1
|
Ma W, Ye S, Tian L, Liu M, Wang R, Yang X, Wang M, Fu F, Ren W, Dang L, Wang T, Wang W, Wang S, Sun Y, Li Y. Association of LHCGR rs2293275 genotype with ovarian aging in Chinese women: a multicenter population-based study. Reprod Biol Endocrinol 2025; 23:41. [PMID: 40089715 PMCID: PMC11909885 DOI: 10.1186/s12958-025-01375-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 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: 12/04/2024] [Accepted: 03/02/2025] [Indexed: 03/17/2025] Open
Abstract
OBJECTIVE To evaluate the association between the LHCGR rs2293275 (N312S) genotype and ovarian aging phenotypes in Han Chinese women, focusing on diminished ovarian reserve (DOR) and primary ovarian insufficiency (POI). STUDY DESIGN This multicenter population-based study included 1,240 women aged 18-40 years diagnosed with DOR (n = 711) or POI (n = 529), alongside 72,846 ethnically and regionally matched controls from the Han Chinese Genomes Database (PGG.Han). Genotyping of rs2293275 was performed, and clinical data (menstrual history, hormonal profiles, maternal menopause age, and ART outcomes) were analyzed. MAIN RESULTS The AA genotype frequency in the ovarian aging cohort (1.85%) was significantly higher than in the general Han population (0.62%, OR 3.04, 95% CI 1.99-4.64, p < 0.001). AA carriers exhibited earlier POI diagnosis (25.5 ± 6.4 vs. 32.0 ± 5.1 years in GG carriers, p < 0.001) and maternal menopause (41.6 ± 3.3 vs. 47.8 ± 4.1 years, p < 0.001). In controlled ovarian stimulation cycles, AA carriers demonstrated reduced ovarian sensitivity (OSI: 3.59 vs. 1.21 in GG, p = 0.019) despite comparable gonadotropin doses. CONCLUSIONS The LHCGR rs2293275 AA genotype is strongly associated with accelerated ovarian aging in Han Chinese women, highlighting its potential as a biomarker for early identification of high-risk individuals. While these findings underscore genetic contributions to ovarian dysfunction, further mechanistic studies are needed to establish causality and optimize clinical translation. TRIAL REGISTRATION NUMBER ClinicalTrials.gov NCT05665010, registered on 2022-11-30.
Collapse
Affiliation(s)
- Wenqing Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuangmei Ye
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lifeng Tian
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Women's Reproductive Health, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, 330000, China
| | - Min Liu
- Changsha Maternal and Child Health Hospital, Changsha, 410000, China
| | - Rui Wang
- Henan Maternal and Child Health Hospital, Zhengzhou, 450000, China
| | - Xuezhou Yang
- Department of Reproductive Center, Affiliated Hospital of Hubei, Xiangyang Central Hospital, University of Arts and Science, Xiangyang, 441000, China
| | - Man Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fangfang Fu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wu Ren
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lei Dang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tian Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenwen Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Sun
- Fujian Maternal and Child Health Hospital, Fuzhou, 350001, China.
| | - Yan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Yang N, Wei XG, Li K, Wang F, Song F, Sun W, Wang Y, Zhao Z, Mu J, Ma H. Transcriptomic analysis of the HPT axis in a model of oligoasthenozoospermia induced by Adenine in rats. Exp Mol Pathol 2025; 141:104948. [PMID: 39700678 DOI: 10.1016/j.yexmp.2024.104948] [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: 11/12/2023] [Revised: 11/25/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
Male infertility is most commonly caused by oligozoospermia, and its pathogenesis is still poorly understood at the molecular level. This study used RNA sequencing (RNA-Seq) technology to identify candidate genes and regulatory pathways that regulate semen quality in the hypothalamic, pituitary, and testicular tissues of healthy rats and Adenine-induced oligozoospermia model rats. Semen quality testing and histological analysis of testicular tissues were performed on both groups of rats. We identified 627, 692, and 437 differentially expressed genes in the hypothalamus, pituitary gland, and testes, respectively. Functional analysis indicates that "neuronal projections," "positive regulation of hormone biosynthetic process," and "neuroactive ligand-receptor interaction pathways" are closely related to the hypothalamic-pituitary-testicular (HPT) axis hormone regulation and sperm production. Seven genes (Pomc, Rxfp1, Tac1, Npy, Insl3, Hsd3b3, Lhcgr) have been identified as key candidate genes responsible for regulating sperm quality within the HPT axis, potentially affecting rat reproductive function by influencing testicular development and testosterone secretion. These data provide a theoretical basis for further understanding the molecular mechanisms of reproductive performance in a rat model of oligoasthenozoospermia.
Collapse
Affiliation(s)
- Nan Yang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China; Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia Medical University, Yinchuan 750000, China
| | - Xiao-Ge Wei
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China; Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia Medical University, Yinchuan 750000, China
| | - Kaiying Li
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China; Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia Medical University, Yinchuan 750000, China
| | - Fei Wang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China; Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia Medical University, Yinchuan 750000, China
| | - Fei Song
- Lianyungang No. 1 People's Hospital, Lianyungang 222000, Jiangsu Province, China
| | - Wenjing Sun
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China; Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia Medical University, Yinchuan 750000, China
| | - Yan Wang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China
| | - Zhenning Zhao
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China
| | - Jing Mu
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China.
| | - Huisheng Ma
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750000, China; Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia Medical University, Yinchuan 750000, China.
| |
Collapse
|
3
|
Conforti A, Di Girolamo R, Guida M, Alviggi C, Casarini L. Pharmacogenomic of LH and its receptor: are we ready for clinical practice? Reprod Biol Endocrinol 2025; 23:29. [PMID: 40001128 PMCID: PMC11863420 DOI: 10.1186/s12958-025-01359-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Luteinizing hormone (LH) is fundamental to support development and reproduction. It acts through a receptor expressed in the gonads, modulating mitogenic, anti-apoptotic, and steroidogenic signals. LH is also marketed as a drug for controlled ovarian stimulation (COS), where it is administered to women to support the action of follicle-stimulating hormone and can lead to specific responses, depending on the individual genetic background. These concepts underline the relevance of a pharmacogenetic approach to COS, in the attempt to optimize clinical outcomes and avoid adverse events. However, knowledge is currently limited by the paucity of clinical studies. This review aims to provide a comprehensive overview of LH and its receptor activity, starting from the description of their molecular pathways from in vitro studies. Data on LH action from in vivo studies were described, as well as the impact of LH and LH/choriogonadotropin (hCG) receptor genetic variants on folliculogenesis and its association with infertility or polycystic ovarian syndrome. Finally, evidence from clinical studies evaluating genetic polymorphisms in the context of assisted reproductive technology treatments and its implications for a pharmacogenomic approach were discussed.
Collapse
Affiliation(s)
- Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Via Sergio Pansini, 5, Napoli, 80131, Italy.
| | | | - Maurizio Guida
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Via Sergio Pansini, 5, Napoli, 80131, Italy
| | - Carlo Alviggi
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
4
|
Behvarz M, Rahmani SA, Siasi Torbati E, Danaei Mehrabad S, Bikhof Torbati M. Correlation between LHCGR and NR5A1 genes polymorphism and male infertility risk. Actas Urol Esp 2024; 48:246-253. [PMID: 37827240 DOI: 10.1016/j.acuroe.2023.10.001] [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: 07/09/2022] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 10/14/2023]
Abstract
INTRODUCTION Infertility is one of the important phenomena in human reproduction. Genetic factors are the most important cause of male infertility. Here, we aimed to investigate the correlation between idiopathic male infertility and SNPs of the LHCGR (rs2293275) and NR5A1 (rs1057517779) genes in the Iranian-Azeri population. METHODS This case-control study consisted of 100 males with infertility and 100 healthy males from the Iranian Azeri population. Genomic DNA isolation from whole blood samples and Tetra-primer amplification refractory mutation system-polymerase chain reaction (Tetra-ARMS-PCR) method was used for genotyping. The data analysis was performed by chi-square (χ2) and Fisher's exact tests. RESULTS Genotyping analysis for LHCGR (rs2293275) polymorphism indicated that the frequency of C in the case group was significantly higher than in the control group (P < .05). Moreover, genotyping analysis for NR5A1 (rs1057517779) polymorphism indicated that the frequencies of the A allele and heterozygote GA genotype in the case group were significantly higher than those in the control group (P < .05). CONCLUSION Our study demonstrated that the SNPs of LHCGR (rs2293275) and NR5A1 (rs1057517779) genes may play a critical role in male infertility in the Iranian Azeri population. However, further studies on other ethnic origins with larger sample sizes are essential for accessing more accurate results. Moreover, functional experiments might be needed to understand the role of these polymorphisms in the molecular pathways involved in male fertility.
Collapse
Affiliation(s)
- M Behvarz
- Departamento de Genética, Facultad de Ciencias, Sede del Norte de Teherán, Universidad Islámica Azad, Teherán, Iran
| | - S A Rahmani
- Departamento de Genética Médica, Facultad de Medicina, Universidad de Ciencias Médicas de Tabriz, Tabriz, Iran
| | - E Siasi Torbati
- Departamento de Genética, Facultad de Ciencias, Sede del Norte de Teherán, Universidad Islámica Azad, Teherán, Iran.
| | - S Danaei Mehrabad
- Departamento de Ginecología, Centro ACECR ART, Sede ACECR Azerbaiyán Oriental, Tabriz, Iran
| | - M Bikhof Torbati
- Departamento de Biología, Sede Yadegar-e-Imam Khomeini (RAH) Shahr-e-Rey, Universidad Islámica Azad, Teherán, Iran
| |
Collapse
|
5
|
Xu Y, Wang E, Liu T, Wang S, Wu F, Zhao X, Wang A. Whole exome sequencing identifies a novel homozygous missense mutation of LHCGR gene in primary infertile women with empty follicle syndrome. J Obstet Gynaecol Res 2023; 49:2436-2445. [PMID: 37462066 DOI: 10.1111/jog.15747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/05/2023] [Indexed: 10/03/2023]
Abstract
AIM The genetic basis of empty follicle syndrome (EFS) is largely unknown, and the aim of this study was to investigate the genetic causes of EFS in primary infertile women. METHODS Four affected women diagnosed with anovulation were recruited, and whole exome sequencing (WES) was requested for the genetic diagnosis of the cases. One hundred healthy controls were verified by Sanger sequencing. RESULTS A novel homozygous variant of the LHCGR gene (NM_000233:c.1847C>A) was revealed in one affected individual by WES. Trios analysis of the mutation revealed an autosomal recessive pattern. This LHCGR variant was absent in 100 healthy controls and predicted to be highly damaging to the function of LHCGR. CONCLUSIONS The novel variant extends the mutational spectrum of the LHCGR gene associated with female sterility, which promotes the prognostic value of testing for LHCGR mutations in infertile women with EFS.
Collapse
Affiliation(s)
- Yang Xu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Department of Reproductive Medicine, Linyi People's Hospital, Shandong University, Linyi, China
| | - Enhua Wang
- Teaching and Research Office of Medical Genetics, Shandong Medical College, Linyi, China
| | - Tianfeng Liu
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, China
| | - Surong Wang
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, China
| | - Fengxia Wu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Xiangyu Zhao
- Department of Medical Genetics, Linyi People's Hospital, Shandong University, Linyi, China
| | - Ancong Wang
- Department of Reproductive Medicine, Linyi People's Hospital, Shandong University, Linyi, China
| |
Collapse
|
6
|
Errico A, Vinco S, Ambrosini G, Dalla Pozza E, Marroncelli N, Zampieri N, Dando I. Mitochondrial Dynamics as Potential Modulators of Hormonal Therapy Effectiveness in Males. BIOLOGY 2023; 12:547. [PMID: 37106748 PMCID: PMC10135745 DOI: 10.3390/biology12040547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/21/2023] [Accepted: 04/01/2023] [Indexed: 04/29/2023]
Abstract
Worldwide the incidence of andrological diseases is rising every year and, together with it, also the interest in them is increasing due to their strict association with disorders of the reproductive system, including impairment of male fertility, alterations of male hormones production, and/or sexual function. Prevention and early diagnosis of andrological dysfunctions have long been neglected, with the consequent increase in the incidence and prevalence of diseases otherwise easy to prevent and treat if diagnosed early. In this review, we report the latest evidence of the effect of andrological alterations on fertility potential in both young and adult patients, with a focus on the link between gonadotropins' mechanism of action and mitochondria. Indeed, mitochondria are highly dynamic cellular organelles that undergo rapid morphological adaptations, conditioning a multitude of aspects, including their size, shape, number, transport, cellular distribution, and, consequently, their function. Since the first step of steroidogenesis takes place in these organelles, we consider that mitochondria dynamics might have a possible role in a plethora of signaling cascades, including testosterone production. In addition, we also hypothesize a central role of mitochondria fission boost on the decreased response to the commonly administrated hormonal therapy used to treat urological disease in pediatric and adolescent patients as well as infertile adults.
Collapse
Affiliation(s)
- Andrea Errico
- Department of Neurosciences, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, 37100 Verona, Italy; (A.E.); (S.V.); (G.A.); (E.D.P.); (N.M.)
| | - Sara Vinco
- Department of Neurosciences, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, 37100 Verona, Italy; (A.E.); (S.V.); (G.A.); (E.D.P.); (N.M.)
| | - Giulia Ambrosini
- Department of Neurosciences, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, 37100 Verona, Italy; (A.E.); (S.V.); (G.A.); (E.D.P.); (N.M.)
| | - Elisa Dalla Pozza
- Department of Neurosciences, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, 37100 Verona, Italy; (A.E.); (S.V.); (G.A.); (E.D.P.); (N.M.)
| | - Nunzio Marroncelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, 37100 Verona, Italy; (A.E.); (S.V.); (G.A.); (E.D.P.); (N.M.)
| | - Nicola Zampieri
- Department of Engineering and Innovation Medicine, Paediatric Fertility Lab, Woman and Child Hospital, Division of Pediatric Surgery, University of Verona, 37100 Verona, Italy;
| | - Ilaria Dando
- Department of Neurosciences, Biomedicine and Movement Sciences, Biochemistry Section, University of Verona, 37100 Verona, Italy; (A.E.); (S.V.); (G.A.); (E.D.P.); (N.M.)
| |
Collapse
|
7
|
Bhattacharya I, Dey S, Banerjee A. Revisiting the gonadotropic regulation of mammalian spermatogenesis: evolving lessons during the past decade. Front Endocrinol (Lausanne) 2023; 14:1110572. [PMID: 37124741 PMCID: PMC10140312 DOI: 10.3389/fendo.2023.1110572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Spermatogenesis is a multi-step process of male germ cell (Gc) division and differentiation which occurs in the seminiferous tubules of the testes under the regulation of gonadotropins - Follicle Stimulating Hormone (FSH) and Luteinising hormone (LH). It is a highly coordinated event regulated by the surrounding somatic testicular cells such as the Sertoli cells (Sc), Leydig cells (Lc), and Peritubular myoid cells (PTc). FSH targets Sc and supports the expansion and differentiation of pre-meiotic Gc, whereas, LH operates via Lc to produce Testosterone (T), the testicular androgen. T acts on all somatic cells e.g.- Lc, PTc and Sc, and promotes the blood-testis barrier (BTB) formation, completion of Gc meiosis, and spermiation. Studies with hypophysectomised or chemically ablated animal models and hypogonadal (hpg) mice supplemented with gonadotropins to genetically manipulated mouse models have revealed the selective and synergistic role(s) of hormones in regulating male fertility. We here have briefly summarized the present concept of hormonal control of spermatogenesis in rodents and primates. We also have highlighted some of the key critical questions yet to be answered in the field of male reproductive health which might have potential implications for infertility and contraceptive research in the future.
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Department of Zoology, School of Biological Science, Central University of Kerala, Kasaragod, Kerala, India
- *Correspondence: Arnab Banerjee, ; Indrashis Bhattacharya,
| | - Souvik Dey
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Arnab Banerjee
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS) Pilani, Goa, India
- *Correspondence: Arnab Banerjee, ; Indrashis Bhattacharya,
| |
Collapse
|
8
|
Rivero-Müller A, Huhtaniemi I. Genetic variants of gonadotrophins and their receptors: Impact on the diagnosis and management of the infertile patient. Best Pract Res Clin Endocrinol Metab 2022; 36:101596. [PMID: 34802912 DOI: 10.1016/j.beem.2021.101596] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This narrative review is concerned with genetic variants of the genes encoding gonadotrophin subunits and their receptors, as well as their implications into the diagnosis and treatment of infertility. We first review briefly the basics of molecular biology and biochemistry of gonadotrophin and gonadotrophin receptor structure and function, then describe the phenotypic effects of polymorphisms and mutations of these genes, followed by diagnostic aspects. We will then summarise the information that inactivating gonadotrophin receptor mutations have provided about the controversial topic of extragonadal gonadotrophin action. Finally, we will close with the current and future therapeutic approaches on patients with gonadotrophin and their receptor mutations.
Collapse
Affiliation(s)
- Adolfo Rivero-Müller
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, 20-093, Poland
| | - Ilpo Huhtaniemi
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK.
| |
Collapse
|
9
|
Koloda YA, Denisova YV, Podzolkova NM. Genetic polymorphisms of reproductive hormones and their receptors in assisted reproduction technology for patients with polycystic ovary syndrome. Drug Metab Pers Ther 2021; 37:111-122. [PMID: 34851566 DOI: 10.1515/dmpt-2021-0123] [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: 03/25/2021] [Accepted: 08/31/2021] [Indexed: 11/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrinopathies in women of childbearing, which is defined by the accumulation of multiple, small fluid-filled ovarian cysts without the selection of a single dominant follicle. Most PCOS phenotypes are characterized by the absence of spontaneous ovulation, resistance toward ovulation inductors, the production of a large immature oocytes number, and the high prevalence of ovarian hyperstimulation syndrome, resulting in reduced assisted reproductive technologies (ART) programs effectiveness. The review analyses current data about the relationship between polymorphism genotypes of KISS genes, follicle stimulating hormone (FSH), luteinizing hormone (LH), anti-Müllerian hormone (AMH) and their receptors genes, gonadotropin-releasing hormone (GnRH), estrogen, and progesterone receptors genes, the PCOS risk and the features of ovarian response to stimulation during ART cycles. The use of single nucleotide polymorphisms (SNPs) as prognostic markers of ART programs outcomes would provide a personalized approach to the drugs and doses choice for ovarian stimulation and significantly increase the chance of pregnancy.
Collapse
Affiliation(s)
- Yulia A Koloda
- Department of Obstetrics and Gynecology, Pediatric Faculty, FSBEI FPE "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of the Russian Federation, Moscow, Russian Federation
| | - Yulia V Denisova
- Department of Obstetrics and Gynecology, Pediatric Faculty, FSBEI FPE "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of the Russian Federation, Moscow, Russian Federation
| | - Natalia M Podzolkova
- Department of Obstetrics and Gynecology, Pediatric Faculty, FSBEI FPE "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of the Russian Federation, Moscow, Russian Federation
| |
Collapse
|
10
|
Ali ARA, Abdul-Rasheed OF, Al-Kawaz UM. The Impact of Luteinizing Hormone/Chorionic Gonadotropin Hormone Receptor Gene Polymorphism rs68073206 in Men with Non-obstructive Azoospermia: A Case-control Study. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: The functional consequences of the luteinizing hormone/chorionic gonadotropin hormone receptor (LHCGR) gene single nucleotide polymorphism (rs68073206) on male infertility in patients with non-obstructive azoospermia (NOA) is not clear.
Objective: To examine whether the presence of LHCGR gene; rs68073206 single nucleotide polymorphisms (SNPs) can be associated with incidence of non-obstructive azoospermia.
Materials and methods: A case-control study comprised of a total of 70 unrelated Iraqi infertile men with non-obstructive azoospermia (zero sperm in semen) whose were on two groups: Group I that were diagnosed to have NOA but didn’t receive infertility treatment yet (33 patient with age of 31.58±1.059 year) and group II that were receiving injectable gonadotropin treatment (37 patient with age of 33.46±1.173 year). In addition to 34 age and BMI matched healthy fertile normozoospermic men (according to the parameters of WHO, 2010). The study population was genotyped by TaqMan assay for LHCGR gene single nucleotide polymorphism (rs68073206). The level of each hormone was estimated by immunoassay technique while the sperm analyses were conducted in accordance with the World Health Organization criteria.
Results: The study revealed a statistically significant higher hormonal level of serum inhibin B in infertile group I patients with wild GG genotype (246.445±224.106 pg/ml), and the p-value is (0.0439) as compared to that hormone levels of GT and TT genotypes carriers that were (85.969±71.685 pg/ml) and (56.420±23.988 pg/ml) respectively. ). The genotyping variations of patients, whether carrying the homozygous GG, heterozygous GT or homozygous TT genotype, did not reveal a statistically significant difference in distribution as compared to control individuals.
Conclusions: The LHCGR gene rs68073206 polymorphisms in our population having non-obstructive azoospermia can be suggested to have a modulating potential in variable gonadotropin sensitivity. The detected non-significant difference in genotypic prevalence can be attributable to the limited sample size.
Collapse
|
11
|
Involvement of single nucleotide polymorphisms in ovarian poor response. J Assist Reprod Genet 2021; 38:2405-2413. [PMID: 34050449 DOI: 10.1007/s10815-021-02242-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/23/2021] [Indexed: 10/21/2022] Open
Abstract
PURPOSE Unpredictability in acquiring an adequate number of high-quality oocytes following ovarian stimulation is one of the major complications in controlled ovarian hyperstimulation (COH). Genetic predispositions of variations could alter the immunological profiles and consequently be implicated in the variability of ovarian response to the stimulation. DESIGN Uncovering the influence of variations in AMHR2, LHCGR, MTHFR, PGR, and SERPINE1 genes with ovarian response to gonadotrophin stimulation in COH of infertile women. METHODS Blood samples of the women with a good ovarian response (GOR) or with a poor ovarian response (POR) were collected. Genomic DNA was extracted, and gene variations were genotyped by TaqMan SNP Genotyping Assays using primer-probe sets or real-time PCR Kit. RESULTS Except for PGR (rs10895068), allele distributions demonstrate that the majority of POR patients carried minor alleles of AMHR2 (rs2002555, G-allele), LHCGR (rs2293275, G-allele), MTHFR (rs1801131, C-allele, and rs1801133, T-allele), and SERPINE1 (rs1799889, 4G allele) genes compared to the GOR. Similarly, genotypes with a minor allele in AMHR2, LHCGR, MTHFR, and SERPINE1 genes had a higher prevalence among POR patients with the polymorphic genotypes. However, further genotype stratification indicated that the minor alleles of these genes are not associated with poor response. Multivariate logistic analysis of clinical-demographic factors and polymorphic genotypes demonstrated a correlation between FSH levels and polymorphic genotypes of SERPINE1 in poor response status. CONCLUSIONS Despite a higher prevalence of AMHR2, LHCGR, MTHFR, and SERPINE1 variations in the patients with poor ovarian response, it seems that these variations are not associated with the ovarian response.
Collapse
|
12
|
Juel Mortensen L, Lorenzen M, Jørgensen A, Albrethsen J, Jørgensen N, Møller S, Andersson AM, Juul A, Blomberg Jensen M. Possible Relevance of Soluble Luteinizing Hormone Receptor during Development and Adulthood in Boys and Men. Cancers (Basel) 2021; 13:cancers13061329. [PMID: 33809538 PMCID: PMC7999540 DOI: 10.3390/cancers13061329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary The reproductive hormones luteinizing hormone (LH) and human chorionic gonadotropin (hCG) are both agonists for the luteinizing hormone receptor (LHCGR) and essential for male reproduction during development and adulthood. LHCGR is expressed and stimulates testosterone production from the testicular Leydig cells. In this study, we demonstrate the presence of soluble LHCGR in blood, urine, and seminal fluid in both healthy boys and men, and patients with aberrations in sex-chromosomes. We show how circulating levels of sLHCGR are associated with pubertal development, testicular function, and semen quality and demonstrate that LHCGR is released from fetal human non-gonadal tissue. sLHCGR is released into serum by testis and other organs, which suggests possible extra-gonadal effects of LH or hCG in boys and men. Abstract Luteinizing hormone (LH) and human chorionic gonadotropin (hCG) are agonists for the luteinizing hormone receptor (LHCGR) which regulates male reproductive function. LHCGR may be released into body fluids. We wish to determine whether soluble LHCGR is a marker for gonadal function. Cross-sectional, longitudinal, and intervention studies on 195 healthy boys and men and 396 men with infertility, anorchia, or Klinefelter Syndrome (KS) were used to correlate LHCGR measured in serum, seminal fluid, urine, and hepatic/renal artery and vein with gonadal function. LHCGR was determined in fluids from in vitro and in vivo models of human testicular tissue and cell lines, xenograft mouse models, and human fetal kidney and adrenal glands. Western blot showed LHCGR fragments in serum and gonadal tissue of similar size using three different antibodies. The LHCGR-ELISA had no species cross-reactivity or unspecific reaction in mouse serum even after human xenografting. Instead, sLHCGR was released into the media after the culture of a human fetal kidney and adrenal glands. Serum sLHCGR decreased markedly during puberty in healthy boys (p = 0.0001). In healthy men, serum sLHCGR was inversely associated with the Inhibin B/FSH ratio (β −0.004, p = 0.027). In infertile men, seminal fluid sLHCGR was inversely associated with serum FSH (β 0.006, p = 0.009), sperm concentration (β −3.5, p = 0.003) and total sperm count (β −3.2, p = 0.007). The injection of hCG lowered sLHCGR in serum and urine of healthy men (p < 0.01). In conclusion, sLHCGR is released into body-fluids and linked with pubertal development and gonadal function. Circulating sLHCGR in anorchid men suggests that sLHCGR in serum may originate from and possibly exert actions in non-gonadal tissues. (ClinicalTrials: NTC01411527, NCT01304927, NCT03418896).
Collapse
Affiliation(s)
- Li Juel Mortensen
- Group of Skeletal, Mineral and Gonadal Endocrinology, University Department of Growth and Reproduction, Rigshospitalet, 2100 Copenhagen, Denmark; (L.J.M.); (M.L.)
| | - Mette Lorenzen
- Group of Skeletal, Mineral and Gonadal Endocrinology, University Department of Growth and Reproduction, Rigshospitalet, 2100 Copenhagen, Denmark; (L.J.M.); (M.L.)
| | - Anne Jørgensen
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; (A.J.); (J.A.); (N.J.); (A.-M.A.); (A.J.)
| | - Jakob Albrethsen
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; (A.J.); (J.A.); (N.J.); (A.-M.A.); (A.J.)
| | - Niels Jørgensen
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; (A.J.); (J.A.); (N.J.); (A.-M.A.); (A.J.)
| | - Søren Møller
- Center for Functional and Diagnostic Imaging and Research, Department of Clinical Physiology and Nuclear Medicine 260, Hvidovre Hospital, 2650 Copenhagen, Denmark;
- Department of Clinical Medicine, Faculty of Health Sciences, Copenhagen University, 2200 Copenhagen, Denmark
| | - Anna-Maria Andersson
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; (A.J.); (J.A.); (N.J.); (A.-M.A.); (A.J.)
| | - Anders Juul
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; (A.J.); (J.A.); (N.J.); (A.-M.A.); (A.J.)
- Department of Clinical Medicine, Faculty of Health Sciences, Copenhagen University, 2200 Copenhagen, Denmark
| | - Martin Blomberg Jensen
- Group of Skeletal, Mineral and Gonadal Endocrinology, University Department of Growth and Reproduction, Rigshospitalet, 2100 Copenhagen, Denmark; (L.J.M.); (M.L.)
- Division of Bone and Mineral Research, Harvard School of Dental Medicine/Harvard Medical School, Boston, MA 02115, USA
- Correspondence: ; Tel.: +45-3545-5064
| |
Collapse
|
13
|
Jorgez CJ, Seth A, Wilken N, Bournat JC, Chen CH, Lamb DJ. E2F1 regulates testicular descent and controls spermatogenesis by influencing WNT4 signaling. Development 2021; 148:dev191189. [PMID: 33441379 PMCID: PMC7823160 DOI: 10.1242/dev.191189] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022]
Abstract
Cryptorchidism is the most common urologic birth defect in men and is a predisposing factor of male infertility and testicular cancer, yet the etiology remains largely unknown. E2F1 microdeletions and microduplications contribute to cryptorchidism, infertility and testicular tumors. Although E2f1 deletion or overexpression in mice causes spermatogenic failure, the mechanism by which E2f1 influences testicular function is unknown. This investigation revealed that E2f1-null mice develop cryptorchidism with severe gubernacular defects and progressive loss of germ cells resulting in infertility and, in rare cases, testicular tumors. It was hypothesized that germ cell depletion resulted from an increase in WNT4 levels. To test this hypothesis, the phenotype of a double-null mouse model lacking both Wnt4 and E2f1 in germ cells was analyzed. Double-null mice are fertile. This finding indicates that germ cell maintenance is dependent on E2f1 repression of Wnt4, supporting a role for Wnt4 in germ cell survival. In the future, modulation of WNT4 expression in men with cryptorchidism and spermatogenic failure due to E2F1 copy number variations may provide a novel approach to improve their spermatogenesis and perhaps their fertility potential after orchidopexy.
Collapse
Affiliation(s)
- Carolina J Jorgez
- Scott Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Surgery, Texas Children's Hospital, Houston, TX 77030, USA
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Abhishek Seth
- Scott Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Surgery, Texas Children's Hospital, Houston, TX 77030, USA
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nathan Wilken
- Scott Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Juan C Bournat
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ching H Chen
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dolores J Lamb
- Scott Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Brady Urology Department, Center for Reproductive Genomics and Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
14
|
GA R, Cheemakurthi R, Kalagara M, Prathigudupu K, Balabomma KL, Mahapatro P, Thota S, Kommaraju AL, Muvvala SPR. Effect of LHCGR Gene Polymorphism (rs2293275) on LH Supplementation Protocol Outcomes in Second IVF Cycles: A Retrospective Study. Front Endocrinol (Lausanne) 2021; 12:628169. [PMID: 34046009 PMCID: PMC8147863 DOI: 10.3389/fendo.2021.628169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/31/2021] [Indexed: 11/24/2022] Open
Abstract
Infertility is a major concern for couples wanting to have progeny. Despite recent advances in the field of IVF, success rates still need improvement. Understanding the patient's variability and addressing it with personalized interventions may improve the success rate of fertilization and live births. This study examined the impact of a personalized pharmacogenomic approach on LH supplementation on the pregnancy and live birth rate outcomes in comparison with the traditional approaches. 193 patients undergoing a second IVF cycle in Krishna IVF Clinic received LH supplementation either as per the conventional methods or based on N312S (rs2293275) LHCGR gene polymorphism. Results showed a significant increase in pregnancy rate (P-value: 0.049) and a trend showing improvement in live birth rates (P-value: 0.082) when r-hLH supplementation protocol was decided as per the genotypes A/A, A/G, and G/G of the N312S variant in the respective patients. This stimulation regimen helped in providing optimum levels of r-hLH supplementation to patients with impaired hormone-receptor interacting activity, to achieve higher success in pregnancy and live birth rates.
Collapse
Affiliation(s)
- Ramaraju GA
- Center for Assisted Reproduction, Krishna IVF Clinic, Visakhapatnam, India
- Department of Biotechnology, Institute of Science, Gitam (Deemed to be) University, Visakhapatnam, India
- *Correspondence: Ramaraju GA,
| | | | - Madan Kalagara
- Center for Assisted Reproduction, Krishna IVF Clinic, Visakhapatnam, India
| | | | | | - Pranati Mahapatro
- Center for Assisted Reproduction, Krishna IVF Clinic, Visakhapatnam, India
| | - Sivanarayana Thota
- Center for Assisted Reproduction, Krishna IVF Clinic, Visakhapatnam, India
| | - Aruna Lakshmi Kommaraju
- Department of Biotechnology, Institute of Science, Gitam (Deemed to be) University, Visakhapatnam, India
| | | |
Collapse
|
15
|
Acién P, Acién M. Disorders of Sex Development: Classification, Review, and Impact on Fertility. J Clin Med 2020; 9:jcm9113555. [PMID: 33158283 PMCID: PMC7694247 DOI: 10.3390/jcm9113555] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/20/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022] Open
Abstract
In this review, the elements included in both sex determination and sex differentiation are briefly analyzed, exposing the pathophysiological and clinical classification of disorders or anomalies of sex development. Anomalies in sex determination without sex ambiguity include gonadal dysgenesis, polysomies, male XX, and Klinefelter syndrome (dysgenesis and polysomies with a female phenotype; and sex reversal and Klinefelter with a male phenotype). Other infertility situations could also be included here as minor degrees of dysgenesis. Anomalies in sex determination with sex ambiguity should (usually) include testicular dysgenesis and ovotesticular disorders. Among the anomalies in sex differentiation, we include: (1) males with androgen deficiency (MAD) that correspond to those individuals whose karyotype and gonads are male (XY and testes), but the phenotype can be female due to different hormonal abnormalities. (2) females with androgen excess (FAE); these patients have ovaries and a 46,XX karyotype, but present varying degrees of external genital virilization as a result of an enzyme abnormality that affects adrenal steroid biosynthesis and leads to congenital adrenal hyperplasia; less frequently, this can be caused by iatrogenia or tumors. (3) Kallman syndrome. All of these anomalies are reviewed and analyzed herein, as well as related fertility problems.
Collapse
Affiliation(s)
- Pedro Acién
- Department of Gynecology, Miguel Hernández University, San Juan Campus, 03550 San Juan, Alicante, Spain;
- Correspondence: ; Tel.: +34-670-097-518, +34-965-919-385; Fax: +34-965-919-550
| | - Maribel Acién
- Department of Gynecology, Miguel Hernández University, San Juan Campus, 03550 San Juan, Alicante, Spain;
- Obstetrics and Gynecology, San Juan University Hospital, San Juan Campus, 03550 San Juan, Alicante, Spain
| |
Collapse
|
16
|
Novel SNPs in the SPAG11 gene and association with testicular biometric variables in Boer goats and application of the levelled-container technique. Anim Reprod Sci 2019; 208:106113. [PMID: 31405472 DOI: 10.1016/j.anireprosci.2019.106113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 04/19/2019] [Accepted: 06/26/2019] [Indexed: 11/22/2022]
Abstract
Testicular volume (TV) is one of the most important traits used in evaluation of the reproductive capacity of male animals. The levelled-container used in the present study was found to be reliable instrument to measure TV, based on a water displacement method. Sperm-associated antigen 11 (SPAG11) is an important gene that affects male reproductive performance. An objective of the present study, therefore, was to determine if single nucleotide polymorphisms (SNPs) in a fragment of the SPAG11 gene could be used to determine associations with values of testicular biometric variables in Boer goats. Primers were designed to amplify the full length of the first two exons of SPAG11. The targeted fragment was generated using a molecular cloning technique. As the result, four SNPs, [g.1256A > G(ss19199134542), g.1270C > T(ss19199134541), g.1325A > G(ss19199134540) and g.1327 G > A (ss19199134543)], were detected using a single-base extension (SBE) method. Two of these SNPs were synonymous (ss19199134540 and ss19199134542). The other two SNPs were nonsynonymous, thus, there were changes in amino acid in the resulting protein: threonine to isoleucine (for ss19199134541) and arginine to glutamine (for ss19199134543). The SNP ss19199134543 was the only locus detected that was associated with TV (P = 0.002). None of the testes dimensions nor TW were associated with detected SPAG11 gene SNPs. Most likely, the ss19199134543 locus affects tissue structures adjacent to the testes, causing the change in TV. In conclusion, among the studied testicular biometric variables, TV had the greatest potential for preselecting of bucks with desirable semen quality. The use of the levelled-container as a TV measurement approach was an accurate and reliable method.
Collapse
|
17
|
Qiao J, Han B. Diseases caused by mutations in luteinizing hormone/chorionic gonadotropin receptor. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 161:69-89. [PMID: 30711030 DOI: 10.1016/bs.pmbts.2018.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Accumulating evidence showed that the luteinizing hormone/chorionic gonadotropin receptor (LHCGR) is an essential regulator of sexual development and reproduction from zebrafish to human. Activating and inactivating mutations of LHCGR gene have been identified from patients of different phenotypes. Familial male-limited precocious puberty, Leydig cell hypoplasia, and empty follicle syndrome are caused by LHCGR mutations. More than 50 mutations have been reported from subjects of different ethnic backgrounds. Functional analyses of the mutant LHCGR revealed multiple defects, including cell surface expression, ligand binding, and signaling. The difference of the two native ligands and signaling pathway activated by LHCGR are illustrated. Potential therapeutic implications from the analyses of the naturally occurring LHCGR mutations, such as pharmacological chaperones, are highlighted.
Collapse
Affiliation(s)
- Jie Qiao
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
| | - Bing Han
- Department of Endocrinology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
18
|
Casarini L, Santi D, Brigante G, Simoni M. Two Hormones for One Receptor: Evolution, Biochemistry, Actions, and Pathophysiology of LH and hCG. Endocr Rev 2018; 39:549-592. [PMID: 29905829 DOI: 10.1210/er.2018-00065] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/08/2018] [Indexed: 01/03/2023]
Abstract
LH and chorionic gonadotropin (CG) are glycoproteins fundamental to sexual development and reproduction. Because they act on the same receptor (LHCGR), the general consensus has been that LH and human CG (hCG) are equivalent. However, separate evolution of LHβ and hCGβ subunits occurred in primates, resulting in two molecules sharing ~85% identity and regulating different physiological events. Pituitary, pulsatile LH production results in an ~90-minute half-life molecule targeting the gonads to regulate gametogenesis and androgen synthesis. Trophoblast hCG, the "pregnancy hormone," exists in several isoforms and glycosylation variants with long half-lives (hours) and angiogenic potential and acts on luteinized ovarian cells as progestational. The different molecular features of LH and hCG lead to hormone-specific LHCGR binding and intracellular signaling cascades. In ovarian cells, LH action is preferentially exerted through kinases, phosphorylated extracellular-regulated kinase 1/2 (pERK1/2) and phosphorylated AKT (also known as protein kinase B), resulting in irreplaceable proliferative/antiapoptotic signals and partial agonism on progesterone production in vitro. In contrast, hCG displays notable cAMP/protein kinase A (PKA)-mediated steroidogenic and proapoptotic potential, which is masked by estrogen action in vivo. In vitro data have been confirmed by a large data set from assisted reproduction, because the steroidogenic potential of hCG positively affects the number of retrieved oocytes, and LH affects the pregnancy rate (per oocyte number). Leydig cell in vitro exposure to hCG results in qualitatively similar cAMP/PKA and pERK1/2 activation compared with LH and testosterone. The supposed equivalence of LH and hCG has been disproved by such data, highlighting their sex-specific functions and thus deeming it an oversight caused by incomplete understanding of clinical data.
Collapse
Affiliation(s)
- Livio Casarini
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Genomic Research, University of Modena and Reggio Emilia, 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, Modena, Italy
| | - 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, Modena, Italy
| | - Manuela Simoni
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy.,Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Modena, Italy
| |
Collapse
|
19
|
Robeva R, Andonova S, Tomova A, Kumanov P, Savov A. LHCG receptor polymorphisms in PCOS patients. BIOTECHNOL BIOTEC EQ 2018. [DOI: 10.1080/13102818.2017.1423246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Ralitsa Robeva
- Medical Faculty, Clinical Center of Endocrinology and Gerontology, Medical University of Sofia, Sofia, Bulgaria
| | - Silvia Andonova
- Medical Faculty, National Genetic Laboratory, USHATOG “Maichin Dom”, Medical University of Sofia, Sofia, Bulgaria
| | - Analia Tomova
- Medical Faculty, Clinical Center of Endocrinology and Gerontology, Medical University of Sofia, Sofia, Bulgaria
| | - Philip Kumanov
- Medical Faculty, Clinical Center of Endocrinology and Gerontology, Medical University of Sofia, Sofia, Bulgaria
| | - Alexey Savov
- Medical Faculty, National Genetic Laboratory, USHATOG “Maichin Dom”, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
20
|
Liu S, Lian S, Yang Y, Fu C, Ma H, Xiong Z, Ling Y, Zhao C. The relationship between the variants in the 5'-untranslated regions of equine chorionic gonadotropin genes and serum equine chorionic gonadotropin levels. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2017; 30:1679-1683. [PMID: 28423874 PMCID: PMC5666169 DOI: 10.5713/ajas.17.0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/23/2017] [Accepted: 03/30/2017] [Indexed: 11/27/2022]
Abstract
Objective An experiment was conducted to study the association between the single nucleotide polymorphisms (SNPs) in 5′-untranslated regions (5′-UTR) of equine chorionic gonadotropin (eCG) genes and the serum eCG levels. Methods SNPs in 5′-UTR of eCG genes were screened across 10 horse breeds, including 7 Chinese indigenous breeds and 3 imported breeds using iPLEX chemistry, and the association between the serum eCG levels of 174 pregnant Da’an mares and their serum eCG levels (determined with ELISA) was analyzed. Results Four SNPs were identified in the 5′-UTR of the eCGα gene, and one of them was unique in the indigenous breeds. There were 2 SNPs detected at the 5′ end of the eCGβ subunit gene, and one of them was only found in the Chinese breeds. The SNP g.39948246T>C at the 5′-UTR of eCGα was associated significantly with eCG levels of 75-day pregnant mare serum (p<0.05) in Da’an mares. Prediction analysis on binding sites of transcription factors showed that the g.39948246T>C mutation causes appearance of the specific binding site of hepatocyte nuclear factor 3 forkhead homolog 2 (HFH-2), which is a transcriptional repressor belonging to the forkhead protein family of transcription factors. Conclusion The SNP g.39948246T>C at the 5′-UTR of eCGα is associated with eCG levels of 75-day pregnant mare serum (p<0.05).
Collapse
Affiliation(s)
- ShuQin Liu
- Equine Center, China Agricultural University, Beijing 100193, China
| | - Song Lian
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - YunZhou Yang
- Equine Center, China Agricultural University, Beijing 100193, China
| | - ChunZheng Fu
- Equine Center, China Agricultural University, Beijing 100193, China
| | - HongYing Ma
- Equine Center, China Agricultural University, Beijing 100193, China
| | - ZhiYao Xiong
- Equine Center, China Agricultural University, Beijing 100193, China
| | - Yao Ling
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - ChunJiang Zhao
- Equine Center, China Agricultural University, Beijing 100193, China.,College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, 100193, China.,National Engineering Laboratory for Animal Breeding, Beijing 100193, China
| |
Collapse
|
21
|
Genetics of gonadotropins and their receptors as markers of ovarian reserve and response in controlled ovarian stimulation. Best Pract Res Clin Obstet Gynaecol 2017; 44:15-25. [PMID: 28506471 DOI: 10.1016/j.bpobgyn.2017.04.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 02/03/2017] [Accepted: 04/01/2017] [Indexed: 01/11/2023]
Abstract
Several controlled ovarian stimulation (COS) protocols have been developed to increase the yield of mature oocytes retrieved in assisted reproductive techniques (ARTs). The ovarian reserve (OR) influences the COS response, and it represents the main parameter that helps clinicians in refining clinical treatments in the perspective of a "personalized" ART. This approach is even more needed in particular conditions such as poor OR or polycystic ovary syndrome. Follicle-stimulating hormone, luteinizing hormone, and human chorionic gonadotropin are currently used in COS at different combinations and with different efficacies, even if the best approach definition is controversial. Differences in individual-specific ovarian response to gonadotropin stimulation can be due to alterations of genes encoding for hormones or their receptors. In particular, FSHB c.-211G>T, FSHR p.Asn680Ser, and c.-29G>A SNP allelic combinations may be used as OR and COS response markers. The purpose of this review is to highlight the evidence-based relevance of mutations and polymorphisms in gonadotropins and their receptor genes as predictive markers of OR and COS response to achieve fine-tuned therapeutic regimens.
Collapse
|
22
|
Polymorphism in the Alternative Donor Site of the Cryptic Exon of LHCGR: Functional Consequences and Associations with Testosterone Level. Sci Rep 2017; 7:45699. [PMID: 28367994 PMCID: PMC5377329 DOI: 10.1038/srep45699] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 03/03/2017] [Indexed: 11/09/2022] Open
Abstract
Selective splicing is a feature of luteinizing hormone receptor (LHCGR). A cryptic exon (LHCGR-exon 6A) was found to be derived from alternative splicing in intron 6 of the LHCGR gene, which including two transcripts LHCGR-exon 6A-long and LHCGR-exon 6A-short. We addressed the functional consequences of SNP rs68073206, located at the +5 position of an alternative 5′ splice donor site, and observed its association with male infertility in the subjects with azoospermia, oligoasthenozoospermia and normozoospermia. The translation product of splicing variant LHCGR-exon 6A was expressed in the cytoplasm and exhibited no affinity with [125I]-hCG. No dominant negative effect was observed in cells co-expressed with LHCGR-exon 6A and wild-type LHCGR. The long transcript (LHCGR-exon 6A-long) was significantly elevated in the granulosa cells with G/G genotypes, which could be reproduced in vitro by mini-gene construct transfection. Genotyping analysis showed no association between rs68073206 and male infertility. However, this polymorphism was significantly associated with testosterone levels in normozoospermic subjects (n = 210). In conclusion, SNP rs68073206 in the splicing site of the cryptic exon 6A of the LHCGR gene affect the splicing pattern in the gene, which may play a role in the modulation of the LHCGR sensitivity in the gonads.
Collapse
|
23
|
Bang AK, Nordkap L, Almstrup K, Priskorn L, Petersen JH, Rajpert-De Meyts E, Andersson AM, Juul A, Jørgensen N. Dynamic GnRH and hCG testing: establishment of new diagnostic reference levels. Eur J Endocrinol 2017; 176:379-391. [PMID: 28077499 DOI: 10.1530/eje-16-0912] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/05/2016] [Accepted: 01/11/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Gonadotropin-releasing hormone (GnRH) and human chorionic gonadotropin (hCG) stimulation tests may be used to evaluate the pituitary and testicular capacity. Our aim was to evaluate changes in follicular-stimulating hormone (FSH), luteinizing hormone (LH) and testosterone after GnRH and hCG stimulation in healthy men and assess the impact of six single nucleotide polymorphisms on the responses. DESIGN GnRH and hCG stimulation tests were performed on 77 healthy men, 18-40 years (reference group) at a specialized andrology referral center at a university hospital. The potential influence of the tests was illustrated by results from 45 patients suspected of disordered hypothalamic-pituitary-gonadal axis. METHODS Baseline, stimulated, relative and absolute changes in serum FSH and LH were determined by ultrasensitive TRIFMA, and testosterone was determined by LC-MS/MS. RESULTS For the reference group, LH and FSH increased almost 400% and 40% during GnRH testing, stimulated levels varied from 4.4 to 58.8 U/L and 0.2 to 11.8 U/L and FSH decreased in nine men. Testosterone increased approximately 110% (range: 18.7-67.6 nmol/L) during hCG testing. None of the polymorphisms had any major impact on the test results. Results from GnRH and hCG tests in patients compared with the reference group showed that the stimulated level and absolute increase in LH showed superior identification of patients compared with the relative increase, and the absolute change in testosterone was superior in identifying men with Leydig cell insufficiency, compared with the relative increase. CONCLUSIONS We provide novel reference ranges for GnRH and hCG test in healthy men, which allows future diagnostic evaluation of hypothalamic-pituitary-gonadal disorders in men.
Collapse
Affiliation(s)
- A Kirstine Bang
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Loa Nordkap
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Kristian Almstrup
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Lærke Priskorn
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Jørgen Holm Petersen
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
- Department of BiostatisticsUniversity of Copenhagen, Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Anna-Maria Andersson
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Anders Juul
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| | - Niels Jørgensen
- Department of Growth and ReproductionRigshospitalet, University of Copenhagen, Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC)Rigshospitalet, Denmark
| |
Collapse
|
24
|
|
25
|
Lindgren I, Bååth M, Uvebrant K, Dejmek A, Kjaer L, Henic E, Bungum M, Bungum L, Cilio C, Leijonhufvud I, Skouby S, Andersen CY, Giwercman YL. Combined assessment of polymorphisms in the LHCGR and FSHR genes predict chance of pregnancy after in vitro fertilization. Hum Reprod 2016; 31:672-83. [PMID: 26769719 DOI: 10.1093/humrep/dev342] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 12/17/2015] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Can gonadotrophin receptor variants separately or in combination, be used for the prediction of pregnancy chances in in vitro fertilization (IVF) trials? SUMMARY ANSWER The luteinizing hormone/human chorionic gonadotrophin receptor (LHCGR) variant N312S and the follicle-stimulating hormone receptor (FSHR) variant N680S can be utilized for the prediction of pregnancy chances in women undergoing IVF. WHAT IS KNOWN ALREADY The FSHR N680S polymorphism has been shown to affect the ovarian response in response to gonadotrophin treatment, while no information is currently available regarding variants of the LHCGR in this context. STUDY DESIGN, SIZE, DURATION Cross-sectional study, duration from September 2010 to February 2015. Women undergoing IVF were consecutively enrolled and genetic variants compared between those who became pregnant and those who did not. The study was subsequently replicated in an independent sample. Granulosa cells from a subset of women were investigated regarding functionality of the genetic variants. PARTICIPANTS/MATERIALS, SETTING, METHODS Women undergoing IVF (n = 384) were enrolled in the study and genotyped. Clinical variables were retrieved from medical records. For replication, an additional group of n = 233 women was utilized. Granulosa cells from n = 135 women were isolated by flow cytometry, stimulated with Follitropin alpha or Menotropin, and the downstream targets 3',5'-cyclic adenosine monophosphate (cAMP) and inositol 1,4,5-trisphosphate (IP3) measured with enzyme-linked immunosorbent assay. MAIN RESULTS AND THE ROLE OF CHANCE Women homozygous for serine (S) in both polymorphisms displayed higher pregnancy rates than women homozygous asparagine (N) (OR = 14.4, 95% CI: [1.65, 126], P = 0.016). Higher pregnancy rates were also evident for women carrying LHCGR S312, regardless of FSHR variant (OR = 1.61, 95% CI: [1.13, 2.29], P = 0.008). These women required higher doses of FSH for follicle recruitment than women homozygous N (161 versus 148 IU, P = 0.030). When combining the study cohort with the replication cohort (n = 606), even stronger associations with pregnancy rates were noted for the combined genotypes (OR = 11.5, 95% CI: [1.86, 71.0], P = 0.009) and for women carrying LHCGR S312 (OR = 1.49, 95% CI: [1.14, 1.96], P = 0.004). A linear significant trend with pregnancy rate and increasing number of G alleles was also evident in the merged study population (OR = 1.34, 95% CI: [1.10, 1.64], P = 0.004). A lower cAMP response in granulosa cells was noted following Follitropin alpha stimulation for women homozygous N in both polymorphisms, compared with women with other genotypes (0.901 pmol cAMP/mg total protein versus 2.19 pmol cAMP/mg total protein, P = 0.035). LIMITATIONS, REASONS FOR CAUTION Due to racial differences in LHCGR genotype distribution, these results may not be applicable for all populations. WIDER IMPLICATIONS OF THE FINDINGS Despite that >250 000 cycles of gonadotrophin stimulations are performed annually worldwide prior to IVF, it has not been possible to predict neither the pregnancy outcome, nor the response to the hormone with accuracy. If LHCGR and FSHR variants are recognized as biomarkers for chance of pregnancy, more individualized and thereby more efficient treatment modalities can be developed. STUDY FUNDING, COMPETING INTERESTS This work was supported by Interreg IV A, EU (grant 167158) and ALF governments grant (F2014/354). Merck-Serono (Darmstadt, Germany) supported the enrollment of the subjects. The authors declare no conflict of interest.
Collapse
Affiliation(s)
- I Lindgren
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 20502 Malmö, Sweden
| | - M Bååth
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 20502 Malmö, Sweden
| | - K Uvebrant
- Department of Clinical Sciences, Cellular Autoimmunity Unit, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 20502 Malmö, Sweden
| | - A Dejmek
- Department of Clinical Pathology, University and Regional Laboratories, Skåne University Hospital, Jan Waldenströms gata 59, SE 20502 Malmö, Sweden
| | - L Kjaer
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 20502 Malmö, Sweden
| | - E Henic
- Reproductive Medicine Centre, Skåne University Hospital, Jan Waldenströms gata 47, Plan 3, SE 20502 Malmö, Sweden
| | - M Bungum
- Reproductive Medicine Centre, Skåne University Hospital, Jan Waldenströms gata 47, Plan 3, SE 20502 Malmö, Sweden
| | - L Bungum
- Department of Obstetrics and Gynecology, Herlev Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - C Cilio
- Department of Clinical Sciences, Cellular Autoimmunity Unit, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 20502 Malmö, Sweden
| | - I Leijonhufvud
- Reproductive Medicine Centre, Skåne University Hospital, Jan Waldenströms gata 47, Plan 3, SE 20502 Malmö, Sweden
| | - S Skouby
- Department of Obstetrics and Gynecology, Herlev Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - C Yding Andersen
- Laboratory of Reproductive Biology, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Y Lundberg Giwercman
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 20502 Malmö, Sweden
| |
Collapse
|
26
|
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
|
27
|
Aschim EL, Oldenburg J, Kristiansen W, Giwercman A, Witczak O, Fosså SD, Haugen TB. Genetic variations associated with the effect of testicular cancer treatment on gonadal hormones. Hum Reprod 2014; 29:2844-51. [PMID: 25336703 DOI: 10.1093/humrep/deu274] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Do genetic variations in the testosterone pathway genes modify the effect of treatment on the levels of testosterone and LH in long-term testicular cancer (TC) survivors (TCSs)? SUMMARY ANSWER Variations in LH receptor (LHR) and in 5α-reductase II (SRD5A2) genes may modify the effect of TC treatment on testosterone levels, whereas genetic variations in the androgen receptor (AR) may modify the effect on LH levels. WHAT IS KNOWN ALREADY TCSs experience variable degrees of long-term reduction in gonadal function after treatment. This variability can in part be explained by treatment intensity, but may also be due to individual variations in genes involved in the function and metabolism of reproductive hormones. STUDY DESIGN, SIZE, DURATION Cross-sectional study on testosterone and LH levels in 637 Norwegian TCSs in relation to genetic variants and TC treatment. PARTICIPANTS/MATERIALS, SETTING, METHODS The single nucleotide polymorphisms LHR Asn291Ser (rs12470652) and Ser312Asn (rs2293275), as well as SRD5A2 Ala49Thr (rs9282858) and Val89Leu (rs523349) were analyzed by allele-specific PCR. The insertion polymorphism LHR InsLQ (rs4539842) was analyzed by sequencing. The numbers of AR CAG and GGN repeats were determined by capillary electrophoresis. Blood samples were collected 5-21 years after diagnosis (median 11 years) and serum total testosterone and LH were analyzed by commercial immunoassays. The TCSs were divided into four groups according to their treatment; surgery only, radiotherapy and chemotherapy with ≤850 or >850 mg of cisplatin. Polymorphisms presenting P < 0.1 for the interaction term with treatment in an initial two-way analysis of covariance (ANCOVA) were investigated further in two consecutive one-way ANCOVA analyses to elucidate the interaction between treatment and genotype. MAIN RESULTS AND THE ROLE OF CHANCE For the whole group of TCSs, there were no significant differences between the hormone levels in homozygotes for the wild type and carriers of at least one polymorphic allele for the investigated polymorphisms. Three of the polymorphisms showed signs of interaction with treatment, i.e. LHR InsLQ, SRD5A2 A49T and the AR CAG repeat. Follow-up analyses revealed three situations where only one of the genotypes of the polymorphism where associated with significantly different hormone levels after surgery compared with after additional cytotoxic treatment: For LHR InsLQ, only the wild-type allele was associated with lower testosterone levels after cisplatin > 850 mg compared with after surgery (24% lower, P < 0.001). For SRD5A2 A49T, testosterone levels were lower after radiotherapy compared with after surgery, but only for the heterozygotes for the polymorphism (39% lower, P = 0.001). In comparison, the testosterone levels were just slightly lower after radiotherapy (6% lower, P = 0.039) or cisplatin ≤ 850 mg (7% lower, P = 0.041), compared with surgery, independent of genotypes. For AR CAG, only the reference length of CAG = 21-22 had significantly higher LH levels after cisplatin ≤ 850 mg compared with after surgery (70% higher, P < 0.001). Independent of genotypes, however, LH levels after cisplatin ≤ 850 mg were only 26% higher than after surgery (P = 0.005). LIMITATIONS, REASONS FOR CAUTION Unadjusted P-values are presented. For analysis involving genotypes, the level of statistical significance was adjusted for the total number of polymorphisms tested, n = 7, i.e. to P < 0.007 (0.5/7). The rather weak associations indicate that additional polymorphisms are involved in the modulation. WIDER IMPLICATIONS OF THE FINDINGS To our knowledge, this is the first study supporting the notion that polymorphisms may explain at least some of the inter-individual differences in endocrine response to TC treatment. Our findings suggest that individuals with certain genotypes may be more vulnerable to certain treatments. Knowledge on genetic predisposition concerning treatment-related endocrine gonadotoxicity to different treatment regimens may help tailoring TC therapy when possible. STUDY FUNDING/COMPETING INTERESTS This study was supported by the Research Council of Norway (Grant No. 160619). There were no competing interests.
Collapse
Affiliation(s)
- E L Aschim
- Faculty of Health Sciences, Oslo and Akershus University College of Applied Sciences, P.O. Box 4, St. Olavs plass, Oslo NO-0130, Norway
| | - J Oldenburg
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo NO-0424, Norway
| | - W Kristiansen
- Faculty of Health Sciences, Oslo and Akershus University College of Applied Sciences, P.O. Box 4, St. Olavs plass, Oslo NO-0130, Norway
| | - A Giwercman
- Molecular Reproduction Research, Department of Clinical Sciences Malmö, Lund University, Malmö SE-205 02, Sweden
| | - O Witczak
- Faculty of Health Sciences, Oslo and Akershus University College of Applied Sciences, P.O. Box 4, St. Olavs plass, Oslo NO-0130, Norway
| | - S D Fosså
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo NO-0424, Norway
| | - T B Haugen
- Faculty of Health Sciences, Oslo and Akershus University College of Applied Sciences, P.O. Box 4, St. Olavs plass, Oslo NO-0130, Norway
| |
Collapse
|
28
|
Raju GAR, Chavan R, Deenadayal M, Gunasheela D, Gutgutia R, Haripriya G, Govindarajan M, Patel NH, Patki AS. Luteinizing hormone and follicle stimulating hormone synergy: A review of role in controlled ovarian hyper-stimulation. J Hum Reprod Sci 2014; 6:227-34. [PMID: 24672160 PMCID: PMC3963304 DOI: 10.4103/0974-1208.126285] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 09/14/2013] [Accepted: 11/06/2013] [Indexed: 01/13/2023] Open
Abstract
Luteinizing hormone (LH) in synergy with follicle stimulating hormone (FSH) stimulates normal follicular growth and ovulation. FSH is frequently used in assisted reproductive technology (ART). Recent studies have facilitated better understanding on the complementary role of the LH to FSH in regulation of the follicle; however, role of LH in stimulation of follicle, optimal dosage of LH in stimulation and its importance in advanced aged patients has been a topic of discussion among medical fraternity. Though the administration of exogenous LH with FSH is obligatory for controlled ovarian stimulation in patients with hypogonadotropic hypogonadism, there is still a paucity of information of its usage in other patient population. In this review we looked in to the multiple roles that LH plays complementary to FSH to better understand the LH requirement in patients undergoing ART.
Collapse
Affiliation(s)
| | - Rahul Chavan
- Department of Pharmacology Medical Affairs, Merck Specialities Private Limited, Mumbai, Maharashtra, India
| | - Mamata Deenadayal
- Department of Reproductive medicine, Infertility Institute and Research Centre, Secunderabad, Andhra Pradesh, India
| | - Devika Gunasheela
- Department of Obstetrics, Gynaecology & Infertility, Gunasheela Assisted Reproduction Centre, Bengaluru, Karnataka, India
| | - Rohit Gutgutia
- Department of Reproductive Medicine, NOVA IVI Fertility, Kolkata, West Bengal, India
| | - Geetha Haripriya
- Department of Reproductive Medicine, Prashanth Fertility Research Centre, Chennai, India
| | - Mirudhubashini Govindarajan
- Department of Obstetrics, Gynaecology & Reproductive Medicine, Womens Center and Hospitals, Coimbatore, Tamil Nadu, India
| | | | | |
Collapse
|
29
|
Bakircioglu ME, Tulay P, Findikli N, Erzik B, Gultomruk M, Bahceci M. Successful testicular sperm recovery and IVF treatment in a man with Leydig cell hypoplasia. J Assist Reprod Genet 2014; 31:817-21. [PMID: 24792890 DOI: 10.1007/s10815-014-0241-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/21/2014] [Indexed: 11/30/2022] Open
Affiliation(s)
- M E Bakircioglu
- Bahceci Assisted Reproductive Technology Centre, Kosuyolu Caddesi, No. 26-28 34,718, Kadikoy, Istanbul, Turkey,
| | | | | | | | | | | |
Collapse
|
30
|
Ramaswamy S, Weinbauer GF. Endocrine control of spermatogenesis: Role of FSH and LH/ testosterone. SPERMATOGENESIS 2014; 4:e996025. [PMID: 26413400 PMCID: PMC4581062 DOI: 10.1080/21565562.2014.996025] [Citation(s) in RCA: 240] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 12/04/2014] [Indexed: 12/21/2022]
Abstract
Evaluation of testicular functions (production of sperm and androgens) is an important aspect of preclinical safety assessment and testicular toxicity is comparatively far more common than ovarian toxicity. This chapter focuses (1) on the histological sequelae of disturbed reproductive endocrinology in rat, dog and nonhuman primates and (2) provides a review of our current understanding of the roles of gonadotropins and androgens. The response of the rodent testis to endocrine disturbances is clearly different from that of dog and primates with different germ cell types and spermatogenic stages being affected initially and also that the end-stage spermatogenic involution is more pronounced in dog and primates compared to rodents. Luteinizing hormone (LH)/testosterone and follicle-stimulating hormone (FSH) are the pivotal endocrine factors controlling testicular functions. The relative importance of either hormone is somewhat different between rodents and primates. Generally, however, both LH/testosterone and FSH are necessary for quantitatively normal spermatogenesis, at least in non-seasonal species.
Collapse
Affiliation(s)
- Suresh Ramaswamy
- Center for Research in Reproductive Physiology (CRRP); Department of Obstetrics, Gynecology & Reproductive Sciences; University of Pittsburgh School of Medicine; Magee-Womens Research Institute; Pittsburgh, PA USA
| | | |
Collapse
|
31
|
Hotaling J, Carrell DT. Clinical genetic testing for male factor infertility: current applications and future directions. Andrology 2014; 2:339-50. [PMID: 24711280 DOI: 10.1111/j.2047-2927.2014.00200.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 01/24/2014] [Accepted: 02/03/2014] [Indexed: 01/06/2023]
Abstract
Spermatogenesis involves the aggregated action of up to 2300 genes, any of which, could, potentially, provide targets for diagnostic tests of male factor infertility. Contrary to the previously proposed common variant hypothesis for common diseases such as male infertility, genome-wide association studies and targeted gene sequencing in cohorts of infertile men have identified only a few gene polymorphisms that are associated with male infertility. Unfortunately, the search for genetic variants associated with male infertility is further hampered by the lack of viable animal models of human spermatogenesis, difficulty in robustly phenotyping infertile men and the complexity of pedigree studies in male factor infertility. In this review, we describe basic genetic principles involved in understanding the genetic basis of male infertility and examine the utility and proper clinical use of the proven genetic assays of male factor infertility, specifically Y chromosome microdeletions, chromosomal translocations, karyotype, cystic fibrosis transmembrane conductance regulator mutation analysis and sperm genetic tests. Unfortunately, these tests are only able to diagnose the cause of about 20% of male factor infertility. The remainder of the review will be devoted to examining novel tests and diagnostic tools that have the potential to explain the other 80% of male factor infertility that is currently classified as idiopathic. Those tests include epigenetic analysis of the spermatozoa and the evaluation of rare genetic variants and copy number variations in patients. Success in advancing to the implementation of such areas is not only dependent on technological advances in the laboratory, but also improved phenotyping in the clinic.
Collapse
Affiliation(s)
- J Hotaling
- Department of Surgery (Urology), University of Utah School of Medicine, Salt Lake City, UT, USA
| | | |
Collapse
|
32
|
Choi J, Smitz J. Luteinizing hormone and human chorionic gonadotropin: origins of difference. Mol Cell Endocrinol 2014; 383:203-13. [PMID: 24365330 DOI: 10.1016/j.mce.2013.12.009] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/06/2013] [Accepted: 12/12/2013] [Indexed: 01/24/2023]
Abstract
Luteinizing hormone (LH) and human chorionic gonadotropin (hCG) are widely recognized for their roles in ovulation and the support of early pregnancy. Aside from the timing of expression, however, the differences between LH and hCG have largely been overlooked in the clinical realm because of their similar molecular structures and shared receptor. With technologic advancements, including the development of highly purified and recombinant gonadotropins, researchers now appreciate that these hormones are not as interchangeable as once believed. Although they bind to a common receptor, emerging evidence suggests that LH and hCG have disparate effects on downstream signaling cascades. Increased understanding of the inherent differences between LH and hCG will foster more effective diagnostic and prognostic assays for use in a variety of clinical contexts and support the individualization of treatment strategies for conditions such as infertility.
Collapse
Affiliation(s)
- Janet Choi
- The Center for Women's Reproductive Care at Columbia University, New York, NY, United States.
| | - Johan Smitz
- UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
33
|
O’Brien TJ, Kalmin MM, Harralson AF, Clark AM, Gindoff I, Simmens SJ, Frankfurter D, Gindoff P. Association between the luteinizing hormone/chorionic gonadotropin receptor (LHCGR) rs4073366 polymorphism and ovarian hyperstimulation syndrome during controlled ovarian hyperstimulation. Reprod Biol Endocrinol 2013; 11:71. [PMID: 23883350 PMCID: PMC3727944 DOI: 10.1186/1477-7827-11-71] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 07/22/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aim of this study was to determine the relationship between a purported luteinizing hormone/chorionic gonadotropin (LHCGR) high function polymorphism (rs4539842/insLQ) and outcome to controlled ovarian hyperstimulation (COH). METHODS This was a prospective study of 172 patients undergoing COH at the Fertility and IVF Center at GWU. DNA was isolated from blood samples and a region encompassing the insLQ polymorphism was sequenced. We also investigated a polymorphism (rs4073366 G > C) that was 142 bp from insLQ. The association of the insLQ and rs4073366 alleles and outcome to COH (number of mature follicles, estradiol level on day of human chorionic gonadotropin (hCG) administration, the number of eggs retrieved and ovarian hyperstimulation syndrome (OHSS)) was determined. RESULTS Increasing age and higher day 3 (basal) FSH levels were significantly associated with poorer response to COH. We found that both insLQ and rs4073366 were in linkage disequilibrium (LD) and no patients were homozygous for both recessive alleles (insLQ/insLQ; C/C). The insLQ variant was not significantly associated with any of the main outcomes to COH. Carrier status for the rs4073366 C variant was associated (P = 0.033) with an increased risk (OR 2.95, 95% CI = 1.09-7.96) of developing OHSS. CONCLUSIONS While age and day 3 FSH levels were predictive of outcome, we found no association between insLQ and patient response to COH. Interestingly, rs4073366 C variant carrier status was associated with OHSS risk. To the best of our knowledge, this is the first report suggesting that LHCGR genetic variation might function in patient risk for OHSS.
Collapse
Affiliation(s)
- Travis J O’Brien
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
| | - Mariah M Kalmin
- Department of Epidemiology and Biostatistics, The George Washington University, Washington, DC, USA
| | - Arthur F Harralson
- Department of Pharmacogenomics, Bernard J. Dunn School of Pharmacy, Shenandoah University, Ashburn, VA, USA
| | - Adam M Clark
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
| | - Ian Gindoff
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
| | - Samuel J Simmens
- Department of Epidemiology and Biostatistics, The George Washington University, Washington, DC, USA
| | - David Frankfurter
- Department of Obstetrics and Gynecology, The George Washington University, Washington, DC, USA
| | - Paul Gindoff
- Department of Obstetrics and Gynecology, The George Washington University, Washington, DC, USA
| |
Collapse
|
34
|
Cannistraci CV, Ogorevc J, Zorc M, Ravasi T, Dovc P, Kunej T. Pivotal role of the muscle-contraction pathway in cryptorchidism and evidence for genomic connections with cardiomyopathy pathways in RASopathies. BMC Med Genomics 2013; 6:5. [PMID: 23410028 PMCID: PMC3626861 DOI: 10.1186/1755-8794-6-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 02/06/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cryptorchidism is the most frequent congenital disorder in male children; however the genetic causes of cryptorchidism remain poorly investigated. Comparative integratomics combined with systems biology approach was employed to elucidate genetic factors and molecular pathways underlying testis descent. METHODS Literature mining was performed to collect genomic loci associated with cryptorchidism in seven mammalian species. Information regarding the collected candidate genes was stored in MySQL relational database. Genomic view of the loci was presented using Flash GViewer web tool (http://gmod.org/wiki/Flashgviewer/). DAVID Bioinformatics Resources 6.7 was used for pathway enrichment analysis. Cytoscape plug-in PiNGO 1.11 was employed for protein-network-based prediction of novel candidate genes. Relevant protein-protein interactions were confirmed and visualized using the STRING database (version 9.0). RESULTS The developed cryptorchidism gene atlas includes 217 candidate loci (genes, regions involved in chromosomal mutations, and copy number variations) identified at the genomic, transcriptomic, and proteomic level. Human orthologs of the collected candidate loci were presented using a genomic map viewer. The cryptorchidism gene atlas is freely available online: http://www.integratomics-time.com/cryptorchidism/. Pathway analysis suggested the presence of twelve enriched pathways associated with the list of 179 literature-derived candidate genes. Additionally, a list of 43 network-predicted novel candidate genes was significantly associated with four enriched pathways. Joint pathway analysis of the collected and predicted candidate genes revealed the pivotal importance of the muscle-contraction pathway in cryptorchidism and evidence for genomic associations with cardiomyopathy pathways in RASopathies. CONCLUSIONS The developed gene atlas represents an important resource for the scientific community researching genetics of cryptorchidism. The collected data will further facilitate development of novel genetic markers and could be of interest for functional studies in animals and human. The proposed network-based systems biology approach elucidates molecular mechanisms underlying co-presence of cryptorchidism and cardiomyopathy in RASopathies. Such approach could also aid in molecular explanation of co-presence of diverse and apparently unrelated clinical manifestations in other syndromes.
Collapse
Affiliation(s)
- Carlo V Cannistraci
- Integrative Systems Biology Laboratory, Biological and Environmental Sciences and Engineering Division, Computational Bioscience Research Center, King Abdullah University for Science and Technology, Thuwal, Saudi Arabia.
| | | | | | | | | | | |
Collapse
|
35
|
Kristiansen W, Aschim EL, Andersen JM, Witczak O, Fosså SD, Haugen TB. Variations in testosterone pathway genes and susceptibility to testicular cancer in Norwegian men. ACTA ACUST UNITED AC 2012; 35:819-827. [DOI: 10.1111/j.1365-2605.2012.01297.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
36
|
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
|
37
|
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
|
38
|
Carrell DT, Aston KI. The search for SNPs, CNVs, and epigenetic variants associated with the complex disease of male infertility. Syst Biol Reprod Med 2011; 57:17-26. [PMID: 21208142 DOI: 10.3109/19396368.2010.521615] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Understanding the genetic basis of idiopathic male infertility has long been the focus of many researchers. Numerous recent studies have attempted to identify relevant single nucleotide polymorphisms (SNPs) through medical re-sequencing studies in which candidate genes are sequenced in large numbers of cases and controls in the search for risk or causative polymorphisms. Two major characteristics have limited the utility of the re-sequencing studies. First, reported SNPs have only accounted for a small percentage of idiopathic male infertility. Second, SNPs reported to have an association with male infertility based on gene re-sequencing studies often fail validation in follow-up studies. Recent advances in the tools available for genetic studies have enabled interrogation of the entire genome in search of common, and more recently, rare variants. In this review, we discuss the progress of studies on genetic and epigenetic variants of male infertility as well as future directions that we predict will be the most productive in identifying the genetic basis for male factor infertility based on our current state of knowledge in this field as well as lessons learned about the genetic basis for complex diseases from other disease models.
Collapse
Affiliation(s)
- Douglas T Carrell
- Andrology and IVF Laboratories, Department of Surgery, University of Utah School of Medicine, Salt Lake City, Utah 84108, USA.
| | | |
Collapse
|
39
|
Zhou W, Bolden-Tiller OU, Shetty G, Shao SH, Weng CC, Pakarinen P, Liu Z, Stivers DN, Meistrich ML. Changes in gene expression in somatic cells of rat testes resulting from hormonal modulation and radiation-induced germ cell depletion. Biol Reprod 2009; 82:54-65. [PMID: 19684331 DOI: 10.1095/biolreprod.109.078048] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Although gonadotropins and androgen are required for normal spermatogenesis and both testosterone and follicle-stimulating hormone (FSH) are responsible for the inhibition of spermatogonial differentiation that occurs in irradiated rats, it has been difficult to identify the specific genes involved. To study specific hormonally regulated changes in somatic cell gene expression in the testis that may be involved in these processes, without the complication of changing populations of germ cells, we used irradiated LBNF(1) rats, the testes of which contain almost exclusively somatic cells except for a few type A spermatogonia. Three different groups of these rats were treated with various combinations of gonadotropin-releasing hormone antagonist, an androgen receptor antagonist (flutamide), testosterone, and FSH, and we compared the gene expression levels 2 wk later to those of irradiated-only rats by microarray analysis. By dividing the gene expression patterns into three major patterns and 11 subpatterns, we successfully distinguished, in a single study, the genes that were specifically regulated by testosterone, by luteinizing hormone (LH), and by FSH from the large number of genes that were not hormonally regulated in the testis. We found that hormones produced more dramatic upregulation than downregulation of gene expression: Testosterone had the strongest upregulatory effect, LH had a modest but appreciable upregulatory effect, and FSH had a minor upregulatory effect. We also separately identified the somatic cell genes that were chronically upregulated by irradiation. Thus, the present study identified gene expression changes that may be responsible for hormonal action on somatic cells to support normal spermatogenesis and the hormone-mediated block in spermatogonial development after irradiation.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Experimental Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Bibliography. Current world literature. Curr Opin Obstet Gynecol 2009; 21:296-300. [PMID: 19458522 DOI: 10.1097/gco.0b013e32832c972c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This bibliography is compiled by clinicians from the journals listed at the end of this publication. It is based on literature entered into our database between 1 February 2008 and 31 January 2009 (articles are generally added to the database about two and a half months after publication). In addition, the bibliography contains every paper annotated by reviewers; these references were obtained from a variety of bibliographic databases and published between the beginning of the review period and the time of going to press. The bibliography has been grouped into topics that relate to the reviews in this issue.
Collapse
|
41
|
Valkenburg O, Uitterlinden A, Piersma D, Hofman A, Themmen A, de Jong F, Fauser B, Laven J. Genetic polymorphisms of GnRH and gonadotrophic hormone receptors affect the phenotype of polycystic ovary syndrome. Hum Reprod 2009; 24:2014-22. [DOI: 10.1093/humrep/dep113] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|