Unit, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe De Placido
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, and Azienda Ospedaliera-Universitaria di Modena, Italy
| |
Collapse
|
12
|
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: 3.7] [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
|
13
|
Čuš M, Vlaisavljević V, Repnik K, Potočnik U, Kovačič B. Could polymorphisms of some hormonal receptor genes, involved in folliculogenesis help in predicting patient response to controlled ovarian stimulation? J Assist Reprod Genet 2019; 36:47-55. [PMID: 30406448 PMCID: PMC6338606 DOI: 10.1007/s10815-018-1357-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/22/2018] [Indexed: 01/21/2023] Open
Abstract
PURPOSE The aim of this study was to investigate whether single nucleotide polymorphisms (SNPs) in selected genes, responsible for hormonal regulation of folliculogenesis, are associated with response to controlled ovarian hyperstimulation (COH) and clinical characteristics of women enrolled in in vitro fertilization (IVF) programs. METHODS In a cross-sectional study, 60 (IVF) patients underwent COH by using gonadotropin-releasing hormone (GnRH) antagonist and recombinant follicle-stimulating hormone (rFSH) protocol. Patients were classified into three groups: poor-responders (according to Bologna criteria), normo-responders (≤ 15 oocytes), and hyper-responders (> 15 oocytes). Genotyping of SNPs AMH rs10407022, AMHR rs3741664, FSHR rs1394205 and rs6166, and ESR1 rs2234693 was performed using high-resolution melting analysis (HRMA). Basal FSH (bFSH), estradiol (E2), and anti-Müllerian hormone (AMH) were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS Patients with GG genotype of FSHR rs1394205 had significantly lower AMH level (P = 0.016) and required higher rFSH dose per oocyte compared to women with AA or AG genotype (P = 0.036). We also found higher frequency of GG genotype of FSHR rs1394205 in poor- (76.5%) than in hyper-responders (37.5%, P = 0.002). Patients with AA genotype of FSHR rs6166 had higher level of measured bFSH compared to those with AG or GG genotypes (P = 0.043). Women with GG genotype of AMHR rs3741664 required higher rFSH dose in comparison with patients carrying genotypes AA or AG (P = 0.028). CONCLUSIONS The GG genotype at position rs1394205 is associated with poor ovarian response to COH. Patients with this genotype may require higher doses of rFSH for ovulation induction.
Collapse
Affiliation(s)
- Maruška Čuš
- Department of Reproductive Medicine and Gynecological Endocrinology, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia
| | | | - Katja Repnik
- Center for human molecular genetics and pharmacogenomics, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty for Chemistry & Chemical Engineering, University of Maribor, Smetanova 17, Maribor, Slovenia
| | - Uroš Potočnik
- Center for human molecular genetics and pharmacogenomics, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty for Chemistry & Chemical Engineering, University of Maribor, Smetanova 17, Maribor, Slovenia
| | - Borut Kovačič
- Department of Reproductive Medicine and Gynecological Endocrinology, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia.
| |
Collapse
|
14
|
Schubert M, Pérez Lanuza L, Gromoll J. Pharmacogenetics of FSH Action in the Male. Front Endocrinol (Lausanne) 2019; 10:47. [PMID: 30873114 PMCID: PMC6403134 DOI: 10.3389/fendo.2019.00047] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/21/2019] [Indexed: 11/28/2022] Open
Abstract
Male infertility is a major contributor to couple infertility, however in most cases it remains "idiopathic" and putative treatment regimens are lacking. This leads to a scenario in which intra-cytoplasmic spermatozoa injection (ICSI) is widely used in idiopathic male infertility, though the treatment burden is high for the couple and it entails considerable costs and risks. Given the crucial role of the Follicle-stimulating hormone (FSH) for spermatogenesis, FSH has been used empirically to improve semen parameters, but the response to FSH varied strongly among treated infertile men. Single nucleotide polymorphisms (SNPs) within FSH ligand/receptor genes (FSHB/FSHR), significantly influencing reproductive parameters in men, represent promising candidates to serve as pharmacogenetic markers to improve prediction of response to FSH. Consequently, several FSH-based pharmacogenetic studies have been conducted within the last years with unfortunately wide divergence concerning selection criteria, treatment and primary endpoints. In this review we therefore outline the current knowledge on single nucleotide polymorphisms (SNPs) in the FSH and FSH receptor genes and their putative functional effects. We compile and critically assess the previously performed pharmacogenetic studies in the male and propose a putative strategy that might allow identifying patients who could benefit from FSH treatment.
Collapse
Affiliation(s)
- Maria Schubert
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Lina Pérez Lanuza
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
| | - Jörg Gromoll
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, Münster, Germany
- *Correspondence: Jörg Gromoll
| |
Collapse
|
15
|
Pharmacogenetic study of Asn680Ser and -29A>G in FSHR gene in Chinese women undergoing controlled ovarian hyperstimulation. JOURNAL OF BIO-X RESEARCH 2018. [DOI: 10.1097/jbr.0000000000000018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
16
|
García-Jiménez G, Zariñán T, Rodríguez-Valentín R, Mejía-Domínguez NR, Gutiérrez-Sagal R, Hernández-Montes G, Tovar A, Arechavaleta-Velasco F, Canto P, Granados J, Moreno-Macias H, Tusié-Luna T, Pellicer A, Ulloa-Aguirre A. Frequency of the T307A, N680S, and -29G>A single-nucleotide polymorphisms in the follicle-stimulating hormone receptor in Mexican subjects of Hispanic ancestry. Reprod Biol Endocrinol 2018; 16:100. [PMID: 30340493 PMCID: PMC6195735 DOI: 10.1186/s12958-018-0420-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/09/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND FSHR SNPs may influence the ovarian sensitivity to endogenous and exogenous FSH stimulation. Given the paucity of data on the FSHR c.919A > G, c.2039A > G and - 29G > A SNPs in Hispanic population, we here analyzed their frequency distribution in Mexican mestizo women. METHODS Samples from 224 Mexican mestizo women enrolled in an IVF program as well as a genotype database from 8182 Mexican mestizo subjects, were analyzed for FSHR SNPs at positions c.919, c.2039 and - 29G > A. Association between the genetic variants and reproductive outcomes was assessed. RESULTS The c.919 and c.2039 SNPs were in strong linkage disequilibrium and their corresponding genotype frequencies in the IVF group were: AA 46.8%, AG 44.2%, and GG 8.9%, and AA 41.9%, AG 48.2% and GG 9.8%, respectively. For the -29G > A SNP, genotype frequencies were 27% (GG), 50% (GA) and 23% (AA). In normal oocyte donors with the c.2039 GG genotype, the number of oocytes recovered after ovarian stimulation (COS) were significantly (p < 0.01) lower than in those bearing other genotypes in this or the -29G > A SNP. Analysis of the large scale database revealed that both allelic and genotype frequencies for the three SNPs were very similar to those detected in the IVF cohort (p ≥ 0.38) and that female carriers of the c.2039 G allele tended to present lower number of pregnancies than women bearing the AA genotype; this trend was stronger when women with more Native American ancestry was separately analyzed (OR = 2.0, C.I. 95% 1.03-3.90, p = 0.04). There were no differences or trends in the number of pregnancies among the different genotypes of the -29G > A SNP. CONCLUSIONS The frequency of the GG/GG combination genotype for the c.919 and c.2039 SNPs in Mexican hispanics is among the lowest reported. The GG genotype is associated with decreased number of oocytes recovered in response to COS as well as to lower pregnancy rates in Hispanic women from the general population. The absence of any effect of the -29AA genotype on the response to COS, indicates that there is no need to perform this particular genotype testing in Hispanic women with the purpose of providing an individually-tailored COS protocol.
Collapse
Affiliation(s)
- Gabriela García-Jiménez
- Instituto Valenciano de Infertilidad (IVINSEMER), Thiers 96, Col. Nueva Anzurez, CP 11590 Mexico City, Mexico
| | - Teresa Zariñán
- 0000 0001 2159 0001grid.9486.3Red de Apoyo a la Investigación (RAI), Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición SZ (INCMNSZ), Calle Vasco de Quiroga 15, Tlalpan, 14000 Ciudad de Mexico, Mexico
| | - Rocío Rodríguez-Valentín
- 0000 0004 1773 4764grid.415771.1Centro de Investigación en Salud Poblacional, Instituto Nacional de Salud Pública, Av. Universidad 655, CP 62100 Cuernavaca, Mor Mexico
| | - Nancy R. Mejía-Domínguez
- 0000 0001 2159 0001grid.9486.3Red de Apoyo a la Investigación (RAI), Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición SZ (INCMNSZ), Calle Vasco de Quiroga 15, Tlalpan, 14000 Ciudad de Mexico, Mexico
| | - Rubén Gutiérrez-Sagal
- 0000 0001 2159 0001grid.9486.3Red de Apoyo a la Investigación (RAI), Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición SZ (INCMNSZ), Calle Vasco de Quiroga 15, Tlalpan, 14000 Ciudad de Mexico, Mexico
| | - Georgina Hernández-Montes
- 0000 0001 2159 0001grid.9486.3Red de Apoyo a la Investigación (RAI), Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición SZ (INCMNSZ), Calle Vasco de Quiroga 15, Tlalpan, 14000 Ciudad de Mexico, Mexico
| | - Armando Tovar
- 0000 0001 0698 4037grid.416850.eDepartment of Physiology of Nutrition, INCMNSZ, Mexico City, Mexico
| | - Fabián Arechavaleta-Velasco
- Research Unit in Reproductive Medicine, UMAE Hospital de Ginecoobstetricia “Luis Castelazo Ayala”, Río de la Magdalena 289, Tizapán San Ángel, Mexico City, 01090 Mexico
| | - Patricia Canto
- 0000 0001 2159 0001grid.9486.3Facultad de Medicina, UNAM, Ciudad Universitaria, Mexico City, Mexico
| | - Julio Granados
- 0000 0001 0698 4037grid.416850.eDepartment of Transplantation, INCMNSZ, Mexico City, Mexico
| | - Hortensia Moreno-Macias
- 0000 0001 2157 0393grid.7220.7Universidad Autónoma Metropolitana-Iztapalapa, Av. San Rafael Atlixco 186, Leyes de Reforma 1ra. Secc., Mexico City, 09340 Mexico
| | - Teresa Tusié-Luna
- Unidad de Biología Molecular y Medicina Genómica, UNAM-INCMNSZ, Mexico City, Mexico
| | - Antonio Pellicer
- Instituto Valenciano de Infertilidad (IVINSEMER), Thiers 96, Col. Nueva Anzurez, CP 11590 Mexico City, Mexico
| | - Alfredo Ulloa-Aguirre
- 0000 0001 2159 0001grid.9486.3Red de Apoyo a la Investigación (RAI), Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición SZ (INCMNSZ), Calle Vasco de Quiroga 15, Tlalpan, 14000 Ciudad de Mexico, Mexico
| |
Collapse
|
17
|
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: 5] [Impact Index Per Article: 0.7] [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
|
18
|
Leutinizing hormone/choriogonadotropin receptor and follicle stimulating hormone receptor gene variants and risk of recurrent pregnancy loss: A case control study. Meta Gene 2018. [DOI: 10.1016/j.mgene.2017.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
19
|
Tamburino L, La Vignera S, Tomaselli V, Condorelli RA, Cannarella R, Mongioì LM, Calogero AE. The -29G/A FSH receptor gene polymorphism is associated with higher FSH and LH levels in normozoospermic men. J Assist Reprod Genet 2017. [PMID: 28624859 DOI: 10.1007/s10815-017-0970-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
PURPOSE The functional role of the FSHR promoter -29G/A polymorphism (rs1394205) in men is not clear. Some studies failed to find a relationship between the FSHR -29G/A and follicle-stimulating hormone (FSH) levels and did not associate the SNP with male infertility. Only one study showed that the FSHR -29 SNP modulates serum FSH levels in Baltic young male cohort. Because the SNP -29G/A has to be shown to have a strong effect on in vitro transcription activity of the FSHR promoter and the activation of FSHR is necessary for a normal FSH function, this study was undertaken to assess whether the FSHR -29G/A SNP modulates the gonadal endocrine function in men. METHODS A total of 200 men with alteration of conventional sperm parameters or normozoospermia (according to the parameters WHO 2010), were genotyped by TaqMan Assay. Hormone levels were measured by immunoassay, and sperm analysis was performed according to the World Health Organization criteria. RESULTS A significant gradient of increasing FSH levels across the FSHR -29G/A genotypes was observed (p < 0.01). Among normozoospermic men (n = 110), those with FSHR -29A-allele carriers (GA + AA and AA) had higher serum FSH (p < 0.01) and LH levels (p < 0.05) and higher body mass index (BMI) (p < 0.01) compared to men with the GG genotype. The carrier status of rs1394205 genotypes did not affect the other endocrine parameters neither in men with altered sperm parameters nor in normozoospermic men. CONCLUSIONS The FSHR -29G/A polymorphism modulates FSH and, for the first time, LH serum levels and BMI in normozoospermic men. These findings underline the importance to pay close attention to the studies of genetic variations associated with clinical-endocrine parameters.
Collapse
Affiliation(s)
- L Tamburino
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", Via S. Sofia 78, Bldg 4, Rm 2C17, 95123, Catania, Italy
| | - S La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", Via S. Sofia 78, Bldg 4, Rm 2C17, 95123, Catania, Italy.
| | - V Tomaselli
- Department of Political and Social Sciences, University of Catania, Via Vittorio Emanuele 49, 95100, Catania, Italy
| | - R A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", Via S. Sofia 78, Bldg 4, Rm 2C17, 95123, Catania, Italy
| | - R Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", Via S. Sofia 78, Bldg 4, Rm 2C17, 95123, Catania, Italy
| | - L M Mongioì
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", Via S. Sofia 78, Bldg 4, Rm 2C17, 95123, Catania, Italy
| | - A E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Policlinico "G. Rodolico", Via S. Sofia 78, Bldg 4, Rm 2C17, 95123, Catania, Italy
| |
Collapse
|
20
|
Borgbo T, Klučková H, Macek M, Chrudimska J, Kristensen SG, Hansen LL, Andersen CY. The Common Follicle-Stimulating Hormone Receptor (FSHR) Promoter Polymorphism FSHR -29G > A Affects Androgen Production in Normal Human Small Antral Follicles. Front Endocrinol (Lausanne) 2017; 8:122. [PMID: 28626448 PMCID: PMC5454067 DOI: 10.3389/fendo.2017.00122] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Follicle-stimulating hormone receptors (FSHRs) are almost exclusively expressed on granulosa cells, and FSH action is probably most clearly reflected in intrafollicular hormone milieu of antral follicles. Little is known about the possible effects of the common single nucleotide polymorphism (SNP) FSHR -29G > A (rs1394205) on hormonal conditions in humsan small antral follicles (hSAFs) obtained from women in the natural menstrual cycle. This study investigated the follicle fluid (FF) concentrations of anti-Müllerian hormone, estradiol, progesterone, androstenedione, and testosterone in hSAF in relation to the different genotypes of FSHR -29G > A. FF from 362 follicles was collected in 95 women undergoing fertility preservation, who did not suffer from a disease that directly affected ovarian function. The testosterone levels of the minor A/A genotype were significantly increased compared to the A/G and the G/G genotype. Furthermore, significantly reduced androstenedione levels were observed for the G/G genotype, as compared to the A/G genotype, while the other hormones did not show statistical significant differences. In conclusion, the androgen levels of hSAF were significantly elevated in the minor SNP genotype in the FSHR promoter polymorphism FSHR -29G > A.
Collapse
Affiliation(s)
- Tanni Borgbo
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Hana Klučková
- Department of Biology and Medical Genetics, 2nd Faculty of Medicine Charles University, University Hospital Motol, Prague, Czechia
| | - Milan Macek
- Department of Biology and Medical Genetics, 2nd Faculty of Medicine Charles University, University Hospital Motol, Prague, Czechia
| | - Jana Chrudimska
- Department of Biology and Medical Genetics, 2nd Faculty of Medicine Charles University, University Hospital Motol, Prague, Czechia
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
| | | | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
- *Correspondence: Claus Yding Andersen,
| |
Collapse
|
21
|
Effect of Follicle Stimulating Hormone Receptor Gene Polymorphisms in Cervical Cancer Risk. Pathol Oncol Res 2016; 23:565-572. [DOI: 10.1007/s12253-016-0152-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 11/09/2016] [Indexed: 02/03/2023]
|
22
|
Association of a promoter polymorphism in FSHR with ovarian reserve and response to ovarian stimulation in women undergoing assisted reproductive treatment. Reprod Biomed Online 2016; 33:391-7. [PMID: 27448492 DOI: 10.1016/j.rbmo.2016.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 06/01/2016] [Accepted: 06/07/2016] [Indexed: 11/23/2022]
Abstract
Previous studies have suggested an association between a variant in the promoter region of the FSHR gene and diminished response to controlled ovarian hyperstimulation (COH) in women undergoing assisted reproduction. FSHR -29G>A was genotyped in 559 women undergoing their first cycle of COH for IVF/intracytoplasmic sperm injection (ICSI) using TaqMan allelic discrimination assay. Correlation and regression analysis was performed to assess the relationship between FSHR promoter genotypes and markers of ovarian reserve and measures of response to COH, including the number of oocytes retrieved, gonadotrophin dose used and the live-birth rate. There were no statistically significant differences between the genotype frequencies and the markers of ovarian reserve or the early measures of response to COH. However, the live-birth rate was higher for women carrying the variant A allele (odds ratio [OR] 1.37; 95% confidence interval [CI] 1.02-1.84 per allele). This relationship did not reach statistical significance after adjustment for the number of embryos transferred (OR 1.33; 95% CI 0.98-1.83 per allele). Results from this study do not provide evidence that the FSHR -29G>A variant can be used in the individualization of the treatment protocol for women undergoing IVF/ICSI.
Collapse
|
23
|
Busch AS, Hagen CP, Almstrup K, Main KM, Juul A. Genetic variations altering FSH action affect circulating hormone levels as well as follicle growth in healthy peripubertal girls. Hum Reprod 2016; 31:897-904. [PMID: 26905078 DOI: 10.1093/humrep/dew022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 01/22/2016] [Indexed: 02/07/2023] Open
Abstract
STUDY QUESTION Do variants of the genes encoding follicle stimulating hormone (FSH) beta subunit (B) and FSH receptor (R) impact circulating reproductive hormone levels and ovarian follicle maturation in healthy peripubertal girls? SUMMARY ANSWER FSHB and FSHR genetic variants exert, alone or their combination, distinct effects on reproductive hormone levels as well as ovarian follicle maturation in healthy peripubertal girls. WHAT IS KNOWN ALREADY FSHB and FSHR genetic variants impact reproductive hormone levels as well as associated pathologies in women. While FSHR c. 2039A>G is known to alter gonadotrophin levels in women, FSHR c.-29G>A has not yet been shown to exert effect and there are conflicting results concerning FSHB c.-211G>T. STUDY DESIGN, SIZE, DURATION This population-based study included 633 girls recruited as part of two cohorts, the COPENHAGEN Puberty Study (2006-2014, a cross-sectional and ongoing longitudinal study) and the Copenhagen Mother-Child Cohort (1997-2002, including transabdominal ultrasound (TAUS) of the ovaries in a subset of 91 peripubertal girls). PARTICIPANTS/MATERIALS, SETTING, METHODS Clinical examinations, including pubertal breast stage (Tanner's classification B1-B5) were performed. Circulating levels of FSH, luteinizing hormone (LH), estradiol, anti-Mullerian hormone (AMH) and inhibin-B were assessed by immunoassays. In a subset of the girls (n = 91), ovarian volume and the number/size of antral follicles were assessed by TAUS. Genotypes were determined by competitive PCR. MAIN RESULTS AND THE ROLE OF CHANCE FSHR c.2039A>G minor alleles were positively associated with serum FSH (β = 0.08, P = 0.004), LH (β = 0.06, P = 0.012) and estradiol (β = 0.06, P = 0.017) (adjusted for Tanner stages). In a combined model, FSHR c.-29G>A and FSHR c.2039A>G alleles were positively associated with FSH levels in early-pubertal girls (B2 + B3, n = 327, r = 0.1, P = 0.02) and in young adolescents (B4 + B5, n = 149, r = 0.2, P = 0.01). Serum AMH and inhibin B levels were not significantly influenced by the single nucleotide polymorphisms (SNPs). Single SNPs were not associated with follicles counts, however, a cumulative minor allele count (FSHB c.-211 G>T and FSHR c.-29G>A) was negatively associated with the number of large follicles (≥5 mm) (n = 91, P = 0.04) (adjusted for Tanner stages). LIMITATIONS, REASONS FOR CAUTION Since we studied girls and young adolescents during pubertal transition, our study may not be fully comparable with previous studies on FSHB and FSHR variants in adult women. The group of young adolescents (Tanner B4 + B5) reflects the endocrine situation in adult women best, however, the group is not large enough to contribute substantially to the conflicting results concerning the influence of FSHB c.-211G>T in adult women. Furthermore, we have no information about the exact day of the menstrual cycle in the subgroup of girls with menarche. WIDER IMPLICATIONS OF THE FINDINGS The sex-specific interaction of FSHB and FSHR genetic variants and physiological as well as pathological conditions is being increasingly elucidated. The variant triplet set might serve as diagnostic and pharmacogenetic marker. For the first time, we show an additional effect of FSHR c.-29G>A on serum FSH levels in healthy girls. Moreover, morphological data suggest impaired FSH-induced maturation of ovarian follicles in minor allele carriers of FSHB c.-211G>T and FSHR c.-29G>A. This may explain previous findings of delayed pubertal onset in these girls. STUDY FUNDING/COMPETING INTERESTS Funding was provided by the Danish Agency for Science, Technology and Innovation (09-067180), Danish Ministry of the Environment, CeHoS (MST-621-00065), Capital Region of Denmark (December 2011), Ministry of Higher Education and Science (DFF-1331-00113) and EDMaRC (Danish Ministry of Health). A.S.B. was funded from December 2015 by ReproUnion (EU Interreg Öresund-Kattegat-Skagerrak). The authors declare no conflict of interest.
Collapse
Affiliation(s)
- Alexander S Busch
- Department of Growth and Reproduction and EDMaRC, Rigshospitalet, Section 5064, University of Copenhagen, Blegdamsvej 9, DK-2100, Copenhagen O, Denmark
| | - Casper P Hagen
- Department of Growth and Reproduction and EDMaRC, Rigshospitalet, Section 5064, University of Copenhagen, Blegdamsvej 9, DK-2100, Copenhagen O, Denmark
| | - Kristian Almstrup
- Department of Growth and Reproduction and EDMaRC, Rigshospitalet, Section 5064, University of Copenhagen, Blegdamsvej 9, DK-2100, Copenhagen O, Denmark
| | - Katharina M Main
- Department of Growth and Reproduction and EDMaRC, Rigshospitalet, Section 5064, University of Copenhagen, Blegdamsvej 9, DK-2100, Copenhagen O, Denmark
| | - Anders Juul
- Department of Growth and Reproduction and EDMaRC, Rigshospitalet, Section 5064, University of Copenhagen, Blegdamsvej 9, DK-2100, Copenhagen O, Denmark
| |
Collapse
|
24
|
Casarini L, Santi D, Marino M. Impact of gene polymorphisms of gonadotropins and their receptors on human reproductive success. Reproduction 2015; 150:R175-84. [PMID: 26370242 DOI: 10.1530/rep-15-0251] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2015] [Indexed: 12/17/2022]
Abstract
Gonadotropins and their receptors' genes carry several single-nucleotide polymorphisms resulting in endocrine genotypes modulating reproductive parameters, diseases, and lifespan leading to important implications for reproductive success and potential relevance during human evolution. Here we illustrate common genotypes of the gonadotropins and gonadotropin receptors' genes and their clinical implications in phenotypes relevant for reproduction such as ovarian cycle length, age of menopause, testosterone levels, polycystic ovary syndrome, and cancer. We then discuss their possible role in human reproduction and adaptation to the environment. Gonadotropins and their receptors' variants are differently distributed among human populations. Some hints suggest that they may be the result of natural selection that occurred in ancient times, increasing the individual chance of successful mating, pregnancy, and effective post-natal parental cares. The gender-related differences in the regulation of the reproductive endocrine systems imply that many of these genotypes may lead to sex-dependent effects, increasing the chance of mating and reproductive success in one sex at the expenses of the other sex. Also, we suggest that sexual conflicts within the FSH and LH-choriogonadotropin receptor genes contributed to maintain genotypes linked to subfertility among humans. Because the distribution of polymorphic markers results in a defined geographical pattern due to human migrations rather than natural selection, these polymorphisms may have had only a weak impact on reproductive success. On the contrary, such genotypes could acquire relevant consequences in the modern, developed societies in which parenthood attempts often occur at a later age, during a short, suboptimal reproductive window, making clinical fertility treatments necessary.
Collapse
Affiliation(s)
- Livio Casarini
- Unit of EndocrinologyDepartment of Biomedical, Metabolic and Neural SciencesCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Via G. Campi, 287, 41125 Modena, ItalyAzienda USL of ModenaNOCSAE, Via P. Giardini 1355, 41126 Modena, Italy Unit of EndocrinologyDepartment of Biomedical, Metabolic and Neural SciencesCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Via G. Campi, 287, 41125 Modena, ItalyAzienda USL of ModenaNOCSAE, Via P. Giardini 1355, 41126 Modena, Italy
| | - Daniele Santi
- Unit of EndocrinologyDepartment of Biomedical, Metabolic and Neural SciencesCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Via G. Campi, 287, 41125 Modena, ItalyAzienda USL of ModenaNOCSAE, Via P. Giardini 1355, 41126 Modena, Italy Unit of EndocrinologyDepartment of Biomedical, Metabolic and Neural SciencesCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Via G. Campi, 287, 41125 Modena, ItalyAzienda USL of ModenaNOCSAE, Via P. Giardini 1355, 41126 Modena, Italy
| | - Marco Marino
- Unit of EndocrinologyDepartment of Biomedical, Metabolic and Neural SciencesCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Via G. Campi, 287, 41125 Modena, ItalyAzienda USL of ModenaNOCSAE, Via P. Giardini 1355, 41126 Modena, Italy Unit of EndocrinologyDepartment of Biomedical, Metabolic and Neural SciencesCenter for Genomic ResearchUniversity of Modena and Reggio Emilia, Via G. Campi, 287, 41125 Modena, ItalyAzienda USL of ModenaNOCSAE, Via P. Giardini 1355, 41126 Modena, Italy
| |
Collapse
|
25
|
Borgbo T, Jeppesen J, Lindgren I, Lundberg Giwercman Y, Hansen L, Yding Andersen C. Effect of the FSH receptor single nucleotide polymorphisms (FSHR 307/680) on the follicular fluid hormone profile and the granulosa cell gene expression in human small antral follicles. ACTA ACUST UNITED AC 2014; 21:255-61. [DOI: 10.1093/molehr/gau106] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
26
|
Hagen CP, Sørensen K, Aksglaede L, Mouritsen A, Mieritz MG, Tinggaard J, Wohlfart-Veje C, Petersen JH, Main KM, Rajpert-De Meyts E, Almstrup K, Juul A. Pubertal onset in girls is strongly influenced by genetic variation affecting FSH action. Sci Rep 2014; 4:6412. [PMID: 25231187 PMCID: PMC4166707 DOI: 10.1038/srep06412] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/28/2014] [Indexed: 01/30/2023] Open
Abstract
Age at pubertal onset varies substantially in healthy girls. Although genetic factors are responsible for more than half of the phenotypic variation, only a small part has been attributed to specific genetic polymorphisms identified so far. Follicle-stimulating hormone (FSH) stimulates ovarian follicle maturation and estradiol synthesis which is responsible for breast development. We assessed the effect of three polymorphisms influencing FSH action on age at breast deveopment in a population-based cohort of 964 healthy girls. Girls homozygous for FSHR -29AA (reduced FSH receptor expression) entered puberty 7.4 (2.5–12.4) months later than carriers of the common variants FSHR -29GG+GA, p = 0.003. To our knowledge, this is the strongest genetic effect on age at pubertal onset in girls published to date.
Collapse
Affiliation(s)
- Casper P Hagen
- Department of Growth and Reproduction, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Kaspar Sørensen
- Department of Growth and Reproduction, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Lise Aksglaede
- Department of Growth and Reproduction, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Annette Mouritsen
- Department of Growth and Reproduction, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Mikkel G Mieritz
- Department of Growth and Reproduction, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jeanette Tinggaard
- Department of Growth and Reproduction, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Christine Wohlfart-Veje
- Department of Growth and Reproduction, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jørgen Holm Petersen
- Department of Growth and Reproduction, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Katharina M Main
- Department of Growth and Reproduction, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- Department of Growth and Reproduction, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Kristian Almstrup
- Department of Growth and Reproduction, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
27
|
Grigorova M, Punab M, Punab AM, Poolamets O, Vihljajev V, Žilaitienė B, Erenpreiss J, Matulevičius V, Laan M. Reproductive physiology in young men is cumulatively affected by FSH-action modulating genetic variants: FSHR -29G/A and c.2039 A/G, FSHB -211G/T. PLoS One 2014; 9:e94244. [PMID: 24718625 PMCID: PMC3981791 DOI: 10.1371/journal.pone.0094244] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 03/14/2014] [Indexed: 12/15/2022] Open
Abstract
Follicle-Stimulating Hormone Receptor (FSHR) -29G/A polymorphism (rs1394205) was reported to modulate gene expression and reproductive parameters in women, but data in men is limited. We aimed to bring evidence to the effect of FSHR -29G/A variants in men. In Baltic young male cohort (n = 982; Estonians, Latvians, Lithuanians; aged 20.2±2.0 years), the FSHR -29 A-allele was significantly associated with higher serum FSH (linear regression: effect 0.27 IU/L; P = 0.0019, resistant to Bonferroni correction for multiple testing) and showed a non-significant trend for association with higher LH (0.19 IU/L) and total testosterone (0.93 nmol/L), but reduced Inhibin B (−7.84 pg/mL) and total testes volume (effect −1.00 mL). Next, we extended the study and tested the effect of FSHR gene haplotypes determined by the allelic combination of FSHR -29G/A and a well-studied variant c.2039 A/G (Asn680Ser, exon 10). Among the FSHR -29A/2039G haplotype carriers (A-Ser; haplotype-based linear regression), this genetic effect was enhanced for FSH (effect 0.40 IU/L), Inhibin B (−16.57 pg/mL) and total testes volume (−2.34 mL). Finally, we estimated the total contribution of three known FSH-action modulating SNPs (FSHB -211G/T; FSHR -29G/A, c.2039 A/G) to phenotypic variance in reproductive parameters among young men. The major FSH-action modulating SNPs explained together 2.3%, 1.4%, 1.0 and 1.1% of the measured variance in serum FSH, Inhibin B, testosterone and total testes volume, respectively. In contrast to the young male cohort, neither FSHR -29G/A nor FSHR haplotypes appeared to systematically modulate the reproductive physiology of oligozoospermic idiopathic infertile patients (n = 641, Estonians; aged 31.5±6.0 years). In summary, this is the first study showing the significant effect of FSHR -29G/A on male serum FSH level. To account for the genetic effect of known common polymorphisms modulating FSH-action, we suggest haplotype-based analysis of FSHR SNPs (FSHR -29G/A, c.2039 A/G) in combination with FSHB -211G/T testing.
Collapse
Affiliation(s)
- Marina Grigorova
- Human Molecular Genetics Research Group, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Margus Punab
- Andrology Unit, Tartu University Hospital, Tartu, Estonia
| | - Anna Maria Punab
- Human Molecular Genetics Research Group, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Olev Poolamets
- Andrology Unit, Tartu University Hospital, Tartu, Estonia
| | | | - Birutė Žilaitienė
- Institute of Endocrinology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | - Valentinas Matulevičius
- Institute of Endocrinology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Maris Laan
- Human Molecular Genetics Research Group, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- * E-mail:
| |
Collapse
|
28
|
Abstract
OBJECTIVE To assess the pharmacogenetic potential of FSH for infertility treatment. DESIGN Review of the literature and genomic databases. METHODS Single-nucleotide polymorphism (SNP) assessed: rs6166 (c.2039A>G, p.N680S), rs6165 (c.919A>G, p.T307A), rs1394205 (c.-29G>A) in FSHR, and rs10835638 (c.-211G>T) in FSHB. Literature search via PubMed. Blast analysis of genomic information available in the NCBI nucleotide database. Comparison of allele frequency and haplotype distribution using the http://spsmart.cesga.estool. RESULTS All these SNPs appear first in Homo, result in reduced FSH action, and are present with variable frequencies and combinations worldwide. Stringent clinical studies demonstrate that the FSHR genotype influences serum FSH levels and gonadal response in both sexes. Serum FSH levels depend on the -211G>T SNP, influencing transcriptional activity of the FSHB promoter. Genotypes reducing FSH action are overrepresented in infertile subjects. CONCLUSIONS Although the clinical relevance of the FSHR polymorphisms alone is limited, the combination of FSHR and FSHB genotypes has a much stronger impact than either one alone in both sexes. About 20% of people are carriers of the alleles associated with lower serum FSH levels/reduced FSHR expression or activity, possibly less favorable for reproduction. Prospective studies need to investigate whether stratification of infertile patients according to their FSHR-FSHB genotypes improves clinical efficacy of FSH treatment compared with the current, naïve approach. A relative enrichment of less favorable FSHR-FSHB genotypes may be related to changes in human reproductive strategies and be a marker of some health-related advantage at the cost of reduced fertility.
Collapse
Affiliation(s)
- Manuela Simoni
- Unit of Endocrinology, NOCSAE, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Pietro Giardini 1355, I- 41126 Modena, Italy
| | | |
Collapse
|
29
|
Desai SS, Roy BS, Mahale SD. Mutations and polymorphisms in FSH receptor: functional implications in human reproduction. Reproduction 2013; 146:R235-48. [PMID: 24051057 DOI: 10.1530/rep-13-0351] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
FSH brings about its physiological actions by activating a specific receptor located on target cells. Normal functioning of the FSH receptor (FSHR) is crucial for follicular development and estradiol production in females and for the regulation of Sertoli cell function and spermatogenesis in males. In the last two decades, the number of inactivating and activating mutations, single nucleotide polymorphisms, and spliced variants of FSHR gene has been identified in selected infertile cases. Information on genotype-phenotype correlation and in vitro functional characterization of the mutants has helped in understanding the possible genetic cause for female infertility in affected individuals. The information is also being used to dissect various extracellular and intracellular events involved in hormone-receptor interaction by studying the differences in the properties of the mutant receptor when compared with WT receptor. Studies on polymorphisms in the FSHR gene have shown variability in clinical outcome among women treated with FSH. These observations are being explored to develop molecular markers to predict the optimum dose of FSH required for controlled ovarian hyperstimulation. Pharmacogenetics is an emerging field in this area that aims at designing individual treatment protocols for reproductive abnormalities based on FSHR gene polymorphisms. The present review discusses the current knowledge of various genetic alterations in FSHR and their impact on receptor function in the female reproductive system.
Collapse
|
30
|
Patnaik M, Pati P, Swain SN, Mohapatra MK, Dwibedi B, Kar SK, Ranjit M. Association of angiotensin-converting enzyme and angiotensin-converting enzyme-2 gene polymorphisms with essential hypertension in the population of Odisha, India. Ann Hum Biol 2013; 41:145-52. [PMID: 24112034 DOI: 10.3109/03014460.2013.837195] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Hypertension is a serious health issue worldwide and essential hypertension, which includes 90-95% of the cases, is influenced by both genetic and environmental factors. Identification of these factors may help in control of this disease. The Insertion/Deletion (I/D) polymorphism in Angiotensin-Converting Enzyme (ACE) gene and rs2106809 (C > T) polymorphism in Angiotensin-Converting Enzyme 2 (ACE2) gene have been reported to be associated with essential hypertension in different populations. AIM To investigate the association of ACE I/D and ACE2 rs2106809 polymorphisms with essential hypertension in the population of Odisha, an eastern Indian state. SUBJECTS AND METHODS A total of 246 hypertensives (159 males and 87 females) and 274 normotensives (158 males and 116 females) were enrolled in the study. Detailed anthropometric data, tobacco, alcohol and food habits were recorded and 2 ml of venous blood was collected for biochemical and genetic analysis. RESULTS The DD genotype of ACE and TT genotype of ACE2 were significantly high among female hypertensives, while T allele of ACE2 was linked to male hypertensives. In the male population, alcohol was also identified as a potential risk factor. CONCLUSION Among females, ACE I/D and ACE2 rs2106809 polymorphisms, while among males, ACE2 rs2106809 polymorphism and alcohol consumption are associated with essential hypertension in the study population.
Collapse
Affiliation(s)
- Manisha Patnaik
- Department of Molecular Biology, Regional Medical Research Centre , Bhubaneswar , India
| | | | | | | | | | | | | |
Collapse
|
31
|
Association of allelic combinations of FSHR gene polymorphisms with ovarian response. Reprod Biomed Online 2013; 27:400-6. [DOI: 10.1016/j.rbmo.2013.07.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 07/08/2013] [Accepted: 07/09/2013] [Indexed: 11/24/2022]
|
32
|
Hoffman MJ, Flister MJ, Nunez L, Xiao B, Greene AS, Jacob HJ, Moreno C. Female-specific hypertension loci on rat chromosome 13. Hypertension 2013; 62:557-63. [PMID: 23817491 DOI: 10.1161/hypertensionaha.113.01708] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A 3.7-Mb region of rat chromosome 13 (45.2-49.0 Mb) affects blood pressure (BP) in females only, indicating the presence of sex-specific BP loci in close proximity to the Renin locus. In the present study, we used a series of Dahl salt-sensitive/Mcwi-13 Brown Norway congenic rat strains to further resolve BP loci within this region. We identified 3 BP loci affecting female rats only, of which the 2 smaller loci (line9BP3 and line9BP4) were functionally characterized by sequence and expression analysis. Compared with SS (SS/HsdMcwiCrl), the presence of a 591-kb region of BN (BN/NHsdMcwi) chromosome 13 (line9BP3) significantly lowered BP by 21 mm Hg on an 8% NaCl diet (153 ± 7 versus 174 ± 5 mm Hg; P<0.001). Unexpectedly, the addition of 23 kb of Brown Norway chromosome 13 (line9BP4) completely erased the female-specific BP protection on 8% NaCl diet, suggesting that BN hypertensive allele(s) reside in this region. The congenic interval of the protective line 9F strain contains 3 genes (Optc, Prelp, and Fmod), and the hypertensive line 9E contains 1 additional gene (Btg2). Sequence analysis of the 2 BP loci revealed a total of 282 intergenic variants, with no coding variants. Analysis of gene expression by quantitative real-time polymerase chain reaction revealed strain- and sex-specific differences in Prelp, Fmod, and Btg2 expression, implicating these as novel candidate genes for female-specific hypertension.
Collapse
Affiliation(s)
- Matthew J Hoffman
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Polymorphisms in gonadotropin and gonadotropin receptor genes as markers of ovarian reserve and response in in vitro fertilization. Fertil Steril 2013; 99:970-8.e1. [PMID: 23380184 DOI: 10.1016/j.fertnstert.2013.01.086] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 01/04/2013] [Accepted: 01/04/2013] [Indexed: 11/22/2022]
Abstract
Since gonadotropins are the fundamental hormones that control ovarian activity, genetic polymorphisms may alter gonadal responsiveness to glycoproteins; hence they are important regulators of hormone activity at the target level. The establishment of the pool of primordial follicles takes place during fetal life and is mainly under genetic control. Consequently, single nucleotide polymorphisms (SNPs) in gonadotropins and their receptors do not seem to be associated with any significant modification in the endowment of nongrowing follicles in the ovary. Indeed, the age at menopause, a biological characteristic strongly related to ovarian reserve, as well as markers of functional ovarian reserve such as anti-Müllerian hormone and antral follicle count, are not different in women with different genetic variants. Conversely, some polymorphisms in FSH receptor (FSHR) seem to be associated with modifications in ovarian activity. In particular, studies suggest that the Ser680 genotype for FSHR is a factor of relative resistance to FSH stimulation resulting in slightly higher FSH serum levels, thus leading to a prolonged duration of the menstrual cycle. Moreover, some FSHR gene polymorphisms show a positive association with ovarian response to exogenous gonadotropin administration, hence exhibiting some potential for a pharmacogenetic estimation of the FSH dosage in controlled ovarian stimulation. The study of SNPs of the FSHR gene is an interesting field of research that could provide us with new information about the way each woman responds to exogenous gonadotropin administration during ovulation induction.
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW To review the current knowledge of genetic variants in the two genes affecting the individual responsiveness to follicle-stimulating hormone (FSH) action-the FSH beta-subunit (FSHB) and the FSH receptor (FSHR), as well as the pharmacogenetic ramifications of the findings. RECENT FINDINGS Four common variants in the FSHB and the FSHR genes were shown to exhibit significant effect on FSH action: linked FSHR variants Thr307Ala and Asn680Ser determining common receptor isoforms, and gene expression affecting polymorphisms FSHR -29G/A and FSHB -211G/T. In women, the FSHR Thr307Ala/Asn680Ser polymorphisms show consistent predictive value for estimating the most optimal recombinant FSH dosage in controlled ovarian hyperstimulation (COH). The same variants exhibit a potential for the pharmacogenetic assessment of the treatment of polycystic ovarian syndrome. The FSHR -29G/A variant was also shown to contribute to ovarian response to COH. Pilot studies have suggested the FSHB -211 TT homozygous oligozoospermic men with genetically determined low concentration of FSH, as potentially the best responders to FSH treatment; furthermore, modulation of this response by FSHR polymorphisms is possible. SUMMARY Genetic variants in FSHB and FSHR exhibit a potential for pharmacogenetic applications in selecting appropriate treatment options (timing and dosage) in male and female conditions requiring or benefiting from FSH therapy.
Collapse
Affiliation(s)
- Maris Laan
- Human Molecular Genetics Research Group, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia.
| | | | | |
Collapse
|
35
|
Abstract
Men have higher blood pressure than women through much of life regardless of race and ethnicity. This is a robust and highly conserved sex difference that it is also observed across species including dogs, rats, mice and chickens and it is found in induced, genetic and transgenic animal models of hypertension. Not only do the differences between the ovarian and testicular hormonal milieu contribute to this sexual dimorphism in blood pressure, the sex chromosomes also play a role in and of themselves. This review primarily focuses on epidemiological studies of blood pressure in men and women and experimental models of hypertension in both sexes. Gaps in current knowledge regarding what underlie male-female differences in blood pressure control are discussed. Elucidating the mechanisms underlying sex differences in hypertension may lead to the development of anti-hypertensives tailored to one's sex and ultimately to improved therapeutic strategies for treating this disease and preventing its devastating consequences.
Collapse
Affiliation(s)
- Kathryn Sandberg
- Center for the Study of Sex Differences in Health, Disease and Aging Georgetown University, Washington, DC 20057
| | - Hong Ji
- Center for the Study of Sex Differences in Health, Disease and Aging Georgetown University, Washington, DC 20057
| |
Collapse
|
36
|
Kuningas M, Altmäe S, Uitterlinden AG, Hofman A, van Duijn CM, Tiemeier H. The relationship between fertility and lifespan in humans. AGE (DORDRECHT, NETHERLANDS) 2011; 33:615-622. [PMID: 21222045 PMCID: PMC3220400 DOI: 10.1007/s11357-010-9202-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Accepted: 12/13/2010] [Indexed: 05/30/2023]
Abstract
Evolutionary theories of aging predict a trade-off between fertility and lifespan, where increased lifespan comes at the cost of reduced fertility. Support for this prediction has been obtained from various sources. However, which genes underlie this relationship is unknown. To assess it, we first analyzed the association of fertility with age at menarche and menopause, and with mortality in 3,575 married female participants of the Rotterdam Study. In addition, we conducted a candidate gene study where 1,664 single nucleotide polymorphisms (SNPs) in 25 candidate genes were analyzed in relation to number of children as a measure of fertility. SNPs that associated with fertility were analyzed for association with mortality. We observed no associations between fertility and age at menarche (p = 0.38) and menopause (p = 0.07). In contrast, fertility was associated with mortality. Women with two to three children had significantly lower mortality (hazard ratio (HR), 0.82; 95% confidence interval (95% CI), 0.69-0.97) compared to women with no children. No such benefit was observed for women with four or more children, who had a similar mortality risk (HR, 0.93; 95% CI, 0.76-1.13) as women with no children. The analysis of candidate genes revealed four genes that influence fertility after correction for multiple testing: CGB/LHB gene cluster (p = 0.0036), FSHR (p = 0.023), FST (p = 0.023), and INHBA (p = 0.021). However, none of the independent SNPs in these genes predicted mortality. In conclusion, women who bear two to three children live longer than those who bear none or many children, but this relationship was not mediated by the candidate genes analyzed in this study.
Collapse
Affiliation(s)
- Maris Kuningas
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
37
|
Casarini L, Pignatti E, Simoni M. Effects of polymorphisms in gonadotropin and gonadotropin receptor genes on reproductive function. Rev Endocr Metab Disord 2011; 12:303-21. [PMID: 21912887 DOI: 10.1007/s11154-011-9192-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Gonadotropins, the action of which is mediated at the level of their gonadal receptors, play a key role in sexual development, reproductive functions and in metabolism. The involvement of the gonadotropins and their receptor genotypes on reproductive function are widely studied. A large number of gonadotropins and their receptors gene polymorphisms are known, but the only one considerable as a clear, absolute genetic marker of reproductive features or disfunctions is the FSHR Asn680Ser polymorphism, since it modulates ovarian response to FSH. The aim of these studies would to be the prediction of the genetic causes of sex-related diseases to enable a customized clinical setting based on individual response of patients undergoing gonadotropin stimulation. In this review we discuss the latest information about the effects of polymorphisms of the gonadotropins and their receptor genes on reproductive functions of both male and female, and discuss their patho-physiological implications.
Collapse
Affiliation(s)
- Livio Casarini
- Department of Medicine, Endocrinology, Metabolism and Geriatrics, University of Modena and Reggio Emilia, via P. Giardini 1355, 41126 Modena, Italy
| | | | | |
Collapse
|
38
|
Desai SS, Achrekar SK, Pathak BR, Desai SK, Mangoli VS, Mangoli RV, Mahale SD. Follicle-stimulating hormone receptor polymorphism (G-29A) is associated with altered level of receptor expression in Granulosa cells. J Clin Endocrinol Metab 2011; 96:2805-12. [PMID: 21752882 DOI: 10.1210/jc.2011-1064] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
CONTEXT Polymorphisms of the FSHR gene are associated with variable ovarian response to FSH stimulation in subjects undergoing in vitro fertilization (IVF) treatment. The type of ovarian response is correlated with the level of FSH receptor (FSHR) expression on granulosa cells. OBJECTIVE We investigated whether the polymorphism at position -29 in the promoter of the FSHR gene may contribute in altered receptor expression. DESIGN AND PATIENTS FSHR polymorphism at position -29 was studied in 100 subjects undergoing IVF treatment. Association of this polymorphism with level of FSHR expression was retrospectively analyzed. SETTING The study was conducted at an academic research institute and private IVF clinic. METHODS The genotype at position -29 of the FSHR gene was studied in IVF subjects by PCR-restriction fragment length polymorphism. Total RNA and protein was extracted from granulosa cells. The relative FSHR mRNA expression was carried out by real-time PCR. The receptor protein expression was evaluated by Western blot and confocal microscopy. RESULTS The clinical and endocrinological parameters revealed that almost 72% of subjects with the AA genotype at position -29 of FSHR gene were poor ovarian responders (odds ratio 8.63, 95% confidential interval 1.84-45.79; P = 0.001). The lower cleavage intensity predicted by in silico analysis for A allele as compared with the G allele suggest the difference in the DNA-protein binding affinity. The relative expression of FSHR at mRNA and protein level was significantly reduced in subjects with AA genotype as compared with the GG genotype. CONCLUSION Poor ovarian response observed in subjects with the AA genotype at position -29 of the FSHR gene is due to reduced receptor expression.
Collapse
Affiliation(s)
- Swapna S Desai
- National Institute for Research in Reproductive Health, Mumbai 400 012, India
| | | | | | | | | | | | | |
Collapse
|
39
|
Altmäe S, Hovatta O, Stavreus-Evers A, Salumets A. Genetic predictors of controlled ovarian hyperstimulation: where do we stand today? Hum Reprod Update 2011; 17:813-28. [DOI: 10.1093/humupd/dmr034] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
40
|
Kang L, Zhang N, Zhang Y, Yan H, Tang H, Yang C, Wang H, Jiang Y. Molecular characterization and identification of a novel polymorphism of 200 bp indel associated with age at first egg of the promoter region in chicken follicle-stimulating hormone receptor (FSHR) gene. Mol Biol Rep 2011; 39:2967-73. [PMID: 21678054 DOI: 10.1007/s11033-011-1058-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 06/08/2011] [Indexed: 01/23/2023]
Abstract
Follicle-stimulating hormone receptor (FSHR) plays an important role in animal follicular development. Polymorphisms in FSHR promoter region likely impact transcription and follicle growth and maturation. In this study, a fragment of ~1.9 kb of cFSHR promoter for Zang, Xianju, Lohmann Brown, Jining Bairi and Wenchang breeds (line) was obtained. Totally 49 variations were revealed, of which 39 are single nucleotide substitutions, one is nucleotide substitution of (TTG) to (CAC) and nine are indels. Polymorphism at -874 site (a 200 bp indel mutation) was identified, and their effects on egg production traits as well as gene expression were analyzed. At this site, allele I(+) was dominant in Lohmann Brown and Xinyang Brown (a synthetic egg-laying line) lines, but very rare in three Chinese indigenous chicken breeds, namely Jining Bairi, Wenchang, Zang and one synthetic boiler line (Luqin). In Xinyang Brown population, the polymorphism was associated with age at first egg (AFE) (P < 0.05) and its effect on egg number at 37 weeks of age (E37) and egg number at 57 weeks of age (E57) was not significantly different (P > 0.05). The cFSHR mRNA level was not significantly different between three genotypes in small white and small yellow follicles of Xinyang Brown hens, however, allele I(+) tends to increase cFSHR transcription.
Collapse
Affiliation(s)
- Li Kang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Taian, Shandong, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Polymorphisms in follicle stimulation hormone receptor, inhibin alpha, inhibin bata A, and prolactin genes, and their association with sperm quality in Chinese Holstein bulls. Anim Reprod Sci 2011; 126:151-6. [PMID: 21684095 DOI: 10.1016/j.anireprosci.2011.04.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Revised: 04/17/2011] [Accepted: 04/26/2011] [Indexed: 11/22/2022]
Abstract
Candidate genes follicle stimulation hormone receptor (FSHR), inhibin alpha (INHA), inhibin bata A (INHBA) and prolactin (PRL) were investigated for their association with sperm quality traits of semen volume per ejaculate (VOL), sperm concentration (SCON), motility (MOT), sperm motility in frozen semen (FMOT), acrosome integrity rate (AIR) and abnormal sperm rate (ASR) in a total of 123 normal mature Holstein bulls. Three reported single nucleotide polymorphisms (SNPs) of FSHR A-234500T (rs43676359), IHNA A192G (rs41257116), and IHNBA C7639T (rs43408735), and 3 novel SNPs (G7550A, C7661T, and T8370C) in exons 4 and 5 of bovine PRL gene (NC 007324) were analyzed. Analysis of variance revealed that FSHR A-234500T and INHBA C7639T polymorphisms significantly associated with VOL (P<0.05) and SCON (P<0.05), and the polymorphism of INHBA C7639T also had significant effects on MOT (P<0.05). Furthermore, the INHA A192G polymorphism significantly associated with AIR (P<0.05). No significant differences were found between the polymorphisms of PRL gene and sperm quality traits.
Collapse
|
42
|
Li Y, Gu A, Yang H, Ding X, Ji G, Lu C, Xia Y, Song L, Wang X. FSH receptor gene polymorphisms in fertile and infertile Han-Chinese males. Clin Chim Acta 2011; 412:1048-1052. [PMID: 21334319 DOI: 10.1016/j.cca.2011.02.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 02/11/2011] [Accepted: 02/11/2011] [Indexed: 11/21/2022]
Abstract
BACKGROUND Follicle stimulating hormone receptor (FSHR), which mediates the effects of FSH, is essential for normal spermatogenesis and male reproduction. This study aimed to investigate the effects of the FSHR polymorphisms on idiopathic male infertility and serum FSH levels in Han-Chinese population. METHODS A case-control study was conducted with 364 idiopathic infertile patients (97 nonobstructive azoospermic, 79 oligozoospermic and 188 normozoospermic) and 281 fertile controls. Three polymorphisms at nucleotide position -29 and codons 307 and 680 of the FSHR gene were genotyped by Taqman allelic discrimination and RFLP. FSH levels were measured by RIA. RESULTS The allele and genotype frequencies of these three polymorphisms were not significantly different between each group of the cases and controls. Serum FSH concentrations did not differ between subjects with different genotypes within each group. Together the three SNPs mainly formed four discrete haplotypes. The distribution of the haplotype was not different between each group of infertile men and fertile controls and did not influence serum FSH levels in each group. CONCLUSIONS Our findings suggest that the FSHR polymorphisms at the studied sites do not associate with idiopathic male infertility and have no influence on FSH levels both in normal and infertile males in the Han-Chinese population.
Collapse
Affiliation(s)
- Yingchun Li
- Key Laboratory of Reproductive Medicine, Institute of Toxicology, Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Morón FJ, Ruiz A. Pharmacogenetics of controlled ovarian hyperstimulation: time to corroborate the clinical utility of FSH receptor genetic markers. Pharmacogenomics 2011; 11:1613-8. [PMID: 21121812 DOI: 10.2217/pgs.10.156] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Several studies have shown high variability in clinical outcome among women undergoing follicle-stimulating hormone treatment. Pharmacogenetic studies have revealed a series of genetic markers involved in controlled ovarian hyperstimulation (COH) response. FSHR gene-associated SNPs, including the N680S missense variant, are the most promising genetic markers available to date. In this paper the state of the art pharmacogenetic analysis of COH outcome is reviewed and a meta-analysis is conducted with available data that confirms that the N680S marker is associated with poor response during COH. Thus, we propose that by pooling together available information, it is possible to go one step further with this biomarker to definitively validate its utility in the clinical field. We propose to conduct clinical trials, to look for algorithms integrating the N680S genotype and to test if such clinical protocols can optimize recombinant follicle-stimulating hormone dose and detect women at risk of a poor response during a COH cycle.
Collapse
Affiliation(s)
- Francisco Jesús Morón
- Department of Structural Genomics, Neocodex, Calle Charles Darwin sn, Parque, Científico y Tecnológico Isla de la Cartuja, 41092, Seville, Spain
| | | |
Collapse
|
44
|
Painter JN, Willemsen G, Nyholt D, Hoekstra C, Duffy DL, Henders AK, Wallace L, Healey S, Cannon-Albright LA, Skolnick M, Martin NG, Boomsma DI, Montgomery GW. A genome wide linkage scan for dizygotic twinning in 525 families of mothers of dizygotic twins. Hum Reprod 2010; 25:1569-80. [PMID: 20378614 DOI: 10.1093/humrep/deq084] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND The tendency to conceive dizygotic (DZ) twins is a complex trait influenced by genetic and environmental factors. To search for new candidate loci for twinning, we conducted a genome-wide linkage scan in 525 families using microsatellite and single nucleotide polymorphism marker panels. METHODS AND RESULTS Non-parametric linkage analyses, including 523 families containing a total of 1115 mothers of DZ twins (MODZT) from Australia and New Zealand (ANZ) and The Netherlands (NL), produced four linkage peaks above the threshold for suggestive linkage, including a highly suggestive peak at the extreme telomeric end of chromosome 6 with an exponential logarithm of odds [(exp)LOD] score of 2.813 (P = 0.0002). Since the DZ twinning rate increases steeply with maternal age independent of genetic effects, we also investigated linkage including only families where at least one MODZT gave birth to her first set of twins before the age of 30. These analyses produced a maximum expLOD score of 2.718 (P = 0.0002), largely due to linkage signal from the ANZ cohort, however, ordered subset analyses indicated this result is most likely a chance finding in the combined dataset. Linkage analyses were also performed for two large DZ twinning families from the USA, one of which produced a peak on chromosome 2 in the region of two potential candidate genes. Sequencing of FSHR and FIGLA, along with INHBB in MODZTs from two large NL families with family specific linkage peaks directly over this gene, revealed a potentially functional variant in the 5' untranslated region of FSHR that segregated with the DZ twinning phenotype in the Utah family. CONCLUSION Our data provide further evidence for complex inheritance of familial DZ twinning.
Collapse
Affiliation(s)
- Jodie N Painter
- Molecular Epidemiology, Genetic Epidemiology and Neurogenetics Laboratories, Queensland Institute of Medical Research, Brisbane, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Follicle stimulating hormone receptor gene variants in women with primary and secondary amenorrhea. J Assist Reprod Genet 2010; 27:317-26. [PMID: 20237833 DOI: 10.1007/s10815-010-9404-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2010] [Accepted: 02/23/2010] [Indexed: 10/19/2022] Open
Abstract
PURPOSE This retrospective study was designed to analyze the FSHR gene variants in subjects with primary and secondary amenorrhea with hypergonadotropic hypogonadism. MATERIALS AND METHODS Eighty six women with primary or secondary amenorrhea and 100 normally cycling proven fertile women of Indian origin were retrospectively studied. These subjects were systematically screened for entire FSHR gene. RESULTS The frequency distribution of polymorphism at -29 position of FSHR gene is altered in women with primary and secondary amenorrhea as compared to controls. AA genotype at -29 position of FSHR gene seems to be associated with increased serum FSH levels in the study subjects. We have identified a novel homozygous mutation C(1723)T (Ala(575)Val) in one woman with primary amenorrhea. CONCLUSIONS Our findings suggest that increased serum FSH levels in subjects with primary amenorrhea correlated to FSHR genotype at position -29. We identified a novel homozygous mutation C(1723)T (Ala(575)Val) in a woman with primary amenorrhea.
Collapse
|
46
|
Deng AY, Ménard A, Xiao C, Roy J. Sexual Dimorphism on Hypertension of Quantitative Trait Loci Entrapped in Dahl Congenic Rats. Clin Exp Hypertens 2009; 30:511-9. [DOI: 10.1080/10641960802251933] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Alan Y. Deng
- Research Centre, Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Annie Ménard
- Research Centre, Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Chunjie Xiao
- Biology Department, Yunnan University, Kunming, Yunnan, China
| | - Julie Roy
- Research Centre, Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
47
|
Achrekar SK, Modi DN, Desai SK, Mangoli VS, Mangoli RV, Mahale SD. Poor ovarian response to gonadotrophin stimulation is associated with FSH receptor polymorphism. Reprod Biomed Online 2009; 18:509-15. [PMID: 19400992 DOI: 10.1016/s1472-6483(10)60127-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Similarities in the phenotype observed in women with FSH receptor mutation and in FSH receptor knockout mice have clearly established a critical role of this protein in normal gonadal function. Two common single nucleotide polymorphisms in the exonic region of the FSH receptor gene have been shown to be associated with altered ovarian response in subjects undergoing gonadotrophin treatment. Recent in-vitro studies have shown that the A allele at the -29 position in the 5 untranslated region of the FSH receptor gene is associated with impaired transcriptional activity. Differential expression of the FSH receptor and its function may be one of the factors responsible for altered ovarian response. These observations prompted a study of the association between FSH receptor genotype at the -29 position and ovarian response in women undergoing gonadotrophin treatment. Analysis of the data revealed that the subjects with AA genotype at the -29 position required the highest amount of exogenous FSH for ovulation induction, and oestradiol concentrations before the day of human chorionic gonadotrophin administration were significantly lower (P = 0.015) compared with the GA genotype. The number of pre-ovulatory follicles and retrieved oocytes were lowest in the subjects with AA genotype. These results indicate that the AA genotype at position -29 may be associated with the poor ovarian response.
Collapse
Affiliation(s)
- Swati K Achrekar
- National Institute for Research in Reproductive Health, J M Street, Parel, Mumbai, India
| | | | | | | | | | | |
Collapse
|
48
|
Nakayama T, Yamamoto T. Comparison between essential hypertension and pregnancy-induced hypertension: a genetic perspective. Endocr J 2009; 56:921-34. [PMID: 19851037 DOI: 10.1507/endocrj.k09e-253] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Essential hypertension (EH) accounts for 80-90% of hypertension, and pregnancy-induced hypertension (PIH) is responsible for hypertension during pregnancy. Both considered multifactorial disorders. While both have many features in common, conditions and causes of these diseases have yet to be clarified. Since both diseases are associated with hypertension, the genetic backgrounds may contain common features. The present study reviewed similarities and differences between women with EH and PIH, with a particular focus on their genetic backgrounds.
Collapse
Affiliation(s)
- Tomohiro Nakayama
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan.
| | | |
Collapse
|
49
|
Ogurtsov AY, Mariño-Ramírez L, Johnson GR, Landsman D, Shabalina SA, Spiridonov NA. Expression patterns of protein kinases correlate with gene architecture and evolutionary rates. PLoS One 2008; 3:e3599. [PMID: 18974838 PMCID: PMC2572838 DOI: 10.1371/journal.pone.0003599] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2008] [Accepted: 10/09/2008] [Indexed: 12/20/2022] Open
Abstract
Background Protein kinase (PK) genes comprise the third largest superfamily that occupy ∼2% of the human genome. They encode regulatory enzymes that control a vast variety of cellular processes through phosphorylation of their protein substrates. Expression of PK genes is subject to complex transcriptional regulation which is not fully understood. Principal Findings Our comparative analysis demonstrates that genomic organization of regulatory PK genes differs from organization of other protein coding genes. PK genes occupy larger genomic loci, have longer introns, spacer regions, and encode larger proteins. The primary transcript length of PK genes, similar to other protein coding genes, inversely correlates with gene expression level and expression breadth, which is likely due to the necessity to reduce metabolic costs of transcription for abundant messages. On average, PK genes evolve slower than other protein coding genes. Breadth of PK expression negatively correlates with rate of non-synonymous substitutions in protein coding regions. This rate is lower for high expression and ubiquitous PKs, relative to low expression PKs, and correlates with divergence in untranslated regions. Conversely, rate of silent mutations is uniform in different PK groups, indicating that differing rates of non-synonymous substitutions reflect variations in selective pressure. Brain and testis employ a considerable number of tissue-specific PKs, indicating high complexity of phosphorylation-dependent regulatory network in these organs. There are considerable differences in genomic organization between PKs up-regulated in the testis and brain. PK genes up-regulated in the highly proliferative testicular tissue are fast evolving and small, with short introns and transcribed regions. In contrast, genes up-regulated in the minimally proliferative nervous tissue carry long introns, extended transcribed regions, and evolve slowly. Conclusions/Significance PK genomic architecture, the size of gene functional domains and evolutionary rates correlate with the pattern of gene expression. Structure and evolutionary divergence of tissue-specific PK genes is related to the proliferative activity of the tissue where these genes are predominantly expressed. Our data provide evidence that physiological requirements for transcription intensity, ubiquitous expression, and tissue-specific regulation shape gene structure and affect rates of evolution.
Collapse
Affiliation(s)
- Aleksey Y. Ogurtsov
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Leonardo Mariño-Ramírez
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gibbes R. Johnson
- Division of Therapeutic Proteins, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Bethesda, Maryland, United States of America
| | - David Landsman
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Svetlana A. Shabalina
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (NAS); (SAS)
| | - Nikolay A. Spiridonov
- Division of Therapeutic Proteins, Center for Drug Evaluation and Research, U. S. Food and Drug Administration, Bethesda, Maryland, United States of America
- * E-mail: (NAS); (SAS)
| |
Collapse
|
50
|
Shih PA, O’Connor D, Mahata S. Human Genomics in Hypertension. Genomics 2008. [DOI: 10.3109/9781420067064-11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|