1
|
Bohaczuk SC, Tonsfeldt KJ, Slaiwa TI, Dunn GA, Gillette DLM, Yeo SE, Shi C, Cassin J, Thackray VG, Mellon PL. A Point Mutation in an Otherwise Dispensable Upstream Fshb Enhancer Moderately Impairs Fertility in Female Mice. Endocrinology 2025; 166:bqaf073. [PMID: 40237337 PMCID: PMC12038155 DOI: 10.1210/endocr/bqaf073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/26/2025] [Accepted: 04/12/2025] [Indexed: 04/18/2025]
Abstract
Follicle-stimulating hormone (FSH) is necessary for fertility in both sexes as a regulator of gametogenesis and hormone synthesis. Humans with loss-of-function mutations within the gene encoding the FSH beta subunit (FSHB) are infertile. Similarly, female Fshb knock-out mice are infertile and fail to ovulate, and males are subfertile. We recently reported the discovery and characterization of an upstream enhancer of FSHB located 26 Kb upstream of the transcriptional start site in humans (-17 Kb in mouse) that also amplifies activin and gonadotropin-releasing hormone induction of FSHB. Notably, the upstream enhancer contains a polymorphic, fertility-associated site in humans, rs11031006 (G/A), and the minor allele (A) increased enhancer activity in vitro as compared to the major allele (G), likely by increasing the affinity of an SF1 binding element. To investigate the role of the novel enhancer and rs11031006 variant in vivo, we created mouse models to assess deletion of the upstream enhancer and the impact of the G>A point mutation at the rs11031006-equivalent base. A full characterization of the -17 Kb enhancer deletion model revealed no apparent differences in fertility or serum FSH/LH levels, nor did a larger deletion that also included an additional putative regulatory element. In contrast, female mice homozygous for the mutated A allele at the rs11031006-equivalent position had fewer litters over a 120-day fertility assay, abnormal estrous cycling at 10 months, and reduced pituitary Lhb transcript abundance. Overall, while the mouse -17 Kb Fshb enhancer is dispensable for fertility, the rs11031006-equivalent G>A mutation results in subfertility in females.
Collapse
Affiliation(s)
- Stephanie C Bohaczuk
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Karen J Tonsfeldt
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Theresa I Slaiwa
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Geneva A Dunn
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dominique L M Gillette
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Seung E Yeo
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chengxian Shi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jessica Cassin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Varykina G Thackray
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
2
|
Mani S, Srivastava V, Shandilya C, Kaushik A, Singh KK. Mitochondria: the epigenetic regulators of ovarian aging and longevity. Front Endocrinol (Lausanne) 2024; 15:1424826. [PMID: 39605943 PMCID: PMC11598335 DOI: 10.3389/fendo.2024.1424826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Ovarian aging is a major health concern for women. Ovarian aging is associated with reduced health span and longevity. Mitochondrial dysfunction is one of the hallmarks of ovarian aging. In addition to providing oocytes with optimal energy, the mitochondria provide a co-substrate that drives epigenetic processes. Studies show epigenetic alterations, both nuclear and mitochondrial contribute to ovarian aging. Both, nuclear and mitochondrial genomes cross-talk with each other, resulting in two ways orchestrated anterograde and retrograde response that involves epigenetic changes in nuclear and mitochondrial compartments. Epigenetic alterations causing changes in metabolism impact ovarian function. Key mitochondrial co-substrate includes acetyl CoA, NAD+, ATP, and α-KG. Thus, enhancing mitochondrial function in aging ovaries may preserve ovarian function and can lead to ovarian longevity and reproductive and better health outcomes in women. This article describes the role of mitochondria-led epigenetics involved in ovarian aging and discusses strategies to restore epigenetic reprogramming in oocytes by preserving, protecting, or promoting mitochondrial function.
Collapse
Affiliation(s)
- Shalini Mani
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Vidushi Srivastava
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Chesta Shandilya
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Aditi Kaushik
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Keshav K. Singh
- Departments of Genetics, Dermatology and Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Center for Women’s Reproductive Health, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
3
|
Xu X, Liu Y, Feng W, Shen J. Strong evidence supports the use of estradiol therapy for the treatment of vaginal inflammation: a two-way Mendelian randomization study. Eur J Med Res 2024; 29:339. [PMID: 38890725 PMCID: PMC11186076 DOI: 10.1186/s40001-024-01914-4] [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: 10/27/2023] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE Nowadays, there has been limited Mendelian randomization (MR) research focusing on the causal relationship between estradiol and vaginitis. Therefore, this study conducted a two-way MR study to clarify the causal effect and related influencing factors between them. METHODS All genetic datasets were obtained using publicly available summary statistics based on individuals of European ancestry from the IEU GWAS database. MR analysis was performed using MR-Egger, weighted median (WM) and inverse variance weighted (IVW) methods to assess the causal relationship between exposure and outcome and to validate the findings by comprehensively evaluating the effects of pleiotropic effects and outliers. RESULTS MR analysis revealed no significant causal relationship between estradiol and vaginitis risk. There was a negative correlation between estradiol and age at menarche (IVW, OR: 0.9996, 95% CI: 0.9992-1.0000, P = 0.0295; WM, OR: 0.9995, 95% CI: 0.9993-0.9998, P = 0.0003), and there was a positive correlation between age at menarche and vaginitis (IVW, OR: 1.5108, 95% CI: 1.1474-2.0930, P = 0.0043; MR-Egger, OR: 2.5575, 95% CI: 1.7664-9.6580, P = 0.0013). Estradiol was negatively correlated with age at menopause (IVW, OR: 0.9872, 95% CI: 0.9786-0.9959, P = 0.0041). However, there was no causal relationship between age at menopause and vaginitis (P > 0.05). In addition, HPV E7 Type 16, HPV E7 Type 18, and Lactobacillus had no direct causal effects on estradiol and vaginitis (P > 0.05). Sensitivity analyses revealed no heterogeneity and horizontal pleiotropy. CONCLUSION When estrogen levels drop, it will lead to a later age of menarche, and a later age of menarche may increase the risk of vaginitis, highlighting that the longer the female reproductive tract receives estrogen stimulation, the stronger the defense ability is formed, and the prevalence of vaginitis is reduced. In conclusion, this study indirectly supports an association between reduced level of estrogen or short time of estrogen stimulation and increased risk of vaginitis.
Collapse
Affiliation(s)
- Xiaosheng Xu
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medcine, 197 Ruijiner Road, Shanghai, 200003, China
| | - Yan Liu
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medcine, 197 Ruijiner Road, Shanghai, 200003, China
| | - Weiwei Feng
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medcine, 197 Ruijiner Road, Shanghai, 200003, China.
| | - Jian Shen
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medcine, 197 Ruijiner Road, Shanghai, 200003, China.
| |
Collapse
|
4
|
Ecochard R, Stanford JB, Fehring RJ, Schneider M, Najmabadi S, Gronfier C. Evidence that the woman's ovarian cycle is driven by an internal circamonthly timing system. SCIENCE ADVANCES 2024; 10:eadg9646. [PMID: 38598621 PMCID: PMC11006216 DOI: 10.1126/sciadv.adg9646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/06/2024] [Indexed: 04/12/2024]
Abstract
The ovarian cycle has a well-established circa-monthly rhythm, but the mechanisms involved in its regularity are unknown. Is the rhythmicity driven by an endogenous clock-like timer or by other internal or external processes? Here, using two large epidemiological datasets (26,912 cycles from 2303 European women and 4786 cycles from 721 North American women), analyzed with time series and circular statistics, we find evidence that the rhythmic characteristics of the menstrual cycle are more likely to be explained by an endogenous clock-like driving mechanism than by any other internal or external process. We also show that the menstrual cycle is weakly but significantly influenced by the 29.5-day lunar cycle and that the phase alignment between the two cycles differs between the European and the North American populations. Given the need to find efficient treatments of subfertility in women, our results should be confirmed in larger populations, and chronobiological approaches to optimize the ovulatory cycle should be evaluated.
Collapse
Affiliation(s)
- René Ecochard
- Pôle de Santé Publique, Service de Biostatistique, Hospices Civils de Lyon, Lyon 69424 Cedex 03, France
- Laboratoire Biostatistique Santé, Université Claude Bernard Lyon I, UMR CNRS 5558 UCBL, Lyon 69000, France
| | - John B. Stanford
- Office of Cooperative Reproductive Health, Division of Public Health, Department of Family and Preventive Medicine, University of Utah, Salt Lake City, 84108 UT, USA
| | - Richard J. Fehring
- College of Nursing, Marquette University, Milwaukee, P.O. Box 1881 WI, USA
| | - Marie Schneider
- College of Nursing, Marquette University, Milwaukee, P.O. Box 1881 WI, USA
- Institute for Natural Family Planning, Milwaukee, P.O. Box 1881 WI, USA
| | - Sam Najmabadi
- Office of Cooperative Reproductive Health, Division of Public Health, Department of Family and Preventive Medicine, University of Utah, Salt Lake City, 84108 UT, USA
| | - Claude Gronfier
- Centre de Recherche en Neurosciences de Lyon (CRNL), Neurocampus, Inserm U1028, CNRS UMR5292, Université de Lyon, Lyon 69500, France
| |
Collapse
|
5
|
Ebong IA, Michos ED, Wilson M, Appiah D, Schreiner PJ, Racette SB, Allison M, Watson K, Bertoni A. Adipokines and adiposity among postmenopausal women of the Multi-Ethnic Study of Atherosclerosis. Menopause 2024; 31:209-217. [PMID: 38270904 PMCID: PMC10989717 DOI: 10.1097/gme.0000000000002261] [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] [Indexed: 01/26/2024]
Abstract
OBJECTIVE We investigated whether the associations of serum adiponectin, leptin, and resistin with adiposity differ with menopausal age. METHODS In this cross-sectional study, we included 751 postmenopausal women from the Multi-Ethnic Study of Atherosclerosis (MESA) who reported their menopausal age (<45, 45-49, 50-54 and ≥55 y) and had anthropometrics, serum adipokines, and abdominal computed tomography measures of visceral and subcutaneous adipose tissue (VAT and SAT) obtained at MESA exam 2 or 3. Linear regression models were used for analysis. RESULTS The mean ± SD age was 65.1 ± 9.0 years for all participants. The median (interquartile range) values for serum adiponectin, leptin and resistin, VAT, and SAT were 21.9 (14.8-31.7) ng/L, 24.3 (12.5-42.4) pg/L, 15.3 (11.8-19.5) pg/L, 183.9 (130.8-251.1) cm2, and 103.7 (65.6-151.5) cm2, respectively. The mean ± SD values for body mass index, waist circumference, and waist-to-hip ratio were 28.3 ± 5.81 kg/m2, 96.6 ± 15.9 cm, and 0.91 ± 0.078, respectively. Adiponectin was inversely associated with all adiposity measures, with similar patterns across menopausal age categories. Leptin was positively associated with all adiposity measures, and the strength of associations varied across menopausal age categories for body mass index, waist circumference, and SAT (Pinteraction ≤ 0.01 for all). The associations of resistin with adiposity measures were mostly nonsignificant except in the 45- to 49-year menopausal age category. CONCLUSIONS Menopausal age category had no influence on the association of serum adiponectin with adiposity. The association of serum leptin and resistin differed according to menopausal age category for generalized adiposity but was inconsistent for measures of abdominal adiposity.
Collapse
Affiliation(s)
- Imo A. Ebong
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California Davis, Sacramento, CA
| | - Erin D. Michos
- Division of Cardiology, John Hopkins University School of Medicine, Baltimore, MD
| | - Machelle Wilson
- Department of Public Health Sciences, Division of Biostatistics, University of California Davis, Sacramento, CA
| | - Duke Appiah
- Department of Public Health, Texas Tech University Health Sciences Center, Lubbock, TX
| | | | - Susan B. Racette
- College of Health Solutions, Arizona State University, Phoenix, AZ
| | - Matthew Allison
- Family Medicine and Public Health, University of California San Diego, San Diego, CA
| | - Karol Watson
- Division of Cardiovascular Medicine, University of California Los Angeles, Los Angeles, CA
| | - Alain Bertoni
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston Salem, NC
| |
Collapse
|
6
|
Amiri M, Rahmati M, Firouzi F, Azizi F, Ramezani Tehran F. A prospective study on the relationship between polycystic ovary syndrome and age at natural menopause. Menopause 2024; 31:130-137. [PMID: 38411437 DOI: 10.1097/gme.0000000000002213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
OBJECTIVE This study aimed to determine whether polycystic ovary syndrome (PCOS) was associated with age at menopause, compared with women without PCOS, after adjusting for potential confounders. METHODS A total of 1,696 reproductive-aged participants from the Tehran Lipid and Glucose Study were included in this population-based prospective study with a follow-up of approximately 20 years. Of these, 348 women with PCOS based on the Rotterdam criteria and 1,348 non-PCOS controls were followed to assess the age at which they reached menopause. An accelerated failure time survival regression model was used to identify the association between PCOS and the age at natural menopause (ANM), with and without adjustment for potential confounders. RESULTS The unadjusted accelerated failure time survival model revealed a significant positive association between PCOS and ANM; PCOS women experienced time to menopause by a factor of 1.05 than non-PCOS controls (95% confidence interval, 1.02-1.06; P < 0.001). After adjusting for age at baseline, menarche age, history of hypertension, history of type 2 diabetes mellitus, parity, oral contraceptive use, body mass index, education level, physical activity, and smoking, the results remained significant (time ratio: 1.03; 95% confidence interval, 1.01-1.06; P = 0.002). CONCLUSIONS This study indicates that ANM is significantly associated with PCOS in women. Our study findings may have implications for the fertility and reproductive health of women with PCOS. However, further large longitudinal studies on diverse populations accounting for other relevant confounders are still needed to provide data on the actual difference in age at menopause and to elucidate the underlying mechanisms of this association.
Collapse
Affiliation(s)
| | - Maryam Rahmati
- From the Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faegheh Firouzi
- Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fahimeh Ramezani Tehran
- From the Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Liang J, Ali F, Ramaiyer M, Borahay MA. Determinants and Assessment of Menstrual Blood Flow. CURR EPIDEMIOL REP 2023; 10:210-220. [PMID: 38275001 PMCID: PMC10810143 DOI: 10.1007/s40471-023-00332-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 01/27/2024]
Abstract
Purpose of review a)The modifiable and non-modifiable determinants and the currently available methods of assessment of menstrual blood flow will be discussed, with the goal of helping healthcare providers, researchers, and those interested in public health. Recent findings b)Several factors can impact menstruation. The determinants include modifiable factors such as smoking, nutrition, exercise, stress, weight fluctuation, and benign gynecologic diseases, and non-modifiable factors such as age, race, and the individual's genes. The intertwined dynamic among these determinants needs more critical attention. Currently, the methods for the assessment of menstruation all have advantages and disadvantages, often with a tradeoff between practicality and accuracy. Summary c)Considered by many as the fifth vital, menstruation provides a window to an individual's general health. The discussion of its determinants and assessment can be more appropriate for individual contexts, especially from a public health perspective as it can improve the reproductive health of the population.
Collapse
Affiliation(s)
- Jinxiao Liang
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Gynecologic Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fiza Ali
- Johns Hopkins University, Krieger School of Arts and Sciences, Baltimore, Maryland, USA
| | - Malini Ramaiyer
- Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mostafa A. Borahay
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Bianco B, Loureiro FA, Trevisan CM, Christofolini DM, Laganà AS, Barbosa CP. Implication of FSHB rs10835638 variant in endometriosis in Brazilian women. EINSTEIN-SAO PAULO 2023; 21:eAO0483. [PMID: 37909652 PMCID: PMC10586852 DOI: 10.31744/einstein_journal/2023ao0483] [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: 02/24/2023] [Accepted: 06/12/2023] [Indexed: 11/03/2023] Open
Abstract
OBJECTIVE The follicle-stimulating hormone subunit beta gene rs10835638 variant (c.-211G>T) may have detrimental effects on fertility and protective effects against endometriosis. A case-control analysis was performed, aiming to investigate the possible relationship between this variant and the development and/or progression of endometriosis. METHODS This study included 326 women with endometriosis and 482 controls without endometriosis, both confirmed by inspection of the pelvic cavity during surgery. Genotyping was performed using a TaqMan real-time polymerase chain reaction assay. Genotype and allele frequencies and genetic models were compared between the groups. RESULTS The genotype and allele frequencies of the rs10835638 variant did not differ between women with and those without endometriosis. Subdividing the endometriosis group into fertile and infertile groups did not result in a significant difference in these frequencies. However, the subgroup with minimal/mild endometriosis had a higher frequency of the GT genotype than the Control Group, regardless of fertility. The T allele was significantly more common in women with minimal/mild endometriosis than in the Control Group in the recessive model. CONCLUSION The T allele is associated with the development of minimal/mild endometriosis in Brazilian women.
Collapse
Affiliation(s)
- Bianca Bianco
- Centro Universitário FMABCSanto AndréSPBrazil Centro Universitário FMABC, Santo André, SP, Brazil.
| | - Flávia Altheman Loureiro
- Centro Universitário FMABCSanto AndréSPBrazil Centro Universitário FMABC, Santo André, SP, Brazil.
| | - Camila Martins Trevisan
- Centro Universitário FMABCSanto AndréSPBrazil Centro Universitário FMABC, Santo André, SP, Brazil.
| | | | - Antonio Simone Laganà
- Unit of Gynecologic Oncology, ARNAS “Civico-Di Cristina-Benfratelli”Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical SpecialtiesUniversity of PalermoPalermoItaly Unit of Gynecologic Oncology, ARNAS “Civico-Di Cristina-Benfratelli”, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.
| | - Caio Parente Barbosa
- Centro Universitário FMABCSanto AndréSPBrazil Centro Universitário FMABC, Santo André, SP, Brazil.
| |
Collapse
|
9
|
Shi Y, Miao BY, Ai XX, Cao P, Gao J, Xu Y, Yang Q, Fei J, Zhang Q, Mai QY, Wen YX, Qu YL, Zhou CQ, Xu YW. Identification of common genetic polymorphisms associated with down-regulated gonadotropin levels in an exome-wide association study. Fertil Steril 2023; 120:671-681. [PMID: 37001689 DOI: 10.1016/j.fertnstert.2023.03.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/17/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
OBJECTIVE To investigate whether common genetic polymorphisms are associated with gonadotropin levels after down-regulation with daily gonadotropin-releasing hormone agonist and whether the polymorphisms of candidate variants influence the ovarian response to exogenous gonadotropins. DESIGN Genetic association study. SETTING University-affiliated in vitro fertilization center. PATIENTS Subjects enrolled in an exploratory exome-wide association study (n = 862), a replication exome-wide association study (n = 86), and a classifier validation study (n = 148) were recruited from September 2016 to October 2018, September 2019 to September 2020, and January 2021 to December 2021, respectively. The included patients were aged ≤40 years and had a basal follicle-stimulating hormone (FSH) ≤12 IU/L. INTERVENTIONS All participants received a luteal phase down-regulation long protocol. Genome DNA was extracted from the peripheral blood leukocytes. For the exploratory and replication cohorts, exome sequencing was conducted on a HiSeq 2500 sequencing platform. The multiplex polymerase chain reaction amplification technique and next-generation sequencing also were performed in the exploratory and replication cohorts. For the samples of the validation cohort, Sanger sequencing was performed. MAIN OUTCOME MEASURES The primary endpoint was the gonadotropin levels after down-regulation, and the secondary endpoints were hormone levels and follicle diameters during stimulation, the total dose of FSH, duration of FSH stimulation, number of oocytes retrieved, and clinical pregnancy rate. RESULTS In the exploratory cohort, we identified that FSHB rs6169 (P=2.71 × 10-24) and its single-nucleotide polymorphisms in high linkage disequilibrium were associated with the down-regulated FSH level. The same locus was confirmed in the replication cohort. Women carrying the C allele of FSHB rs6169 exhibited higher average estradiol level during stimulation (P=6.82 × 10-5), shorter duration of stimulation, and less amount of exogenous FSH (Pduration=0.0002; Pdose=0.0024). In the independent validation set, adding rs6169 genotypes into the prediction model for FSH level after down-regulation enhanced the area under the curve from 0.560 to 0.712 in a logistic regression model, and increased prediction accuracy by 41.05% when a support vector machine classifier was applied. CONCLUSION The C allele of FSHB rs6169 is a susceptibility site for the relatively high level of FSH after down-regulation, which may be associated with increased ovarian FSH sensitivity.
Collapse
Affiliation(s)
- Yue Shi
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China
| | - Ben-Yu Miao
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China
| | - Xi-Xiong Ai
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China; Reproductive Medicine Center, The Affiliated Shenzhen Maternity and Child Healthcare Hospital of the South Medical University, Shenzhen, Guangdong, China
| | - Ping Cao
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China; Research School for Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands; Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Jun Gao
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China
| | - Yan Xu
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China
| | - Qun Yang
- Peking Medriv Academy of Genetics and Reproduction, Peking, China
| | - Jia Fei
- Peking Medriv Academy of Genetics and Reproduction, Peking, China
| | - Qian Zhang
- Peking Medriv Academy of Genetics and Reproduction, Peking, China
| | - Qing-Yun Mai
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China
| | - Yang-Xing Wen
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China
| | - Yan-Lin Qu
- Department of Management Science and Engineering, Stanford University, Stanford, California
| | - Can-Quan Zhou
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China
| | - Yan-Wen Xu
- Reproductive Medicine Center, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Rahmioglu N, Mortlock S, Ghiasi M, Møller PL, Stefansdottir L, Galarneau G, Turman C, Danning R, Law MH, Sapkota Y, Christofidou P, Skarp S, Giri A, Banasik K, Krassowski M, Lepamets M, Marciniak B, Nõukas M, Perro D, Sliz E, Sobalska-Kwapis M, Thorleifsson G, Topbas-Selcuki NF, Vitonis A, Westergaard D, Arnadottir R, Burgdorf KS, Campbell A, Cheuk CSK, Clementi C, Cook J, De Vivo I, DiVasta A, Dorien O, Donoghue JF, Edwards T, Fontanillas P, Fung JN, Geirsson RT, Girling JE, Harkki P, Harris HR, Healey M, Heikinheimo O, Holdsworth-Carson S, Hostettler IC, Houlden H, Houshdaran S, Irwin JC, Jarvelin MR, Kamatani Y, Kennedy SH, Kepka E, Kettunen J, Kubo M, Kulig B, Kurra V, Laivuori H, Laufer MR, Lindgren CM, MacGregor S, Mangino M, Martin NG, Matalliotaki C, Matalliotakis M, Murray AD, Ndungu A, Nezhat C, Olsen CM, Opoku-Anane J, Padmanabhan S, Paranjpe M, Peters M, Polak G, Porteous DJ, Rabban J, Rexrode KM, Romanowicz H, Saare M, Saavalainen L, Schork AJ, Sen S, Shafrir AL, Siewierska-Górska A, Słomka M, Smith BH, Smolarz B, Szaflik T, Szyłło K, Takahashi A, Terry KL, Tomassetti C, Treloar SA, Vanhie A, Vincent K, Vo KC, Werring DJ, Zeggini E, Zervou MI, DBDS Genomic Consortium, et alRahmioglu N, Mortlock S, Ghiasi M, Møller PL, Stefansdottir L, Galarneau G, Turman C, Danning R, Law MH, Sapkota Y, Christofidou P, Skarp S, Giri A, Banasik K, Krassowski M, Lepamets M, Marciniak B, Nõukas M, Perro D, Sliz E, Sobalska-Kwapis M, Thorleifsson G, Topbas-Selcuki NF, Vitonis A, Westergaard D, Arnadottir R, Burgdorf KS, Campbell A, Cheuk CSK, Clementi C, Cook J, De Vivo I, DiVasta A, Dorien O, Donoghue JF, Edwards T, Fontanillas P, Fung JN, Geirsson RT, Girling JE, Harkki P, Harris HR, Healey M, Heikinheimo O, Holdsworth-Carson S, Hostettler IC, Houlden H, Houshdaran S, Irwin JC, Jarvelin MR, Kamatani Y, Kennedy SH, Kepka E, Kettunen J, Kubo M, Kulig B, Kurra V, Laivuori H, Laufer MR, Lindgren CM, MacGregor S, Mangino M, Martin NG, Matalliotaki C, Matalliotakis M, Murray AD, Ndungu A, Nezhat C, Olsen CM, Opoku-Anane J, Padmanabhan S, Paranjpe M, Peters M, Polak G, Porteous DJ, Rabban J, Rexrode KM, Romanowicz H, Saare M, Saavalainen L, Schork AJ, Sen S, Shafrir AL, Siewierska-Górska A, Słomka M, Smith BH, Smolarz B, Szaflik T, Szyłło K, Takahashi A, Terry KL, Tomassetti C, Treloar SA, Vanhie A, Vincent K, Vo KC, Werring DJ, Zeggini E, Zervou MI, DBDS Genomic Consortium, FinnGen Study, The Celmatix Research Team, The 23andMe Research Team, Adachi S, Buring JE, Ridker PM, D’Hooghe T, Goulielmos GN, Hapangama DK, Hayward C, Horne AW, Low SK, Martikainen H, Chasman DI, Rogers PAW, Saunders PT, Sirota M, Spector T, Strapagiel D, Tung JY, Whiteman DC, Giudice LC, Velez-Edwards DR, Uimari O, Kraft P, Salumets A, Nyholt DR, Mägi R, Stefansson K, Becker CM, Yurttas-Beim P, Steinthorsdottir V, Nyegaard M, Missmer SA, Montgomery GW, Morris AP, Zondervan KT. The genetic basis of endometriosis and comorbidity with other pain and inflammatory conditions. Nat Genet 2023; 55:423-436. [PMID: 36914876 PMCID: PMC10042257 DOI: 10.1038/s41588-023-01323-z] [Show More Authors] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/27/2023] [Indexed: 03/16/2023]
Abstract
Endometriosis is a common condition associated with debilitating pelvic pain and infertility. A genome-wide association study meta-analysis, including 60,674 cases and 701,926 controls of European and East Asian descent, identified 42 genome-wide significant loci comprising 49 distinct association signals. Effect sizes were largest for stage 3/4 disease, driven by ovarian endometriosis. Identified signals explained up to 5.01% of disease variance and regulated expression or methylation of genes in endometrium and blood, many of which were associated with pain perception/maintenance (SRP14/BMF, GDAP1, MLLT10, BSN and NGF). We observed significant genetic correlations between endometriosis and 11 pain conditions, including migraine, back and multisite chronic pain (MCP), as well as inflammatory conditions, including asthma and osteoarthritis. Multitrait genetic analyses identified substantial sharing of variants associated with endometriosis and MCP/migraine. Targeted investigations of genetically regulated mechanisms shared between endometriosis and other pain conditions are needed to aid the development of new treatments and facilitate early symptomatic intervention.
Collapse
Affiliation(s)
- Nilufer Rahmioglu
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Sally Mortlock
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Marzieh Ghiasi
- Department of Epidemiology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Peter L Møller
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | - Constance Turman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Rebecca Danning
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston MA, USA
| | - Matthew H Law
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Faculty of Health, and Institute of health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Paraskevi Christofidou
- Department of Twin Research and Genetic Epidemiology, St. Thomas’ Hospital, Kings College London, London, UK
| | - Sini Skarp
- Northern Finland Birth Cohorts, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Ayush Giri
- Department of Obstetrics and Gynecology, Institute of Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michal Krassowski
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Maarja Lepamets
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Błażej Marciniak
- Biobank Lab, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | - Margit Nõukas
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Danielle Perro
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Eeva Sliz
- Computational Medicine and Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Marta Sobalska-Kwapis
- Biobank Lab, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | | | - Nura F Topbas-Selcuki
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Allison Vitonis
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ragnheidur Arnadottir
- Department of Obstetrics and Gynecology, Landspitali University Hospital, Reykjavik, Iceland
| | - Kristoffer S Burgdorf
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics & Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Cecilia SK Cheuk
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | - James Cook
- Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Immaculata De Vivo
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Amy DiVasta
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
- Division of Adolescent and Young Adult Medicine, Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - O Dorien
- Department of Obstetrics and Gynaecology, Leuven University Fertility Centre, University Hospital Leuven, Leuven, Belgium
- KULeuven (University of Leuven), Department of Development and Regeneration, Organ systems, Leuven, Belgium
| | - Jacqueline F Donoghue
- University of Melbourne Department of Obstetrics and Gynaecology, Royal Women’s Hospital, Melbourne, Australia
| | - Todd Edwards
- Department of Obstetrics and Gynecology, Institute of Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Jenny N Fung
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Reynir T Geirsson
- Department of Obstetrics and Gynecology, Landspitali University Hospital, Reykjavik, Iceland
| | - Jane E Girling
- University of Melbourne Department of Obstetrics and Gynaecology, Royal Women’s Hospital, Melbourne, Australia
- Department of Anatomy, School of Biomedical Sciences, University of Otago, New Zealand
| | - Paivi Harkki
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Holly R Harris
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Martin Healey
- University of Melbourne Department of Obstetrics and Gynaecology, Royal Women’s Hospital, Melbourne, Australia
| | - Oskari Heikinheimo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sarah Holdsworth-Carson
- University of Melbourne Department of Obstetrics and Gynaecology, Royal Women’s Hospital, Melbourne, Australia
| | - Isabel C Hostettler
- Stroke Research Centre, University College London, Institute of Neurology, London, UK
- Neurogenetics Laboratory, The National Hospital of Neurology and Neurosurgery, London, UK
- Department of Neurosurgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Henry Houlden
- Neurogenetics Laboratory, The National Hospital of Neurology and Neurosurgery, London, UK
| | - Sahar Houshdaran
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Juan C Irwin
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Computational Medicine and Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, Oulu, Finland
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, Middlesex, UK
| | | | - Stephen H Kennedy
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Ewa Kepka
- Biobank Lab, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | - Johannes Kettunen
- Computational Medicine and Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Institute for Health and Welfare, Helsinki, Finland
| | - Michiaki Kubo
- Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Bartosz Kulig
- Department of Operative Gynecology and Oncological Gynecology, Polish Mother’s Memorial Hospital - Research Institute, Łódź, Poland
| | - Venla Kurra
- Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Hannele Laivuori
- Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Marc R Laufer
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
- Division of Adolescent and Young Adult Medicine, Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gynecology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Cecilia M Lindgren
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Queensland, Australia
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, St. Thomas’ Hospital, Kings College London, London, UK
- NIHR Biomedical Research Centre at Guy’s and St Thomas’ Foundation Trust, London, UK
| | - Nicholas G Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Charoula Matalliotaki
- Third Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Michail Matalliotakis
- Third Department of Obstetrics and Gynecology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alison D Murray
- The Institute of Medical Sciences, Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - Anne Ndungu
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Camran Nezhat
- Center For Special Minimally Invasive and Robotic Surgery, Camran Nezhat Institute, Palo Alto, CA, USA
| | - Catherine M Olsen
- Department of Population Health, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jessica Opoku-Anane
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Manish Paranjpe
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
| | - Maire Peters
- Institute of Clinical Medicine, Department of Obstetrics and Gynecology, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Grzegorz Polak
- 1st Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Poland
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics & Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Joseph Rabban
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Kathyrn M Rexrode
- Division of Women’s Health, Brigham and Women’s Hospital, Boston MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Hanna Romanowicz
- Laboratory of Cancer Genetics, Department of Clinical Pathomorphology, Polish Mother’s Memorial Hospital - Research Institute, Łódź, Poland
| | - Merli Saare
- Institute of Clinical Medicine, Department of Obstetrics and Gynecology, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Liisu Saavalainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Andrew J Schork
- Institute of Biological Psychiatry, Mental Health Center, Sct. Hans, Mental Health Services, Copenhagen, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
- Neurogenomics Division, The Translational Genomics Research Institute (TGEN), Phoenix, AZ, USA
| | - Sushmita Sen
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Amy L Shafrir
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
- Division of Adolescent and Young Adult Medicine, Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Anna Siewierska-Górska
- Computational Medicine and Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Marcin Słomka
- Computational Medicine and Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Blair H Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Beata Smolarz
- Laboratory of Cancer Genetics, Department of Clinical Pathomorphology, Polish Mother’s Memorial Hospital - Research Institute, Łódź, Poland
| | - Tomasz Szaflik
- Department of Operative Gynecology and Oncological Gynecology, Polish Mother’s Memorial Hospital - Research Institute, Łódź, Poland
| | - Krzysztof Szyłło
- Department of Operative Gynecology and Oncological Gynecology, Polish Mother’s Memorial Hospital - Research Institute, Łódź, Poland
| | - Atsushi Takahashi
- Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
- Research Institute, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Kathryn L Terry
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Carla Tomassetti
- Department of Obstetrics and Gynaecology, Leuven University Fertility Centre, University Hospital Leuven, Leuven, Belgium
- KULeuven (University of Leuven), Department of Development and Regeneration, Organ systems, Leuven, Belgium
| | - Susan A Treloar
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Arne Vanhie
- Department of Obstetrics and Gynaecology, Leuven University Fertility Centre, University Hospital Leuven, Leuven, Belgium
- KULeuven (University of Leuven), Department of Development and Regeneration, Organ systems, Leuven, Belgium
| | - Katy Vincent
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Kim C Vo
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - David J Werring
- Stroke Research Centre, University College London, Institute of Neurology, London, UK
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Wellcome Sanger Institute, Hinxton, United Kingdom
- TUM School of Medicine, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany
| | - Maria I Zervou
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | | | | | | | | | - Sosuke Adachi
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Julie E Buring
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Thomas D’Hooghe
- KULeuven (University of Leuven), Department of Development and Regeneration, Organ systems, Leuven, Belgium
- Global Medical Affairs Fertility, Research and Development, Merck, Darmstadt, Germany
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - George N Goulielmos
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Dharani K Hapangama
- Department of Women’s and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Andrew W Horne
- MRC Centre for Reproductive Health, University of Edinburgh, Institute for Regeneration and Repair, Edinburgh, UK
| | - Siew-Kee Low
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hannu Martikainen
- Department of Obstetrics and Gynecology, Oulu University Hospital, Oulu, Finland
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, Oulu, Finland
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Peter AW Rogers
- University of Melbourne Department of Obstetrics and Gynaecology, Royal Women’s Hospital, Melbourne, Australia
| | - Philippa T Saunders
- Centre for Inflammation Research, University of Edinburgh, Institute for Regeneration and Repair, Edinburgh, UK
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Tim Spector
- Department of Twin Research and Genetic Epidemiology, St. Thomas’ Hospital, Kings College London, London, UK
| | - Dominik Strapagiel
- Biobank Lab, Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Łódź, Poland
| | | | - David C Whiteman
- Department of Population Health, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Linda C Giudice
- Center for Reproductive Sciences, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Digna R Velez-Edwards
- Department of Obstetrics and Gynecology, Institute of Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Outi Uimari
- Department of Obstetrics and Gynecology, Oulu University Hospital, Oulu, Finland
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, Oulu, Finland
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andres Salumets
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Clinical Medicine, Department of Obstetrics and Gynecology, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Dale R Nyholt
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Faculty of Health, and Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kari Stefansson
- deCODE genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Christian M Becker
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | | | - Mette Nyegaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Health, Science and Technology, Aalborg University, Aalborg, Denmark
| | - Stacey A Missmer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
- Division of Adolescent and Young Adult Medicine, Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Obstetrics, Gynecology, and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Grant W Montgomery
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
| | - Krina T Zondervan
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
11
|
Wegrzynowicz AK, Beckley A, Eyvazzadeh A, Levy G, Park J, Klein J. Complete Cycle Mapping Using a Quantitative At-Home Hormone Monitoring System in Prediction of Fertile Days, Confirmation of Ovulation, and Screening for Ovulation Issues Preventing Conception. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1853. [PMID: 36557055 PMCID: PMC9783738 DOI: 10.3390/medicina58121853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Background and Objectives: To achieve pregnancy, it is highly beneficial to identify the time of ovulation as well as the greater period of fertile days during which sperm may survive leading up to ovulation. Confirming successful ovulation is also critical to accurately diagnose ovulatory disorders. Ovulation predictor kits, fertility monitors, and tracking apps are all available to assist with detecting ovulation, but often fall short. They may not detect the full fertile window, provide accurate or real-time information, or are simply expensive and impractical. Finally, few over-the-counter products provide information to women about their ovarian reserve and future fertility. Therefore, there is a need for an easy, over-the-counter, at-home quantitative hormone monitoring system that assesses ovarian reserve, predicts the entire fertile window, and can screen for ovulatory disorders. Materials and Methods: Proov Complete is a four-in-one at-home multihormone testing system that utilizes lateral flow assay test strips paired with the free Proov Insight App to guide testing of four hormones-FSH, E1G, LH, and PdG-across the woman's cycle. In a pilot study, 40 women (including 16 with a fertility-related diagnosis or using fertility treatments) used Complete for one cycle. Results: Here, we demonstrate that Proov Complete can accurately and sensitively predict ovarian reserve, detect up to 6 fertile days and confirm if ovulation was successful, in one easy-to-use kit. Ovulation was confirmed in 38 cycles with a detectable PdG rise. An average of 5.3 fertile days (from E1G rise to PdG rise) were detected, with an average of 2.7 days prior to LH surge. Ovulation was confirmed via PdG rise an average of 2.6 days following the LH surge. While 38/40 women had a PdG rise, only 22 had a sustained PdG level above 5 μg/mL throughout the critical implantation window, indicating ovulatory dysfunction in 16 women. Conclusions: Proov Complete can detect the entire fertile window of up to 6 fertile days and confirm ovulation, while also providing information on ovarian reserve and guidance to clinicians and patients.
Collapse
Affiliation(s)
- Andrea K. Wegrzynowicz
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- MFB Fertility, Inc., 720 Austin Ave Suite 100-305, Erie, CO 80516, USA
| | - Amy Beckley
- MFB Fertility, Inc., 720 Austin Ave Suite 100-305, Erie, CO 80516, USA
| | - Aimee Eyvazzadeh
- Aimee Eyvazzedeh MD, Inc., 5401 Norris Canyon Road, Suite 106, San Ramon, CA 94583, USA
| | - Gary Levy
- Fertility Cloud, Inc., 2100 Geng Rd, Palo Alto, CA 94303, USA
| | - John Park
- Carolina Conceptions, 2601 Lake Dr 301, Raleigh, NC 27607, USA
| | - Joshua Klein
- Extend Fertility, 200 W 57th St 1101, New York, NY 10019, USA
| |
Collapse
|
12
|
Dapas M, Dunaif A. Deconstructing a Syndrome: Genomic Insights Into PCOS Causal Mechanisms and Classification. Endocr Rev 2022; 43:927-965. [PMID: 35026001 PMCID: PMC9695127 DOI: 10.1210/endrev/bnac001] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Indexed: 01/16/2023]
Abstract
Polycystic ovary syndrome (PCOS) is among the most common disorders in women of reproductive age, affecting up to 15% worldwide, depending on the diagnostic criteria. PCOS is characterized by a constellation of interrelated reproductive abnormalities, including disordered gonadotropin secretion, increased androgen production, chronic anovulation, and polycystic ovarian morphology. It is frequently associated with insulin resistance and obesity. These reproductive and metabolic derangements cause major morbidities across the lifespan, including anovulatory infertility and type 2 diabetes (T2D). Despite decades of investigative effort, the etiology of PCOS remains unknown. Familial clustering of PCOS cases has indicated a genetic contribution to PCOS. There are rare Mendelian forms of PCOS associated with extreme phenotypes, but PCOS typically follows a non-Mendelian pattern of inheritance consistent with a complex genetic architecture, analogous to T2D and obesity, that reflects the interaction of susceptibility genes and environmental factors. Genomic studies of PCOS have provided important insights into disease pathways and have indicated that current diagnostic criteria do not capture underlying differences in biology associated with different forms of PCOS. We provide a state-of-the-science review of genetic analyses of PCOS, including an overview of genomic methodologies aimed at a general audience of non-geneticists and clinicians. Applications in PCOS will be discussed, including strengths and limitations of each study. The contributions of environmental factors, including developmental origins, will be reviewed. Insights into the pathogenesis and genetic architecture of PCOS will be summarized. Future directions for PCOS genetic studies will be outlined.
Collapse
Affiliation(s)
- Matthew Dapas
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Andrea Dunaif
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
13
|
Golovchenko I, Aizikovich B, Golovchenko O, Reshetnikov E, Churnosova M, Aristova I, Ponomarenko I, Churnosov M. Sex Hormone Candidate Gene Polymorphisms Are Associated with Endometriosis. Int J Mol Sci 2022; 23:13691. [PMID: 36430184 PMCID: PMC9697627 DOI: 10.3390/ijms232213691] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/07/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
The present study was designed to examine whether sex hormone polymorphisms proven by GWAS are associated with endometriosis risk. Unrelated female participants totaling 1376 in number (395 endometriosis patients and 981 controls) were recruited into the study. Nine single-nucleotide polymorphisms (SNPs) which GWAS correlated with circulating levels of sex hormones were genotyped using a TaqMan allelic discrimination assay. FSH-lowering, and LH- and testosterone-heightening polymorphisms of the FSHB promoter (allelic variants A rs11031002 and C rs11031005) exhibit a protective effect for endometriosis (OR = 0.60-0.68). By contrast, the TT haplotype loci that were GWAS correlated with higher FSH levels and lower LH and testosterone concentrations determined an increased risk for endometriosis (OR = 2.03). Endometriosis-involved epistatic interactions were found between eight loci of sex hormone genes (without rs148982377 ZNF789) within twelve genetic simulation models. In silico examination established that 8 disorder-related loci and 80 proxy SNPs are genome variants affecting the expression, splicing, epigenetic and amino acid conformation of the 34 genes which enrich the organic anion transport and secondary carrier transporter pathways. In conclusion, the present study showed that sex hormone polymorphisms proven by GWAS are associated with endometriosis risk and involved in the molecular pathophysiology of the disease due to their functionality.
Collapse
Affiliation(s)
- Ilya Golovchenko
- Department of Medical Biological Disciplines, Belgorod State University, 308015 Belgorod, Russia
| | - Boris Aizikovich
- Department of Fundamental Medicine, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Oleg Golovchenko
- Department of Obstetrics and Gynecology, Belgorod State University, 308015 Belgorod, Russia
| | - Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State University, 308015 Belgorod, Russia
| | - Maria Churnosova
- Department of Medical Biological Disciplines, Belgorod State University, 308015 Belgorod, Russia
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State University, 308015 Belgorod, Russia
| | - Irina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State University, 308015 Belgorod, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State University, 308015 Belgorod, Russia
| |
Collapse
|
14
|
Schubert M, Pérez Lanuza L, Wöste M, Dugas M, Carmona FD, Palomino-Morales RJ, Rassam Y, Heilmann-Heimbach S, Tüttelmann F, Kliesch S, Gromoll J. A GWAS in Idiopathic/Unexplained Infertile Men Detects a Genomic Region Determining Follicle-Stimulating Hormone Levels. J Clin Endocrinol Metab 2022; 107:2350-2361. [PMID: 35305013 PMCID: PMC9282256 DOI: 10.1210/clinem/dgac165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Approximately 70% of infertile men are diagnosed with idiopathic (abnormal semen parameters) or unexplained (normozoospermia) infertility, with the common feature of lacking etiologic factors. Follicle-stimulating hormone (FSH) is essential for initiation and maintenance of spermatogenesis. Certain single-nucleotide variations (SNVs; formerly single-nucleotide polymorphisms [SNPs]) (ie, FSHB c.-211G > T, FSHR c.2039A > G) are associated with FSH, testicular volume, and spermatogenesis. It is unknown to what extent other variants are associated with FSH levels and therewith resemble causative factors for infertility. OBJECTIVE We aimed to identify further genetic determinants modulating FSH levels in a cohort of men presenting with idiopathic or unexplained infertility. METHODS We retrospectively (2010-2018) selected 1900 men with idiopathic/unexplained infertility. In the discovery study (n = 760), a genome-wide association study (GWAS) was performed (Infinium PsychArrays) in association with FSH values (Illumina GenomeStudio, v2.0). Minor allele frequencies (MAFs) were analyzed for the discovery and an independent normozoospermic cohort. In the validation study (n = 1140), TaqMan SNV polymerase chain reaction was conducted for rs11031005 and rs10835638 in association with andrological parameters. RESULTS Imputation revealed 9 SNVs in high linkage disequilibrium, with genome-wide significance (P < 4.28e-07) at the FSHB locus 11p.14.1 being associated with FSH. The 9 SNVs accounted for up to a 4.65% variance in FSH level. In the oligozoospermic subgroup, this was increased up to 6.95% and the MAF was enhanced compared to an independent cohort of normozoospermic men. By validation, a significant association for rs11031005/rs10835638 with FSH (P = 4.71e-06/5.55e-07) and FSH/luteinizing hormone ratio (P = 2.08e-12/6.4e-12) was evident. CONCLUSIONS This GWAS delineates the polymorphic FSHB genomic region as the main determinant of FSH levels in men with unexplained or idiopathic infertility. Given the essential role of FSH, molecular detection of one of the identified SNVs that causes lowered FSH and therewith decreases spermatogenesis could resolve the idiopathic/unexplained origin by this etiologic factor.
Collapse
Affiliation(s)
| | | | - Marius Wöste
- Institute of Medical Informatics, University of Münster, Münster, North Rhine-Westphalia 48149, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Münster, Münster, North Rhine-Westphalia 48149, Germany
- Institute of Medical Informatics, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - F David Carmona
- Department of Genetics and Institute of Biotechnology, University of Granada, Granada, Andalusia 18016, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Andalusia 18012, Spain
| | - Rogelio J Palomino-Morales
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Andalusia 18012, Spain
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Granada, Andalusia 18071, Spain
| | - Yousif Rassam
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University of Münster, Münster, North Rhine-Westphalia 48149, Germany
| | - Stefanie Heilmann-Heimbach
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital, Bonn, North Rhine-Westphalia 53127, Germany
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, North Rhine-Westphalia 48149, Germany
| | - Sabine Kliesch
- Department of Clinical and Surgical Andrology, Centre of Reproductive Medicine and Andrology, University of Münster, Münster, North Rhine-Westphalia 48149, Germany
| | - Jörg Gromoll
- Correspondence: Jörg Gromoll, Dr. rer. nat., Institute of Reproductive and Regenerative Biology, Centre of Reproductive Medicine and Andrology, University of Münster, Albert-Schweitzer-Campus 1, Geb. D11, 48149 Münster, Germany.
| |
Collapse
|
15
|
Crespi B, Yang N. Three laws of teleonometrics. Biol J Linn Soc Lond 2022. [DOI: 10.1093/biolinnean/blac068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Abstract
We define teleonometrics as the theoretical and empirical study of teleonomy. We propose three laws for teleonometrics. The first law describes the hierarchical organization of teleonomic functions across biological levels from genes to individuals. According to this law, the number of goal-directed functions increases from individuals (one goal, maximizing inclusive fitness) to intermediate levels and to genes and alleles (myriad time-, space- and context-dependent goals, depending upon degrees and patterns of pleiotropy). The second law describes the operation of teleonomic functions under trade-offs, coadaptations and negative and positive pleiotropies, which are universal in biological systems. According to this law, the functions of an allele, gene or trait are described and defined by patterns of antagonistic (trading off) and compatible (coadapted) functions. The third law of teleonometrics is that the major transitions in evolution are driven by the origins of novel, emergent goals associated with functional changes and by the breaking and reshaping of trade-offs, especially by mechanisms involving increases in resources or time, and new divisions of labour or function. We illustrate the application of these laws using data from three empirical vignettes, which help to show the usefulness of teleonometric viewpoints for understanding the interfaces between function, trade-offs and dysfunctions manifest as disease.
Collapse
Affiliation(s)
- Bernard Crespi
- Department of Biological Sciences, Simon Fraser University , Burnaby, British Columbia, V5A 1S6 , Canada
| | - Nancy Yang
- Department of Biological Sciences, Simon Fraser University , Burnaby, British Columbia, V5A 1S6 , Canada
| |
Collapse
|
16
|
Nabhan AF, Mburu G, Elshafeey F, Magdi R, Kamel M, Elshebiny M, Abuelnaga YG, Ghonim M, Abdelhamid MH, Ghonim M, Eid P, Morsy A, Nasser M, Abdelwahab N, Elhayatmy F, Hussein AA, Elgabaly N, Sawires E, Tarkhan Y, Doas Y, Farrag N, Amir A, Gobran MF, Maged M, Abdulhady M, Sherif Y, Dyab M, Kiarie J. Women's reproductive span: a systematic scoping review. Hum Reprod Open 2022; 2022:hoac005. [PMID: 35280216 PMCID: PMC8907405 DOI: 10.1093/hropen/hoac005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/31/2022] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION What is the scope of literature regarding women's reproductive span in terms of definitions, trends and determinants? SUMMARY ANSWER The scoping review found a wide variation in definitions, trends and determinants of biological, social and effective women's reproductive span. WHAT IS KNOWN ALREADY A woman's reproductive span refers to her childbearing years. Its span influences a woman's reproductive decisions. STUDY DESIGN SIZE DURATION A systematic scoping review was conducted. We searched MEDLINE, PubMed, JSTOR, CINAHL, Web of Science and Scopus electronic databases from inception to January 2021 without imposing language or date restrictions. We searched unpublished sources including the Global Burden of Disease, Demographic and Health Surveys, and National Health and Nutrition Examination Surveys. The list of relevant references was searched by hand. Sixty-seven reports on women's reproductive span were included in this review. PARTICIPANTS/MATERIALS SETTING METHODS This scoping systematic review followed an established framework. The reporting of this scoping review followed the reporting requirements provided in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses, Extension for Scoping Reviews. Identified records were independently screened and data were extracted. We performed conceptual synthesis by grouping the studies by available concepts of reproductive span and then summarized definitions, measures used, temporal trends, determinants, and broad findings of implications on population demographics and assisted reproduction. Structured tabulation and graphical synthesis were used to show patterns in the data and convey detailed information efficiently, along with a narrative commentary. MAIN RESULTS AND THE ROLE OF CHANCE A total of 67 relevant reports on women's reproductive span were published between 1980 and 2020 from 74 countries. Most reports (42/67) were cross-sectional in design. Literature on reproductive span was conceptually grouped as biological (the interval between age at menarche and age at menopause), effective (when a woman is both fertile and engaging in sexual activity) and social (period of exposure to sexual activity). We summarized the working definitions, trends and determinants of each concept. Few articles addressed implications on demographics and assisted reproduction. LIMITATIONS REASONS FOR CAUTION A formal assessment of methodological quality of the included studies was not performed because the aim of this review was to provide an overview of the existing evidence base regardless of quality. WIDER IMPLICATIONS OF THE FINDINGS The review produced a comprehensive set of possible definitions of women's reproductive span, trends, and potential determinants. Further advancement of these findings will involve collaboration with relevant stakeholders to rate the importance of each definition in relation to demography and fertility care, outline a set of core definitions, identify implications for policy, practice or research and define future research opportunities to explore linkages between reproductive spans, their determinants, and the need for assisted reproduction. STUDY FUNDING/COMPETING INTERESTS This work received funding from the UNDP-UNFPA-UNICEF-WHO-World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP), a cosponsored programme executed by the World Health Organization (WHO). The authors had no competing interests. STUDY REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- A F Nabhan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - G Mburu
- The UNDP/UNFPA/UNICEF/WHO/World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP Research), World Health Organization, Geneva, Switzerland
| | - F Elshafeey
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - R Magdi
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - M Kamel
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - M Elshebiny
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - Y G Abuelnaga
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - M Ghonim
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - M H Abdelhamid
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - Mo Ghonim
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - P Eid
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - A Morsy
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - M Nasser
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - N Abdelwahab
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - F Elhayatmy
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - A A Hussein
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - N Elgabaly
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - E Sawires
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - Y Tarkhan
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - Y Doas
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - N Farrag
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - A Amir
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - M F Gobran
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - M Maged
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - M Abdulhady
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - Y Sherif
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - M Dyab
- Egyptian Center for Evidence Based Medicine, Cairo, Egypt
| | - J Kiarie
- The UNDP/UNFPA/UNICEF/WHO/World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP Research), World Health Organization, Geneva, Switzerland
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW The aim of this review is to evaluate biological, life history, environmental, and lifestyle factors and exposures that cause variability in menstrual cycle length (MCL). RECENT FINDINGS Recent literature has detailed a number of factors that influence MCL, with particular emphasis placed on novel environmental exposures, such as air pollution and endocrine disrupting chemicals. SUMMARY MCL varies widely in response to intrinsic and extrinsic inputs and is a useful predictor of reproductive health and fecundability. VIDEO ABSTRACT http://links.lww.com/COE/A28.
Collapse
Affiliation(s)
| | - Ariel L. Scalise
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | - Shruthi Mahalingaiah
- Department of Environmental Health, Harvard T.H. Chan School of Public Health
- Massachusetts General Hospital Fertility Center, Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology, and Reproductive Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Younis JS, Iskander R, Fauser BCJM, Izhaki I. Does an association exist between menstrual cycle length within the normal range and ovarian reserve biomarkers during the reproductive years? A systematic review and meta-analysis. Hum Reprod Update 2021; 26:904-928. [PMID: 32514566 DOI: 10.1093/humupd/dmaa013] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/05/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Regular menstrual cycling during the reproductive years is an indicator of spontaneous ovulation but sometimes falsely perceived as an indicator of preserved fertility. In contrast, menstrual cycle shortening, a physiologic occurrence preceding the menopausal transition, is not usually perceived as an indicator of decreased ovarian reserve in the general population. OBJECTIVE AND RATIONALE The individual decrease in menstrual cycle length (MCL) might represent a sensitive biomarker of diminishing ovarian reserve. The aim of this systematic review and meta-analysis is to examine the possible association between MCL in regularly cycling women (21-35 days) and ovarian reserve tests (ORT), fecundability in natural cycles and IVF outcomes. SEARCH METHODS An electronic database search employing PubMed, Web of Science, Trip, EBSCO, ClinicalTrials.gov and the Cochrane library was performed to identify research articles, only on human, published between January 1978 and August 2019. Search terms were pregnancy OR fertility OR fecundity OR fecundability, anti-Müllerian hormone OR AMH OR antral follicle count OR AFC OR ovarian reserve OR ovarian reserve test, in vitro fertilization OR ART OR assisted reproductive therapy OR assisted reproductive treatment OR assisted reproductive technology OR IVF OR ICSI, menstrual cycle length OR menstrual cycle characteristics. We combined these terms to complete the search. All prospective and retrospective studies exploring an association between MCL and proxies of ovarian reserve were included. The exclusions included studies of PCOS, ovarian failure, oral contraception treatment, prior chemotherapy and/or radiotherapy or ovarian surgery. The Newcastle-Ottawa scale was used to assess the quality of studies that were eligible for meta-analysis. OUTCOMES Eleven studies were eligible for meta-analysis, including 12 031 women. The included studies had a low risk of bias. Short MCL (21-27 days) was associated with lower ORT values as compared to normal (28-31 days), long (32-35 days) and all other (28-35 days) MCL sets. The estimated weighted mean difference (WMD) of AMH level was -1.3 ng/mL (95% CI: -1.75 to -0.86, P < 0.001) between the short and normal MCL sets. The estimated WMD of AFC values was -5.17 (95% CI: -5.96 to -4.37, P < 0.001) between the short and normal MCL sets. The weighted overall odds ratio (OR) of fecundability in natural cycles between women with short versus normal MCL sets was statistically significant (overall OR 0.81; 95% CI 0.72-0.91, P < 0.001). In the IVF setting, fewer oocytes were retrieved in short MCL in comparison to normal, long and all other MCL sets, with an estimated WMD of -1.8 oocytes (95% CI: -2.5 to -1.1, P < 0.001) in the short versus normal MCL sets. The weighted overall OR of clinical pregnancy rate between women with short versus all other MCL sets was statistically significant (overall OR 0.76; 95% CI: 0.60 to 0.96, P = 0.02). Low levels of heterogeneity were found in most meta-analyses of MCL and qualitative ovarian reserve biomarkers, while heterogeneity was high in meta-analyses performed for quantitative measures. WIDER IMPLICATIONS MCL in regularly cycling women is closely related to ovarian reserve biomarkers during the reproductive years. A short MCL, as compared to normal, is significantly associated with lower ORT values, reduced fecundability and inferior IVF outcomes, independent of age. The results imply that short MCL may be a sign of ovarian aging, combining the quantitative and qualitative facets of ovarian reserve. Educational efforts ought to be designed to guide women with short MCL at a young age, who desire children in the future, to seek professional counselling.
Collapse
Affiliation(s)
- Johnny S Younis
- Reproductive Medicine, Department of Obstetrics and Gynecology, Baruch-Padeh Medical Center, Poriya 15208, Israel.,Azrieili Faculty of Medicine, Galilee, Bar-Ilan University, Safed, Israel
| | - Rula Iskander
- Reproductive Medicine, Department of Obstetrics and Gynecology, Baruch-Padeh Medical Center, Poriya 15208, Israel
| | - Bart C J M Fauser
- Department of Reproductive Medicine and Gynecology, University of Utrecht and University Medical Center Utrecht, 3508 TC, Utrecht, The Netherlands
| | - Ido Izhaki
- Department of Evolutionary and Environmental Biology, University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
19
|
Bosch E, Alviggi C, Lispi M, Conforti A, Hanyaloglu AC, Chuderland D, Simoni M, Raine-Fenning N, Crépieux P, Kol S, Rochira V, D'Hooghe T, Humaidan P. Reduced FSH and LH action: implications for medically assisted reproduction. Hum Reprod 2021; 36:1469-1480. [PMID: 33792685 PMCID: PMC8129594 DOI: 10.1093/humrep/deab065] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
Luteinizing hormone (LH) and follicle-stimulating hormone (FSH) play complementary roles in follicle development and ovulation via a complex interaction in the hypothalamus, anterior pituitary gland, reproductive organs, and oocytes. Impairment of the production or action of gonadotropins causes relative or absolute LH and FSH deficiency that compromises gametogenesis and gonadal steroid production, thereby reducing fertility. In women, LH and FSH deficiency is a spectrum of conditions with different functional or organic causes that are characterized by low or normal gonadotropin levels and low oestradiol levels. While the causes and effects of reduced LH and FSH production are very well known, the notion of reduced action has received less attention by researchers. Recent evidence shows that molecular characteristics, signalling as well as ageing, and some polymorphisms negatively affect gonadotropin action. These findings have important clinical implications, in particular for medically assisted reproduction in which diminished action determined by the afore-mentioned factors, combined with reduced endogenous gonadotropin production caused by GnRH analogue protocols, may lead to resistance to gonadotropins and, thus, to an unexpected hypo-response to ovarian stimulation. Indeed, the importance of LH and FSH action has been highlighted by the International Committee for Monitoring Assisted Reproduction Technologies (ICMART) in their definition of hypogonadotropic hypogonadism as gonadal failure associated with reduced gametogenesis and gonadal steroid production due to reduced gonadotropin production or action. The aim of this review is to provide an overview of determinants of reduced FSH and LH action that are associated with a reduced response to ovarian stimulation.
Collapse
Affiliation(s)
| | - C Alviggi
- Department of Neuroscience, Reproductive Science and Odontostomatology, University Federico II, Naples, Italy
| | - M Lispi
- Global Medical Affairs Fertility, Merck KGaA, Darmstadt, Germany.,International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Modena, Italy
| | - A Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University Federico II, Naples, Italy
| | - A C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - D Chuderland
- Global Medical Affairs Fertility, Merck KGaA, Darmstadt, Germany
| | - M Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - N Raine-Fenning
- Department of Child Health, Obstetrics and Gynaecology, School of Medicine, University of Nottingham, Nottingham, UK
| | - P Crépieux
- Physiologie de la Reproduction et des Comportements, UMR INRA 085, CNRS 7247, Université de Tours, Nouzilly, France
| | - S Kol
- IVF Unit, Elisha Hospital, Haifa, Israel
| | - V Rochira
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.,Unit of Endocrinology, Azienda Ospedaliero-Universitaria of Modena, Ospedale Civile di Baggiovara, Modena, Italy
| | - T D'Hooghe
- Global Medical Affairs Fertility, Merck KGaA, Darmstadt, Germany.,Department of Development & Regeneration, University of Leuven (KU Leuven), Leuven, Belgium.,Department of Obstetrics and Gynecology, Yale University, New Haven, CT, USA
| | - P Humaidan
- Fertility Clinic, Skive Regional Hospital, and the Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
20
|
Zhao W, Smith JA, Bielak LF, Ruiz-Narvaez EA, Yu M, Hood MM, Peyser PA, Kardia SL, Harlow SD. Associations between polygenic risk score for age at menarche and menopause, reproductive timing, and serum hormone levels in multiple race/ethnic groups. Menopause 2021; 28:819-828. [PMID: 33878091 PMCID: PMC8225555 DOI: 10.1097/gme.0000000000001775] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We assessed associations of genetic loci that contribute to age at menarche and menopause with sentinel menopausal traits in multiple race/ethnic groups. METHODS Genetic data from the Study of Women's Health Across the Nation include 738 White, 366 Black, 139 Chinese, and 145 Japanese women aged 42 to 52 at baseline. We constructed standardized polygenic risk scores (PRSs) using single nucleotide polymorphisms identified from large-scale genome-wide association studies meta-analyses of ages at menopause and menarche, evaluating associations with each trait within each race/ethnic group. RESULTS Menopause PRS was significantly associated with age at menopause in White women after Bonferroni correction (P < 0.004) and nominally associated in Chinese and Japanese women (P < 0.05) (7.4-8.5 mo delay for one standard deviation [SD] increase in PRS). Menarche PRS was significantly associated with age at menarche in White (P < 0.004) and nominally associated in Black and Japanese women (P < 0.05) (2.6-4.8 mo delay for one SD increase). In White women, menarche and menopause PRSs were significantly associated (P < 0.004) with shorter and longer (5.9 and 9.6 mo for one SD increase) reproductive lifespans, respectively, and menopause PRS with a longer menopausal transition (7.1 mo for one SD increase). We observed a significant positive association (P < 0.004) between menopause PRS and E2 level 2 years before menopause and a nominal association (P < 0.05) 2 years after menopause in Japanese women. CONCLUSIONS In addition to menopausal timing, PRSs associated with onset and ending of reproductive life were associated with reproductive lifespan, length of the menopausal transition, and E2 levels in different race/ethnic groups.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109
| | - Lawrence F. Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109
| | - Edward A. Ruiz-Narvaez
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, 48109
| | - Miao Yu
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109
| | - Michelle M. Hood
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109
| | - Patricia A. Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109
| | - Sharon L.R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109
| | - Sioban D. Harlow
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109
| |
Collapse
|
21
|
Abstract
Gonadotropins are glycoprotein sex hormones regulating development and reproduction and bind to specific G protein–coupled receptors expressed in the gonads. Their effects on multiple signaling cascades and intracellular events have recently been characterized using novel technological and scientific tools. The impact of allosteric modulators on gonadotropin signaling, the role of sugars linked to the hormone backbone, the detection of endosomal compartments supporting signaling modules, and the dissection of different effects mediated by these molecules are areas that have advanced significantly in the last decade. The classic view providing the exclusive activation of the cAMP/protein kinase A (PKA) and the steroidogenic pathway by these hormones has been expanded with the addition of novel signaling cascades as determined by high-resolution imaging techniques. These new findings provided new potential therapeutic applications. Despite these improvements, unanswered issues of gonadotropin physiology, such as the intrinsic pro-apoptotic potential to these hormones, the existence of receptors assembled as heteromers, and their expression in extragonadal tissues, remain to be studied. Elucidating these issues is a challenge for future research.
Collapse
Affiliation(s)
- Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria di Modena, Via P. Giardini 1355, 41126 Modena, Italy
| |
Collapse
|
22
|
Dinsdale N, Nepomnaschy P, Crespi B. The evolutionary biology of endometriosis. EVOLUTION MEDICINE AND PUBLIC HEALTH 2021; 9:174-191. [PMID: 33854783 PMCID: PMC8030264 DOI: 10.1093/emph/eoab008] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/04/2021] [Indexed: 12/14/2022]
Abstract
We provide the first analysis and synthesis of the evolutionary and mechanistic bases for risk of endometriosis in humans, structured around Niko Tinbergen's four questions about phenotypes: phylogenetic history, development, mechanism and adaptive significance. Endometriosis, which is characterized by the proliferation of endometrial tissue outside of the uterus, has its phylogenetic roots in the evolution of three causally linked traits: (1) highly invasive placentation, (2) spontaneous rather than implantation-driven endometrial decidualization and (3) frequent extensive estrogen-driven endometrial proliferation and inflammation, followed by heavy menstrual bleeding. Endometriosis is potentiated by these traits and appears to be driven, proximately, by relatively low levels of prenatal and postnatal testosterone. Testosterone affects the developing hypothalamic-pituitary-ovarian (HPO) axis, and at low levels, it can result in an altered trajectory of reproductive and physiological phenotypes that in extreme cases can mediate the symptoms of endometriosis. Polycystic ovary syndrome, by contrast, is known from previous work to be caused primarily by high prenatal and postnatal testosterone, and it demonstrates a set of phenotypes opposite to those found in endometriosis. The hypothesis that endometriosis risk is driven by low prenatal testosterone, and involves extreme expression of some reproductive phenotypes, is supported by a suite of evidence from genetics, development, endocrinology, morphology and life history. The hypothesis also provides insights into why these two diametric, fitness-reducing disorders are maintained at such high frequencies in human populations. Finally, the hypotheses described and evaluated here lead to numerous testable predictions and have direct implications for the treatment and study of endometriosis. Lay summary: Endometriosis is caused by endometrial tissue outside of the uterus. We explain why and how humans are vulnerable to this disease, and new perspectives on understanding and treating it. Endometriosis shows evidence of being caused in part by relatively low testosterone during fetal development, that 'programs' female reproductive development. By contrast, polycystic ovary syndrome is associated with relatively high testosterone in prenatal development. These two disorders can thus be seen as 'opposite' to one another in their major causes and correlates. Important new insights regarding diagnosis, study and treatment of endometriosis follow from these considerations.
Collapse
Affiliation(s)
- Natalie Dinsdale
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Pablo Nepomnaschy
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Bernard Crespi
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
23
|
Sinnott-Armstrong N, Naqvi S, Rivas M, Pritchard JK. GWAS of three molecular traits highlights core genes and pathways alongside a highly polygenic background. eLife 2021; 10:e58615. [PMID: 33587031 PMCID: PMC7884075 DOI: 10.7554/elife.58615] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 01/18/2021] [Indexed: 12/30/2022] Open
Abstract
Genome-wide association studies (GWAS) have been used to study the genetic basis of a wide variety of complex diseases and other traits. We describe UK Biobank GWAS results for three molecular traits-urate, IGF-1, and testosterone-with better-understood biology than most other complex traits. We find that many of the most significant hits are readily interpretable. We observe huge enrichment of associations near genes involved in the relevant biosynthesis, transport, or signaling pathways. We show how GWAS data illuminate the biology of each trait, including differences in testosterone regulation between females and males. At the same time, even these molecular traits are highly polygenic, with many thousands of variants spread across the genome contributing to trait variance. In summary, for these three molecular traits we identify strong enrichment of signal in putative core gene sets, even while most of the SNP-based heritability is driven by a massively polygenic background.
Collapse
Affiliation(s)
| | - Sahin Naqvi
- Department of Genetics, Stanford UniversityStanfordUnited States
- Department of Chemical and Systems Biology, Stanford UniversityStanfordUnited States
| | - Manuel Rivas
- Department of Biomedical Data Sciences, Stanford UniversityStanfordUnited States
| | - Jonathan K Pritchard
- Department of Genetics, Stanford UniversityStanfordUnited States
- Department of Biology, Stanford UniversityStanfordUnited States
| |
Collapse
|
24
|
Feng W, Zhang Y, Pan Y, Zhang Y, Liu M, Huang Y, Xiao Y, Mo W, Jiao J, Wang X, Tian D, Yang L, Ma Y. Association of three missense mutations in the homocysteine-related MTHFR and MTRR gene with risk of polycystic ovary syndrome in Southern Chinese women. Reprod Biol Endocrinol 2021; 19:5. [PMID: 33407572 PMCID: PMC7789417 DOI: 10.1186/s12958-020-00688-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The etiology between homocysteine and polycystic ovary syndrome (PCOS) is unclear. In humans, the level of homocysteine is mainly affected by two enzymes: methylene tetrahydrofolate reductase (MTHFR) and methionine synthase reductase (MTRR). While the activity of these two enzymes is mainly affected by three missense mutations, namely C677T (MTHFR), A1298C (MTHFR), and A66G (MTRR). This study aims to examine the association between the three missense mutations and PCOS and investigate whether the three missense mutations exerted their effect on PCOS by affecting the homocysteine level. METHODS A case-control study was designed, comprising 150 people with PCOS and 300 controls. Logistic regression analysis was used to assess the association between the three missense mutations and PCOS. Linear regression analysis was used to assess the association between the three missense mutations and the homocysteine level. Mediation analysis was used to investigate whether the three missense mutations exerted their effect on PCOS by affecting the homocysteine level. RESULTS Following adjustments and multiple rounds of testing, MTHFR A1298C was found to be significantly associated with PCOS in a dose-dependent manner (compared to AA, OR = 2.142 for AC & OR = 3.755 for CC; P < 0.001). MTRR A66G was nominally associated with PCOS. Mutations in MTHFR A1298C and MTRR A66G were significantly associated with the homocysteine level. Mediation analysis suggested the effect of MTHFR A1298C on PCOS was mediated by homocysteine. CONCLUSIONS MTHFR A1298C and MTRR A66G were associated with PCOS, and MTHFR A1298C might affect the risk of PCOS by influencing the homocysteine level.
Collapse
Affiliation(s)
- Wanqin Feng
- Department of Gynecology, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, No.253, Gongye Middle Avenue, Haizhu District, 510280, Guangzhou, Guangdong, China
| | - Yan Zhang
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Yuan Pan
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Yi Zhang
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Minjuan Liu
- Department of Gynecology, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, No.253, Gongye Middle Avenue, Haizhu District, 510280, Guangzhou, Guangdong, China
- Department of Obstetrics and Gynecology, Dongguan People's Hospital, 523000, Dongguan, China
| | - Yuxin Huang
- Department of Obstetrics, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Yuanling Xiao
- Department of Obstetrics, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Wenyu Mo
- Department of Gynecology, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, No.253, Gongye Middle Avenue, Haizhu District, 510280, Guangzhou, Guangdong, China
| | - Junjie Jiao
- Department of Fetal Medicine and Prenatal Diagnosis, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Xiaoyang Wang
- Department of Gynecology, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, No.253, Gongye Middle Avenue, Haizhu District, 510280, Guangzhou, Guangdong, China
| | - Dan Tian
- Department of Obstetrics, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Lixia Yang
- Department of Emergency, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
| | - Ying Ma
- Department of Gynecology, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, No.253, Gongye Middle Avenue, Haizhu District, 510280, Guangzhou, Guangdong, China.
| |
Collapse
|
25
|
A Systematic Two-Sample Mendelian Randomization Analysis Identifies Shared Genetic Origin of Endometriosis and Associated Phenotypes. Life (Basel) 2021; 11:life11010024. [PMID: 33401535 PMCID: PMC7824623 DOI: 10.3390/life11010024] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 12/18/2022] Open
Abstract
Endometriosis, one of the most common gynecological disorders, is a complex disease characterized by the growth of endometrial-like tissue in extra-uterine locations and is a cause of pelvic pain and infertility. Evidence from observational studies indicate that endometriosis usually appears together with several other phenotypes. These include a list of autoimmune diseases, most of them more prevalent in women, anthropometric traits associated with leanness in the adulthood, as well as female reproductive traits, including altered hormone levels and those associated with a prolonged exposure to menstruation. However, the biological mechanisms underlying their co-morbidity remains unknown. To explore whether those phenotypes and endometriosis share a common genetic origin, we performed a systematic Two-Sample Mendelian Randomization (2SMR) analysis using public GWAS data. Our results suggest potential common genetic roots between endometriosis and female anthropometric and reproductive traits. Particularly, our data suggests that reduced weight and BMI might be mediating the genetic susceptibility to suffer endometriosis. Furthermore, data on female reproductive traits strongly suggest that genetic variants that predispose to a more frequent exposure to menstruation, through earlier age at menarche and shorter menstrual cycles, might also increase the risk to suffer from endometriosis.
Collapse
|
26
|
Bianco B, Loureiro FA, Trevisan CM, Peluso C, Christofolini DM, Montagna E, Laganà AS, Barbosa CP. Effects of FSHR and FSHB Variants on Hormonal Profile and Reproductive Outcomes of Infertile Women With Endometriosis. Front Endocrinol (Lausanne) 2021; 12:760616. [PMID: 34659133 PMCID: PMC8514996 DOI: 10.3389/fendo.2021.760616] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Single nucleotide variants (SNVs) FSHB:c.-211G>T, FSHR:c.919G>A, and FSHR:c.2039G>A were reported to be associated with the variability in FSH and LH levels, and in vitro fertilization (IVF) outcomes. In this study, we aimed to evaluate the effects of FSHB:c.-211G>T, FSHR:c.919G>A, and FSHR:c.2039G>A variants, alone and combined, on the hormonal profile and reproduction outcomes of women with endometriosis. METHODS A cross-sectional study was performed comprising 213 infertile Brazilian women with endometriosis who underwent IVF treatment. Genotyping was performed using TaqMan real-time PCR. Variables were compared according to the genotypes of each variant and genetic models, and the combined effects of the SNVs were evaluated using the multifactorial dimensionality reduction method. RESULTS FSHB:c.-211G>T affected LH levels in women with overall endometriosis and minimal/mild disease. FSHR:c.919G>A affected FSH levels in women with overall endometriosis and the number of oocytes retrieved in those with moderate/severe endometriosis. Moreover, the FSHR:c.2039G>A affected FSH levels in women with overall endometriosis, LH levels and total amount of rFSH in those with minimal/mild disease, and number of follicles and number of oocytes retrieved in those with moderate/severe endometriosis. No effect on hormone profile or reproductive outcomes was observed when the genotypes were combined. CONCLUSIONS Variants of the FSHB and FSHR genes separately interfered with the hormonal profiles and IVF outcomes of women with endometriosis.
Collapse
Affiliation(s)
- Bianca Bianco
- Discipline of Sexual and Reproductive Health, and Populational Genetics – Department of Collective Health, Faculdade de Medicina do ABC/Centro Universitário FMABC, Santo André, Brazil
- Department of Urology, Instituto Israelita de Ensino e Pesquisa Albert Einstein, São Paulo, Brazil
- *Correspondence: Bianca Bianco,
| | - Flavia Altheman Loureiro
- Discipline of Sexual and Reproductive Health, and Populational Genetics – Department of Collective Health, Faculdade de Medicina do ABC/Centro Universitário FMABC, Santo André, Brazil
| | - Camila Martins Trevisan
- Discipline of Sexual and Reproductive Health, and Populational Genetics – Department of Collective Health, Faculdade de Medicina do ABC/Centro Universitário FMABC, Santo André, Brazil
| | - Carla Peluso
- Discipline of Sexual and Reproductive Health, and Populational Genetics – Department of Collective Health, Faculdade de Medicina do ABC/Centro Universitário FMABC, Santo André, Brazil
| | - Denise Maria Christofolini
- Discipline of Sexual and Reproductive Health, and Populational Genetics – Department of Collective Health, Faculdade de Medicina do ABC/Centro Universitário FMABC, Santo André, Brazil
| | - Erik Montagna
- Postgraduate Program in Health Sciences, Faculdade de Medicina do ABC/Centro Universitário FMABC, Santo André, Brazil
| | - Antonio Simone Laganà
- Department of Obstetrics and Gynecology, “Filippo Del Ponte” Hospital, University of Insubria, Varese, Italy
| | - Caio Parente Barbosa
- Discipline of Sexual and Reproductive Health, and Populational Genetics – Department of Collective Health, Faculdade de Medicina do ABC/Centro Universitário FMABC, Santo André, Brazil
| |
Collapse
|
27
|
Adewuyi EO, Mehta D, Sapkota Y, Auta A, Yoshihara K, Nyegaard M, Griffiths LR, Montgomery GW, Chasman DI, Nyholt DR. Genetic analysis of endometriosis and depression identifies shared loci and implicates causal links with gastric mucosa abnormality. Hum Genet 2020; 140:529-552. [PMID: 32959083 DOI: 10.1007/s00439-020-02223-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
Abstract
Evidence from observational studies indicates that endometriosis and depression often co-occur. However, conflicting evidence exists, and the etiology as well as biological mechanisms underlying their comorbidity remain unknown. Utilizing genome-wide association study (GWAS) data, we comprehensively assessed the relationship between endometriosis and depression. Single nucleotide polymorphism effect concordance analysis (SECA) found a significant genetic overlap between endometriosis and depression (PFsig-permuted = 9.99 × 10-4). Linkage disequilibrium score regression (LDSC) analysis estimated a positive and highly significant genetic correlation between the two traits (rG = 0.27, P = 8.85 × 10-27). A meta-analysis of endometriosis and depression GWAS (sample size = 709,111), identified 20 independent genome-wide significant loci (P < 5 × 10-8), of which eight are novel. Mendelian randomization analysis (MR) suggests a causal effect of depression on endometriosis. Combining gene-based association results across endometriosis and depression GWAS, we identified 22 genes with a genome-wide significant Fisher's combined P value (FCPgene < 2.75 × 10-6). Genes with a nominal gene-based association (Pgene < 0.05) were significantly enriched across endometriosis and depression (Pbinomial-test = 2.90 × 10-4). Also, genes overlapping the two traits at Pgene < 0.1 (Pbinomial-test = 1.31 × 10-5) were significantly enriched for the biological pathways 'cell-cell adhesion', 'inositol phosphate metabolism', 'Hippo-Merlin signaling dysregulation' and 'gastric mucosa abnormality'. These results reveal a shared genetic etiology for endometriosis and depression. Indeed, additional analyses found evidence of a causal association between each of endometriosis and depression and at least one abnormal condition of gastric mucosa. Our study confirms the comorbidity of endometriosis and depression, implicates links with gastric mucosa abnormalities in their causal pathways and reveals potential therapeutic targets for further investigation.
Collapse
Affiliation(s)
- Emmanuel O Adewuyi
- School of Biomedical Sciences, Faculty of Health, and Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD, Australia.
| | - Divya Mehta
- School of Biomedical Sciences, Faculty of Health, and Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Yadav Sapkota
- Department of Epidemiology And Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | | | | | - Asa Auta
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, UK
| | - Kosuke Yoshihara
- Department of Obstetrics And Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 950-2181, Japan
| | - Mette Nyegaard
- Department of Biomedicine - Human Genetics, Aarhus University, 8000, Aarhus,, Denmark.,iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, 2100, Copenhagen, Denmark
| | - Lyn R Griffiths
- School of Biomedical Sciences, Faculty of Health, and Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Grant W Montgomery
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Daniel I Chasman
- Divisions of Preventive Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Dale R Nyholt
- School of Biomedical Sciences, Faculty of Health, and Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD, Australia.
| |
Collapse
|
28
|
Laisk T, Tšuiko O, Jatsenko T, Hõrak P, Otala M, Lahdenperä M, Lummaa V, Tuuri T, Salumets A, Tapanainen JS. Demographic and evolutionary trends in ovarian function and aging. Hum Reprod Update 2020; 25:34-50. [PMID: 30346539 DOI: 10.1093/humupd/dmy031] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/03/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The human female reproductive lifespan is regulated by the dynamics of ovarian function, which in turn is influenced by several factors: from the basic molecular biological mechanisms governing folliculogenesis, to environmental and lifestyle factors affecting the ovarian reserve between conception and menopause. From a broader point of view, global and regional demographic trends play an additional important role in shaping the female reproductive lifespan, and finally, influences on an evolutionary scale have led to the reproductive senescence that precedes somatic senescence in humans. OBJECTIVE AND RATIONALE The narrative review covers reproductive medicine, by integrating the molecular mechanisms of ovarian function and aging with short-term demographic and long-term evolutionary trends. SEARCH METHODS PubMed and Google Scholar searches were performed with relevant keywords (menopause, folliculogenesis, reproductive aging, reproductive lifespan and life history theory). The reviewed articles and their references were restricted to those written in English. OUTCOMES We discuss and summarize the rapidly accumulating information from large-scale population-based and single-reproductive-cell genomic studies, their constraints and advantages in the context of female reproductive aging as well as their possible evolutionary significance on the life history trajectory from foetal-stage folliculogenesis until cessation of ovarian function in menopause. The relevant environmental and lifestyle factors and demographic trends are also discussed in the framework of predominant evolutionary hypotheses explaining the origin and maintenance of menopause. WIDER IMPLICATIONS The high speed at which new data are generated has so far raised more questions than it has provided solid answers and has been paralleled by a lack of satisfactory interpretations of the findings in the context of human life history theory. Therefore, the recent flood of data could offer an unprecedented tool for future research to possibly confirm or rewrite human evolutionary reproductive history, at the same time providing novel grounds for patient counselling and family planning strategies.
Collapse
Affiliation(s)
- Triin Laisk
- Competence Centre on Health Technologies, Tiigi 61b, Tartu, Estonia.,Institute of Clinical Medicine, Department of Obstetrics and Gynaecology, University of Tartu, L. Puusepa 8, Tartu, Estonia
| | - Olga Tšuiko
- Competence Centre on Health Technologies, Tiigi 61b, Tartu, Estonia.,Institute of Biomedicine and Translational Medicine, Department of Biomedicine, University of Tartu, Ravila 19, Tartu, Estonia
| | - Tatjana Jatsenko
- Competence Centre on Health Technologies, Tiigi 61b, Tartu, Estonia
| | - Peeter Hõrak
- Department of Zoology, University of Tartu, Vanemuise 46, Tartu, Estonia
| | - Marjut Otala
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 2, Helsinki, Finland
| | - Mirkka Lahdenperä
- Department of Biology, University of Turku, Turun yliopisto, Turku, Finland
| | - Virpi Lummaa
- Department of Biology, University of Turku, Turun yliopisto, Turku, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 2, Helsinki, Finland
| | - Andres Salumets
- Competence Centre on Health Technologies, Tiigi 61b, Tartu, Estonia.,Institute of Clinical Medicine, Department of Obstetrics and Gynaecology, University of Tartu, L. Puusepa 8, Tartu, Estonia.,Institute of Biomedicine and Translational Medicine, Department of Biomedicine, University of Tartu, Ravila 19, Tartu, Estonia.,Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 2, Helsinki, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 2, Helsinki, Finland.,Department of Obstetrics and Gynecology, University Hospital of Oulu, University of Oulu, Medical Research Center Oulu and PEDEGO Research Unit, OYS Oulu, Finland
| |
Collapse
|
29
|
Genetic Characterization of Endometriosis Patients: Review of the Literature and a Prospective Cohort Study on a Mediterranean Population. Int J Mol Sci 2020; 21:ijms21051765. [PMID: 32143537 PMCID: PMC7084255 DOI: 10.3390/ijms21051765] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/20/2020] [Accepted: 03/02/2020] [Indexed: 12/21/2022] Open
Abstract
The pathogenesis of endometriosis is unknown, but some evidence supports a genetic predisposition. The purpose of this study was to evaluate the recent literature on the genetic characterization of women affected by endometriosis and to evaluate the influence of polymorphisms of the wingless-type mammalian mouse tumour virus integration site family member 4 (WNT4), vezatin (VEZT), and follicle stimulating hormone beta polypeptide (FSHB) genes, already known to be involved in molecular mechanisms associated with the proliferation and development of endometriotic lesions in the Sardinian population. Materials and Methods: In order to provide a comprehensive and systematic tool for those approaching the genetics of endometriosis, the most cited review, observational, cohort and case-control studies that have evaluated the genetics of endometriosis in the last 20 years were collected. Moreover, 72 women were recruited for a molecular biology analysis of whole-blood samples—41 patients affected by symptomatic endometriosis and 31 controls. The molecular typing of three single nucleotide polymorphisms (SNPs) was evaluated in patients and controls: rs7521902, rs10859871 and rs11031006, mapped respectively in the WNT4, VEZT and FSHB genes. In this work, the frequency of alleles, genotypes and haplotypes of these SNPs in Sardinian women is described. Results: From the initial search, a total of 73 articles were chosen. An analysis of the literature showed that in endometriosis pathogenesis, the contribution of genetics has been well supported by many studies. The frequency of genotypes observed in the groups of the study population of 72 women was globally coherent with the law of the Hardy–Weinberg equilibrium. For the SNP rs11031006 (FSHB), the endometriosis group did not show an increase in genotypic or allelic frequency due to this polymorphism compared to the control group (p = 0.9999, odds ratio (OR) = 0.000, 95% confidence interval (CI), 0.000–15.000 and p = 0.731, OR = 1639, 95% CI, 0.39–683, respectively, for the heterozygous genotype and the polymorphic minor allele). For the SNP rs10859871 (VEZT), we found a significant difference in the frequency of the homozygous genotype in the control group compared to the affected women (p = 0.0111, OR = 0.0602, 95% CI, 0.005–0.501). For the SNP rs7521902 (WNT4), no increase in genotypic or allelic frequency between the two groups was shown (p = 0.3088, OR = 0.4133, 95% CI, 0.10–1.8 and p = 0.3297, OR = 2257, 95% CI, 0.55–914, respectively, for the heterozygous genotype and the polymorphic minor allele). Conclusion: An analysis of recent publications on the genetics of endometriosis showed a discrepancy in the results obtained in different populations. In the Sardinian population, the results obtained do not show a significant association between the investigated variants of the genes and a greater risk of developing endometriosis, although several other studies in the literature have shown the opposite. Anyway, the data underline the importance of evaluating genetic variants in different populations. In fact, in different ethnic groups, it is possible that specific risk alleles could act differently in the pathogenesis of the disease.
Collapse
|
30
|
Adewuyi EO, Sapkota Y, International Endogene Consortium (IEC), 23andMe Research Team, International Headache Genetics Consortium (IHGC), Auta A, Yoshihara K, Nyegaard M, Griffiths LR, Montgomery GW, Chasman DI, Nyholt DR. Shared Molecular Genetic Mechanisms Underlie Endometriosis and Migraine Comorbidity. Genes (Basel) 2020; 11:E268. [PMID: 32121467 PMCID: PMC7140889 DOI: 10.3390/genes11030268] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 01/02/2023] Open
Abstract
Observational epidemiological studies indicate that endometriosis and migraine co-occur within individuals more than expected by chance. However, the aetiology and biological mechanisms underlying their comorbidity remain unknown. Here we examined the relationship between endometriosis and migraine using genome-wide association study (GWAS) data. Single nucleotide polymorphism (SNP) effect concordance analysis found a significant concordance of SNP risk effects across endometriosis and migraine GWAS. Linkage disequilibrium score regression analysis found a positive and highly significant genetic correlation (rG = 0.38, P = 2.30 × 10-25) between endometriosis and migraine. A meta-analysis of endometriosis and migraine GWAS data did not reveal novel genome-wide significant SNPs, and Mendelian randomisation analysis found no evidence for a causal relationship between the two traits. However, gene-based analyses identified two novel loci for migraine. Also, we found significant enrichment of genes nominally associated (Pgene < 0.05) with both traits (Pbinomial-test = 9.83 × 10-6). Combining gene-based p-values across endometriosis and migraine, three genes, two (TRIM32 and SLC35G6) of which are at novel loci, were genome-wide significant. Genes having Pgene < 0.1 for both endometriosis and migraine (Pbinomial-test = 1.85 ×10-°3) were significantly enriched for biological pathways, including interleukin-1 receptor binding, focal adhesion-PI3K-Akt-mTOR-signaling, MAPK and TNF-α signalling. Our findings further confirm the comorbidity of endometriosis and migraine and indicate a non-causal relationship between the two traits, with shared genetically-controlled biological mechanisms underlying the co-occurrence of the two disorders.
Collapse
Affiliation(s)
- Emmanuel O. Adewuyi
- School of Biomedical Sciences, Faculty of Health, and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland 4000, Australia;
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, USA;
| | | | | | | | - Asa Auta
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 950-2181, Japan;
| | - Mette Nyegaard
- Department of Biomedicine – Human Genetics, Aarhus University, DK-8000 Aarhus, Denmark;
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, DK-2100 Copenhagen, Denmark
| | - Lyn R. Griffiths
- School of Biomedical Sciences, Faculty of Health, and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland 4000, Australia;
| | - Grant W. Montgomery
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia;
| | - Daniel I. Chasman
- Divisions of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA;
| | - Dale R. Nyholt
- School of Biomedical Sciences, Faculty of Health, and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland 4000, Australia;
| |
Collapse
|
31
|
Thomas N, Gurvich C, Hudaib AR, Gavrilidis E, Kulkarni J. Dissecting the syndrome of schizophrenia: Associations between symptomatology and hormone levels in women with schizophrenia. Psychiatry Res 2019; 280:112510. [PMID: 31415936 DOI: 10.1016/j.psychres.2019.112510] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/22/2022]
Abstract
Despite many studies implicating reproductive hormones in the development and outcome of schizophrenia, few have characterised the association between symptomatology and hormonal trajectories. To understand the influence of hormones on schizophrenia symptoms, serum steroids (estradiol, progesterone, follicular stimulating hormone (FSH), luteinising hormone (LH), and dehydroepiandrosterone (DHEA)) and psychopathology (The positive-and-negative-syndrome-scale(PANSS)) and depression (Montgomery-Asberg-Depression-Rating Scale(MADRS)) were collected across 12-weeks in 45 women (mean age 46) diagnosed with schizophrenia. To account for potential heterogeneity, Group-based-trajectory-modelling of psychopathology was used to identify distinct subgroups of individuals following a similar pattern of association between symptom score and hormone levels over-time. Two trajectories were identified for PANSS: one subgroup with lower symptom severity was associated with FSH, DHEA, LH, and another high severity subgroup associated with LH. Two trajectories were identified for MADRS: 'depressed' (associated with FSH), and non-depressed. The result delineates subpopulations with unique psychopathology and hormone associations that support the hypothesis that reproductive hormones play a role in the pathophysiology of schizophrenia, and that heterogeneity may exist in hormonal sensitivities in the schizophrenia population. Stratification of subjects according to biological phenotype may help improve existing treatments through personalised-medicine strategies. The endocrine system may be one such biological mechanism to continue dissecting the syndrome.
Collapse
Affiliation(s)
- Natalie Thomas
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and The Alfred Hospital, Level 4, 607 St Kilda Rd Melbourne, Australia.
| | - Caroline Gurvich
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and The Alfred Hospital, Level 4, 607 St Kilda Rd Melbourne, Australia
| | - Abdul-Rahman Hudaib
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and The Alfred Hospital, Level 4, 607 St Kilda Rd Melbourne, Australia
| | - Emorfia Gavrilidis
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and The Alfred Hospital, Level 4, 607 St Kilda Rd Melbourne, Australia
| | - Jayashri Kulkarni
- Monash Alfred Psychiatry Research Centre, Monash University Central Clinical School and The Alfred Hospital, Level 4, 607 St Kilda Rd Melbourne, Australia
| |
Collapse
|
32
|
Association between menstrual cycle irregularity and tinnitus: a nationwide population-based study. Sci Rep 2019; 9:14038. [PMID: 31575925 PMCID: PMC6773700 DOI: 10.1038/s41598-019-50559-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/11/2019] [Indexed: 01/01/2023] Open
Abstract
This population-based cross-sectional study investigated the association between menstrual cycle irregularity and tinnitus in premenopausal Korean women. We used data from the 5th Korea National Health and Nutrition Examination Survey (2010-2012). A total of 4633 premenopausal women were included. Hierarchical multivariable logistic regression analysis was performed. Individuals with tinnitus accounted for 21.6%. Women with tinnitus or menstrual irregularity had significantly higher rates of stress, depressive mood, and suicidal ideation than those without. The proportion of individuals with irregular menstrual cycles with duration of longer than 3 months increased as the severity of tinnitus increased (P = 0.01). After adjusting for confounding variables, the odds of tinnitus increased in individuals with irregular menstrual cycles compared to those with regular menstrual cycles. The odds ratios (ORs) of tinnitus tended to increase as the duration of menstrual irregularity became longer (1.37, 95% confidence interval: 1.06-1.78 for duration of up to 3 months; 1.71, 1.03-2.85 for duration of longer than 3 months, P for trend = 0.002). Our study found a positive association between menstrual cycle irregularity and tinnitus. Menstrual cycle irregularity may be a related factor of tinnitus in women with childbearing age.
Collapse
|
33
|
Rull K, Grigorova M, Ehrenberg A, Vaas P, Sekavin A, Nõmmemees D, Adler M, Hanson E, Juhanson P, Laan M. FSHB -211 G>T is a major genetic modulator of reproductive physiology and health in childbearing age women. Hum Reprod 2019; 33:954-966. [PMID: 29617818 DOI: 10.1093/humrep/dey057] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/05/2018] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Are the genetic variants FSHB -211 G>T (rs10835638), FSHR c.2039 A>G (Asn680Ser, rs6166) and FSHR -29 G>A (rs1394205) associated with serum FSH, LH and anti-Müllerian hormone (AMH) levels in reproductive age women, their menstrual cycle parameters and risk of infertility? SUMMARY ANSWER Only the FSHB -211 G>T variant was a major genetic determinant of serum gonadotropin levels in both, eumenorrheic healthy women and female infertility patients, and the T-allele carrier status was enriched among idiopathic infertility cases. WHAT IS KNOWN ALREADY There are accumulating data on common genetic variants modulating reproductive parameters and fertility potential. FSHB -211 G>T represents the strongest acknowledged genetic factor contributing to male circulating gonadotropins levels. Respective data in women are limited and the two previously published studies have reached conflicting results. In addition, previous studies have consistently associated FSHR c.2039 A>G (but not FSHR -29 G>A) with female serum FSH level. STUDY DESIGN, SIZE, DURATION The study aimed to test robust and clinically meaningful genetic effects (if present) of the FSHB -211 G>T, FSHR c.2039 A>G and FSHR -29 G>A variants on female basal FSH, LH and AMH levels, and linked reproductive parameters. Genetic association testing was performed in two independent and clinically different study groups (i) eumenorrheic healthy women without known fertility problems (n = 169; 27.6 ± 6.1 years) and (ii) female partners of infertile couples (n = 186; 32.4 ± 4.7 years). The study groups were compared for allelic and genotypic distributions of the analysed variants. PARTICIPANTS/MATERIALS, SETTING, METHODS All participants were recruited during the HAPPY PREGNANCY study (2013-2015) at the Women's Clinic, Tartu University Hospital, Estonia. Serum FSH, LH and AMH were measured in the follicular phase (Days 2-6) of the menstrual cycle. All three single nucleotide polymorphisms (SNPs) were genotyped by PCR and Taqman allelic discrimination assay. The effect of the analysed variants on hormonal measurements and menstrual cycle data was assessed using linear regression under additive and recessive models adjusted by age, BMI and smoking status. Results of the two subgroups were combined in a meta-analysis applying the fixed effects model. Restricted maximum likelihood analysis was applied to estimate the proportion of total phenotypic variance of analysed reproductive parameters, explainable by the tested genetic variants. In case-control analysis, genetic association with infertility status was tested using Fisher's exact test and logistic regression adjusted by age, BMI and smoking status. MAIN RESULTS AND THE ROLE OF CHANCE In both study groups, T-allele of the FSHB -211 G>T was associated with significantly higher serum levels of FSH and LH. Results of the meta-analysis (additive genetic model) remained significant after Bonferroni correction for multiple testing: FSH, T-allele effect 0.80 IU/L, P = 1.2 × 10-3; LH, 1.58 IU/L, P = 1.8×10-8. A more pronounced effect of T-allele of the FSHB -211 G>T on circulating LH was identified as a driving factor to increased LH/FSH ratio (meta-analysis, P = 4.7 × 10-3). In healthy women, the FSHB -211 G>T variant was estimated to explain 3.5 and 7.1% of the total variance of the measured serum FSH and LH levels, respectively. The corresponding numbers for the infertility patients were 1.6 and 10.5%. Women with idiopathic infertility compared to controls exhibited a doubled T-allele frequency (23.6 versus 12.4%; P = 8.9 × 10-3) and a >3-fold excess of TT homozygotes (5.6 versus 1.8%; P = 3.5 × 10-2). The only association of the FSHR c.2039 A>G was detected with serum FSH levels in eumenorrheic healthy women, explaining 3.9% of the total parameter variance (G-allele effect 0.56 IU/L, P = 4.6 × 10-3). In the study group of healthy reproductive age women, the highest serum FSH levels were detected among the FSHB -211 T-allele carriers with the FSHR c.2039 GG-genotype (median 7.7 IU/L). In contrast, the lowest hormone concentrations were measured for the women carrying the combination of the FSHB -211 GG- and the FSHR c.2039 AA-homozygosity (median 5.8 IU/L, P = 9.6 × 10-3). None of the analysed reproductive parameters was associated with the FSHR -29 G>A variant. In our study groups, the tested polymorphisms did not reach significant associations with serum AMH measurements, menstrual cycle length or age at menarche. LIMITATIONS, REASONS FOR CAUTION Small sample size and the design involving two clinical groups with different reproductive histories may have limited the capacity to replicate the associations with the age at menarche and length of menstrual cycle, initially reported in large genome-wide association studies. Small sample size may have also affected the accuracy in estimating the contribution of the tested variants to the total phenotypic variance of measured gonadotropin concentrations. The group of eumenorrheic healthy women had its limitations as a control to estimate the true effect of analysed genetic variants on individual's fertility potential as the recruitment strategy had been targeted mostly towards younger women, who may not yet have planned to conceive a child by this age. WIDER IMPLICATIONS OF THE FINDINGS We propose that like in men, also in women the FSHB -211 G>T represents a key genetic modulator of circulating gonadotropin, leading to various possible downstream effects on reproductive physiology. This claim is strongly supported by the reports of genome-wide association studies on various female reproductive traits and diseases. In perspective, FSHB -211 G>T may have a diagnostic value in fertility clinics to detect female patients with genetically inherited elevated basal FSH and LH levels. STUDY FUNDING/COMPETING INTEREST(S) The study was supported by Estonian Science Foundation Grant (ETF9030 for M.L.); Institutional Research Grant (IUT34-12 for M.L.) and European Union through the European Regional Development Fund (project HAPPY PREGNANCY, 3.2.0701.12-0047; for M.L. and K.R.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the article. We have no competing interests to declare. TRAIL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Kristiina Rull
- Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, Tartu 50411, Estonia.,Department of Obstetrics and Gynaecology, University of Tartu, L. Puusepa St. 8, Tartu 51014, Estonia.,Women's Clinic of Tartu University Hospital, L. Puusepa St. 8, Tartu 51014, Estonia
| | - Marina Grigorova
- Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, Tartu 50411, Estonia
| | - Aivar Ehrenberg
- Department of Obstetrics and Gynaecology, University of Tartu, L. Puusepa St. 8, Tartu 51014, Estonia.,Women's Clinic of Tartu University Hospital, L. Puusepa St. 8, Tartu 51014, Estonia
| | - Pille Vaas
- Department of Obstetrics and Gynaecology, University of Tartu, L. Puusepa St. 8, Tartu 51014, Estonia.,Women's Clinic of Tartu University Hospital, L. Puusepa St. 8, Tartu 51014, Estonia
| | - Aire Sekavin
- Department of Obstetrics and Gynaecology, University of Tartu, L. Puusepa St. 8, Tartu 51014, Estonia
| | - Diana Nõmmemees
- Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, Tartu 50411, Estonia
| | - Mart Adler
- Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, Tartu 50411, Estonia
| | - Ele Hanson
- Department of Obstetrics and Gynaecology, University of Tartu, L. Puusepa St. 8, Tartu 51014, Estonia.,Women's Clinic of Tartu University Hospital, L. Puusepa St. 8, Tartu 51014, Estonia
| | - Peeter Juhanson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, Tartu 50411, Estonia
| | - Maris Laan
- Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila St. 19, Tartu 50411, Estonia
| |
Collapse
|
34
|
Batiha O, Alahmad NA, Sindiani A, Bodoor K, Shaaban S, Al-Smadi M. Genetics of Female Infertility: Molecular Study of Newborn Ovary Homeobox Gene in Poor Ovarian Responders. J Hum Reprod Sci 2019; 12:85-91. [PMID: 31293321 PMCID: PMC6594122 DOI: 10.4103/jhrs.jhrs_112_18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background Newborn ovary homeobox (NOBOX) gene plays a critical role in the transcriptional regulation of oocyte-specific genes. Previous studies have demonstrated a pathogenic effect of NOBOX variants on premature ovarian insufficiency (POI) patients. Poor ovarian response (POR) is a risk factor for POI. Therefore, genetic variants in the NOBOX gene may also be studied as risk factors for POR development. Aims The aim of the study is to investigate the association between seven known NOBOX single-nucleotide polymorphisms (SNPs) and POR in Jordanian females. Settings and Design This was a case-control study of 60 females with POR for controlled ovarian hyperstimulation and 59 healthy females with no history of reproductive problems. Blood samples were collected from the participants and seven SNPs of NOBOX gene were screened. Subjects and Methods DNA was extracted from blood samples. Polymerase chain reaction with primers specific for seven known SNPs in NOBOX gene was used to amplify the specified region within the gene followed by Sanger sequencing. Results The seven SNPs investigated in this study, namely, rs77587352 (c.271G>T, p. Gly91Trp), rs7800847 (c.349C>T, p. Arg117Trp), rs193303102 (c.907C>T, p. Arg303X), rs193303103 (c.1025G>C, p. Ser342Thr), rs193303104 (c.1048G>T, p. Val350Leu), rs201947677 (c.1064G>A, p. Arg355His), and rs146227301 (c.1856C>T, p. Pro619Leu), only represent the wild-type allele in both females with POR and healthy participants. Conclusions The results show that only monomorphic genotype of the NOBOX variants was found in Jordanian females studied.
Collapse
Affiliation(s)
- Osamah Batiha
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Nour Alhoda Alahmad
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Amer Sindiani
- Department of Obstetrics and Gynecology, Jordan University of Science and Technology, Irbid, Jordan
| | - Khaldon Bodoor
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Sherin Shaaban
- Department of Applied Biological Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Mohammad Al-Smadi
- Department of Reproductive Endocrinology and IVF Unit, King Hussein Medical Center, Amman, Jordan
| |
Collapse
|
35
|
Conforti A, Vaiarelli A, Cimadomo D, Bagnulo F, Peluso S, Carbone L, Di Rella F, De Placido G, Ubaldi FM, Huhtaniemi I, Alviggi C. Pharmacogenetics of FSH Action in the Female. Front Endocrinol (Lausanne) 2019; 10:398. [PMID: 31293516 PMCID: PMC6606727 DOI: 10.3389/fendo.2019.00398] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/05/2019] [Indexed: 12/19/2022] Open
Abstract
The purpose of a pharmacogenomic approach is to tailor treatment on the basis of an individual human genotype. This strategy is becoming increasingly common in medicine, and important results have been obtained in oncologic and antimicrobial therapies. The rapid technological developments and availability of innovative methodologies have revealed the existence of numerous genotypes that can influence the action of medications and give rise to the idea that a true "individualized" approach could become in the future a reality in clinical practice. Moreover, compared to the past, genotype analyses are now more easily available at accessible cost. Concerning human reproduction, there is ample evidence that several variants of gonadotropins and their receptors influence female reproductive health and ovarian response to exogenous gonadotropins. In more detail, variants in genes of follicle-stimulating hormone β-chain (FSH-B) and its receptor (FSH-R) seem to be the most promising candidates for a pharmacogenomic approach to controlled ovarian stimulation in assisted reproductive technologies. In the present review, we summarize the evidence regarding FSH-B and FSH-R variants, with special reference to their impact on reproductive health and assisted reproductive technology treatments.
Collapse
Affiliation(s)
- Alessandro Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
- *Correspondence: Alessandro Conforti
| | - Alberto Vaiarelli
- G.E.N.E.R.A. Centre for Reproductive Medicine, Clinica Valle Giulia, Rome, Italy
| | - Danilo Cimadomo
- G.E.N.E.R.A. Centre for Reproductive Medicine, Clinica Valle Giulia, Rome, Italy
| | - Francesca Bagnulo
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Stefania Peluso
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Luigi Carbone
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Francesca Di Rella
- Medical Oncology, Department of Senology, National Cancer Institute, IRCCS Fondazione G. Pascale, Naples, Italy
| | - Giuseppe De Placido
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Filippo Maria Ubaldi
- G.E.N.E.R.A. Centre for Reproductive Medicine, Clinica Valle Giulia, Rome, Italy
| | - Ilpo Huhtaniemi
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Carlo Alviggi
- Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, Naples, Italy
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale (IEOS) Consiglio Nazionale delle Ricerche, Naples, Italy
| |
Collapse
|
36
|
Trevisan CM, de Oliveira R, Christofolini DM, Barbosa CP, Bianco B. Effects of a Polymorphism in the Promoter Region of the Follicle-Stimulating Hormone Subunit Beta (FSHB) Gene on Female Reproductive Outcomes. Genet Test Mol Biomarkers 2018; 23:39-44. [PMID: 30585745 DOI: 10.1089/gtmb.2018.0182] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Follicle-stimulating hormone (FSH) is essential to the hypothalamic-pituitary-gonadal axis, playing a key role in human reproduction. It is a heterodimer comprised of a hormone-specific β-chain (FSH-β) that is associated with an α-chain. It exerts its biological activities by binding to the FSH receptor (FSHR). The β-subunit, which is encoded by the FSHB gene, is responsible for ensuring binding specificity to the FSHR. There is a promoter polymorphism in this gene, c.-211G>T (rs10835638), upstream of the transcription start site; and in vitro studies have reported that the T allele decreases FSHB transcription in gonadotrophic cells. AIMS Investigate the possible effects of the FSHB c.-211G/T polymorphism on hormonal profile and in in vitro fertilization (IVF)/intracytoplasmic sperm injection outcomes in normoovulatory Brazilian women. METHODS A cross-sectional study of 140 women (median age = 33 years [CI: 32-34]) with infertility mainly caused by male (n = 85) or tuboperitoneal (n = 55) factors. In this study we evaluated FSH, estradiol, luteinizing hormone (LH), progesterone, prolactin and anti-Mullerian hormone levels, and antral follicle counting (AFC). Genotyping was performed using the TaqMan real-time polymerase chain reaction methodology. RESULTS The wild-type allele G was found in 86.4% and the polymorphic allele T in 13.6% of the women respectively. The TT genotype was not found in any women. Women carrying the GT genotype had a poorer response more frequently to controlled ovarian hyperstimulation when compared to individuals with the GG genotype (47.4% vs. 26.5%, p = 0.010), higher LH levels (3.1 IU/mL vs. 2.4 IU/mL, p = <0.001), lower AFC (8.0 vs. 10.0, p = 0.03), oocytes retrieved (3.0 vs. 5.0, p = 0.03), MII (3.0 vs. 4.0, p = 0.02), and embryos (2.0 vs. 3.0, p = 0.02). Despite these findings, no difference was observed in pregnancy rate. CONCLUSION Our findings suggest that the FSHB c.-211G/T polymorphism may modestly alter some aspects of the female reproductive system, but they are not associated with significantly different IVF outcomes.
Collapse
Affiliation(s)
- Camila Martins Trevisan
- Human Reproduction and Genetics, Department of Collective Health, Faculdade de Medicina do ABC , Santo André/SP, Brazil
| | - Renato de Oliveira
- Human Reproduction and Genetics, Department of Collective Health, Faculdade de Medicina do ABC , Santo André/SP, Brazil
| | - Denise Maria Christofolini
- Human Reproduction and Genetics, Department of Collective Health, Faculdade de Medicina do ABC , Santo André/SP, Brazil
| | - Caio Parente Barbosa
- Human Reproduction and Genetics, Department of Collective Health, Faculdade de Medicina do ABC , Santo André/SP, Brazil
| | - Bianca Bianco
- Human Reproduction and Genetics, Department of Collective Health, Faculdade de Medicina do ABC , Santo André/SP, Brazil
| |
Collapse
|
37
|
Laisk T, Kukuškina V, Palmer D, Laber S, Chen CY, Ferreira T, Rahmioglu N, Zondervan K, Becker C, Smoller JW, Lippincott M, Salumets A, Granne I, Seminara S, Neale B, Mägi R, Lindgren CM. Large-scale meta-analysis highlights the hypothalamic-pituitary-gonadal axis in the genetic regulation of menstrual cycle length. Hum Mol Genet 2018; 27:4323-4332. [PMID: 30202859 PMCID: PMC6276838 DOI: 10.1093/hmg/ddy317] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/17/2018] [Accepted: 09/03/2018] [Indexed: 12/31/2022] Open
Abstract
The normal menstrual cycle requires a delicate interplay between the hypothalamus, pituitary and ovary. Therefore, its length is an important indicator of female reproductive health. Menstrual cycle length has been shown to be partially controlled by genetic factors, especially in the follicle-stimulating hormone beta-subunit (FSHB) locus. A genome-wide association study meta-analysis of menstrual cycle length in 44 871 women of European ancestry confirmed the previously observed association with the FSHB locus and identified four additional novel signals in, or near, the GNRH1, PGR, NR5A2 and INS-IGF2 genes. These findings not only confirm the role of the hypothalamic-pituitary-gonadal axis in the genetic regulation of menstrual cycle length but also highlight potential novel local regulatory mechanisms, such as those mediated by IGF2.
Collapse
Affiliation(s)
- Triin Laisk
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Viktorija Kukuškina
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Duncan Palmer
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Samantha Laber
- Big Data Institute, Li Ka Shing Center for Health for Health Information and Discovery, Oxford University, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Chia-Yen Chen
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Boston, Massachusetts, USA
| | - Teresa Ferreira
- Big Data Institute, Li Ka Shing Center for Health for Health Information and Discovery, Oxford University, Oxford, UK
| | - Nilufer Rahmioglu
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Krina Zondervan
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, University of Oxford, UK
| | - Christian Becker
- Oxford Endometriosis CaRe Centre, Nuffield Department of Women’s and Reproductive Health, University of Oxford, UK
| | - Jordan W Smoller
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Margaret Lippincott
- Harvard Reproductive Sciences Center and Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Andres Salumets
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Biomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Ingrid Granne
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, UK
| | - Stephanie Seminara
- Harvard Reproductive Sciences Center and Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Benjamin Neale
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Cecilia M Lindgren
- Big Data Institute, Li Ka Shing Center for Health for Health Information and Discovery, Oxford University, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Program in Medical and Population Genetics, Broad Institute, Boston, MA, USA
| |
Collapse
|
38
|
Guo LC, Pan S, Yu S, Liu T, Xiao J, Zhu B, Qu Y, Huang W, Li M, Li X, Zeng W, Rutherford S, Lin L, Zhang Y, Ma W. Human Sex Hormone Disrupting Effects of New Flame Retardants and Their Interactions with Polychlorinated Biphenyls, Polybrominated Diphenyl Ethers, a Case Study in South China. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:13935-13941. [PMID: 30384584 DOI: 10.1021/acs.est.8b01540] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Even though sex hormone disrupting effects of polychlorinated biphenyls (PCBs) and polybrominated diphenyl ethers (PBDEs) are widely understood, similar effects associated with new flame retardants (NFRs) have not been so well studied. This study aimed to explore the sex hormone disruption of NFRs and their interactions with PCBs and PBDEs through the conduct of an ecological study in an e-waste dismantling and control region in South China. Questionnaires and blood samples were collected from local adult residents. Results of generalized additive model and linear regression analyses indicate that several species of NFRs showed similar disrupting effects with PBDE congeners on female follicle-stimulating hormone (FSH) and male testosterone. Judged by the curved shape and statistical significance, ΣNFR (sum of 8 species of NFRs) showed stronger disrupting effects on male testosterone and female FSH compared to ΣPBDE (sum of 13 congeners of PBDEs). The interactions induced by NFRs complicated the original sex hormone disruption led by PCBs and PBDEs. The disrupting effects and interactions induced by NFRs decreased female FSH levels in the exposed group. Comprehensive evaluation is needed to provide the evidence base for judging the health risks arising from the increased usage of NFRs.
Collapse
Affiliation(s)
- Ling-Chuan Guo
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
| | - Shangxia Pan
- Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
| | - Shengbing Yu
- Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
| | - Tao Liu
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
| | - Jianpeng Xiao
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
- School of Public Health , Southern Medical University , Guangzhou 510515 , China
| | - Binghui Zhu
- Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
| | - Yabin Qu
- Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
| | - Weixiong Huang
- Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
| | - Min Li
- Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
| | - Xing Li
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
- School of Public Health , Southern Medical University , Guangzhou 510515 , China
| | - Weilin Zeng
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
| | - Shannon Rutherford
- Centre for Environment and Population Health, School of Medicine , Griffith University , Brisbane , Queensland 4111 , Australia
| | - Lifeng Lin
- Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
| | - Yonghui Zhang
- Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
| | - Wenjun Ma
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention , Guangzhou 511430 , China
| |
Collapse
|
39
|
Gajbhiye R, Fung JN, Montgomery GW. Complex genetics of female fertility. NPJ Genom Med 2018; 3:29. [PMID: 30345074 PMCID: PMC6185946 DOI: 10.1038/s41525-018-0068-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 08/13/2018] [Accepted: 09/12/2018] [Indexed: 01/10/2023] Open
Abstract
Variation in reproductive lifespan and female fertility have implications for health, population size and ageing. Fertility declines well before general signs of menopause and is also adversely affected by common reproductive diseases, including polycystic ovarian syndrome (PCOS) and endometriosis. Understanding the factors that regulate the timing of puberty and menopause, and the relationships with fertility are important for individuals and for policy. Substantial genetic variation exists for common traits associated with reproductive lifespan and for common diseases influencing female fertility. Genetic studies have identified mutations in genes contributing to disorders of reproduction, and in the last ten years, genome-wide association studies (GWAS) have transformed our understanding of common genetic contributions to these complex traits and diseases. These studies have made great progress towards understanding the genetic factors contributing to variation in traits and diseases influencing female fertility. The data emerging from GWAS demonstrate the utility of genetics to explain epidemiological observations, revealing shared biological pathways linking puberty timing, fertility, reproductive ageing and health outcomes. Many variants implicate DNA damage/repair genes in variation in the age at menopause with implications for follicle health and ageing. In addition to the discovery of individual genes and pathways, the increasingly powerful studies on common genetic risk factors help interpret the underlying relationships and direction of causation in the regulation of reproductive lifespan, fertility and related traits.
Collapse
Affiliation(s)
- Rahul Gajbhiye
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, QLD 4072 Australia
- Department of Clinical Research, ICMR-National Institute for Research in Reproductive Health, J. M. Street, Parel Mumbai, 400012 India
| | - Jenny N. Fung
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, QLD 4072 Australia
| | - Grant W. Montgomery
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, QLD 4072 Australia
| |
Collapse
|
40
|
An Ancient Fecundability-Associated Polymorphism Creates a GATA2 Binding Site in a Distal Enhancer of HLA-F. Am J Hum Genet 2018; 103:509-521. [PMID: 30245028 DOI: 10.1016/j.ajhg.2018.08.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 08/17/2018] [Indexed: 12/20/2022] Open
Abstract
Variation in female reproductive traits, such as fertility, fecundity, and fecundability, are heritable in humans, but identifying and functionally characterizing genetic variants associated with these traits have been challenging. Here, we explore the functional significance and evolutionary history of a G/A polymorphism at SNP rs2523393, which is an eQTL for HLA-F and is significantly associated with fecundability (the probability of being pregnant within a single menstrual cycle). We replicated the association between the rs2523393 genotype and HLA-F expression by using GTEx data and demonstrate that HLA-F is upregulated in the endometrium during the window of implantation and by progesterone in decidual stromal cells. Next, we show that the rs2523393 A allele creates a GATA2 binding site in a progesterone-responsive distal enhancer that loops to the HLA-F promoter. Remarkably, we found that the A allele is derived in the human lineage and that the G/A polymorphism arose before the divergence of modern and archaic humans and segregates at intermediate to high frequencies across human populations. Remarkably, the derived A allele is has also been identified in a GWAS as a risk allele for multiple sclerosis. These data suggest that the polymorphism is maintained by antagonistic pleiotropy and a reproduction-health tradeoff in human evolution.
Collapse
|
41
|
van der Kemp J, van der Schouw YT, Asselbergs FW, Onland-Moret NC. Women-specific risk factors for heart failure: A genetic approach. Maturitas 2018; 109:104-111. [DOI: 10.1016/j.maturitas.2017.12.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 12/22/2017] [Accepted: 12/29/2017] [Indexed: 02/07/2023]
|
42
|
Sapkota Y, Steinthorsdottir V, Morris AP, Fassbender A, Rahmioglu N, De Vivo I, Buring JE, Zhang F, Edwards TL, Jones S, O D, Peterse D, Rexrode KM, Ridker PM, Schork AJ, MacGregor S, Martin NG, Becker CM, Adachi S, Yoshihara K, Enomoto T, Takahashi A, Kamatani Y, Matsuda K, Kubo M, Thorleifsson G, Geirsson RT, Thorsteinsdottir U, Wallace LM, Yang J, Velez Edwards DR, Nyegaard M, Low SK, Zondervan KT, Missmer SA, D'Hooghe T, Montgomery GW, Chasman DI, Stefansson K, Tung JY, Nyholt DR. Meta-analysis identifies five novel loci associated with endometriosis highlighting key genes involved in hormone metabolism. Nat Commun 2017; 8:15539. [PMID: 28537267 PMCID: PMC5458088 DOI: 10.1038/ncomms15539] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 04/07/2017] [Indexed: 12/27/2022] Open
Abstract
Endometriosis is a heritable hormone-dependent gynecological disorder, associated with severe pelvic pain and reduced fertility; however, its molecular mechanisms remain largely unknown. Here we perform a meta-analysis of 11 genome-wide association case-control data sets, totalling 17,045 endometriosis cases and 191,596 controls. In addition to replicating previously reported loci, we identify five novel loci significantly associated with endometriosis risk (P<5 × 10-8), implicating genes involved in sex steroid hormone pathways (FN1, CCDC170, ESR1, SYNE1 and FSHB). Conditional analysis identified five secondary association signals, including two at the ESR1 locus, resulting in 19 independent single nucleotide polymorphisms (SNPs) robustly associated with endometriosis, which together explain up to 5.19% of variance in endometriosis. These results highlight novel variants in or near specific genes with important roles in sex steroid hormone signalling and function, and offer unique opportunities for more targeted functional research efforts.
Collapse
Affiliation(s)
- Yadav Sapkota
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | - Andrew P. Morris
- Department of Biostatistics, University of Liverpool, Liverpool L69 3GL, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Amelie Fassbender
- KULeuven, Department of Development and Regeneration, Organ systems, 3000 Leuven, Belgium
- Department of Obstetrics and Gynaecology, Leuven University Fertility Centre, University Hospital Leuven, 3000 Leuven, Belgium
| | - Nilufer Rahmioglu
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Immaculata De Vivo
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Julie E. Buring
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
| | - Futao Zhang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Todd L. Edwards
- Institute of Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA
| | - Sarah Jones
- Vanderbilt Genetics Institute, Division of Epidemiology, Institute of Medicine and Public Health, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA
| | - Dorien O
- KULeuven, Department of Development and Regeneration, Organ systems, 3000 Leuven, Belgium
- Department of Obstetrics and Gynaecology, Leuven University Fertility Centre, University Hospital Leuven, 3000 Leuven, Belgium
| | - Daniëlle Peterse
- KULeuven, Department of Development and Regeneration, Organ systems, 3000 Leuven, Belgium
- Department of Obstetrics and Gynaecology, Leuven University Fertility Centre, University Hospital Leuven, 3000 Leuven, Belgium
| | - Kathryn M. Rexrode
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
| | - Paul M. Ridker
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
| | - Andrew J. Schork
- Cognitive Science Department, University of California, San Diego, La Jolla, California 92093, USA
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Copenhagen University Hospital, DK-2100 Copenhagen, Denmark
| | - Stuart MacGregor
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Nicholas G. Martin
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Christian M. Becker
- Endometriosis CaRe Centre, Nuffield Dept of Obstetrics & Gynaecology, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Sosuke Adachi
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 950-2181, Japan
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 950-2181, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 950-2181, Japan
| | - Atsushi Takahashi
- Center for Integrative Medical Sciences, RIKEN, Yokohama 230-0045, Japan
| | - Yoichiro Kamatani
- Center for Integrative Medical Sciences, RIKEN, Yokohama 230-0045, Japan
| | - Koichi Matsuda
- Institute of Medical Sciences, The University of Tokyo, Tokyo 108-8639, Japan
| | - Michiaki Kubo
- Center for Integrative Medical Sciences, RIKEN, Yokohama 230-0045, Japan
| | | | - Reynir T. Geirsson
- Department of Obstetrics and Gynecology, Landspitali University Hospital, 101 Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, 101 Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen, 101 Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, 101 Reykjavik, Iceland
| | - Leanne M. Wallace
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jian Yang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Digna R. Velez Edwards
- Vanderbilt Genetics Institute, Vanderbilt Epidemiology Center, Institute of Medicine and Public Health, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA
| | - Mette Nyegaard
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
- iPSYCH, The Lundbeck Foundation Initiative for Integrative Psychiatric Research, DK-2100 Copenhagen, Denmark
| | - Siew-Kee Low
- Center for Integrative Medical Sciences, RIKEN, Yokohama 230-0045, Japan
| | - Krina T. Zondervan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Endometriosis CaRe Centre, Nuffield Dept of Obstetrics & Gynaecology, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Stacey A. Missmer
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Thomas D'Hooghe
- KULeuven, Department of Development and Regeneration, Organ systems, 3000 Leuven, Belgium
- Department of Obstetrics and Gynaecology, Leuven University Fertility Centre, University Hospital Leuven, 3000 Leuven, Belgium
- Global Medical Affairs Fertility, Research and Development, Merck KGaA, Darmstadt, Germany
| | - Grant W. Montgomery
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Daniel I. Chasman
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
| | - Kari Stefansson
- deCODE Genetics/Amgen, 101 Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, 101 Reykjavik, Iceland
| | - Joyce Y. Tung
- 23andMe, Inc., 899 W. Evelyn Avenue, Mountain View, California 94041, USA
| | - Dale R. Nyholt
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland 4059, Australia
| |
Collapse
|
43
|
Busch AS, Hagen CP, Main KM, Pereira A, Corvalan C, Almstrup K, Mericq V, Juul A. Genetic Variation of Follicle-Stimulating Hormone Action Is Associated With Age at Testicular Growth in Boys. J Clin Endocrinol Metab 2017; 102:1740-1749. [PMID: 28323923 DOI: 10.1210/jc.2016-4013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/22/2017] [Indexed: 02/07/2023]
Abstract
CONTEXT Although genetic factors play a pivotal role in male pubertal timing, genome-wide association studies have identified only a few loci. Genetic variation of follicle-stimulating hormone (FSH) action affects adult reproductive parameters and female pubertal timing. OBJECTIVE To investigate whether genetic variation affecting FSH action is associated with onset of puberty in boys. DESIGN Cross-sectional and longitudinal study of two cohorts of healthy boys. SETTING This was a population-based study. PATIENTS OR OTHER PARTICIPANTS Danish (n = 1130) and Chilean (n = 424) boys were followed through puberty and genotyped for FSHB c.-211G>T, FSHR c.-29A>G, and FSHR c.2039G>A. MAIN OUTCOME MEASURES Clinical pubertal staging including orchidometry, anthropometry, and serum gonadotropin levels. RESULTS Although the cohorts differed markedly (e.g., body composition and genotype frequencies), genetic variation affecting FSH production (FSHB c.-211G>T) was associated with age at pubertal onset, as assessed by testicular enlargement, in both cohorts. The effect appeared further modified by coexistence of genetic variation affecting FSH sensitivity (FSHR c.-29G>A): After correcting for body mass index (BMI), boys with a ligand-receptor variant combination resulting in weak FSH action (i.e., FSHB c.-211GT/TT and FSHR c.-29AA) entered puberty 0.64 years [95% confidence interval (CI), 0.12 to 1.17 years; Denmark] and 0.94 years (95% CI, 0.00 to 1.88 years; Chile) later than boys with the most effective FSH action. Effects explained 1.7% (Denmark) and 1.5% (Chile) of the variance. In addition, BMI z score was negatively associated with pubertal timing (β = -0.35 years in both cohorts), explaining 17.2% (Denmark) and 7.2% (Chile) of the variance. CONCLUSION In two ethnically distinct populations, we independently identified an association of two genetic loci with male pubertal timing.
Collapse
Affiliation(s)
- Alexander S Busch
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Casper P Hagen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Katharina M Main
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Anita Pereira
- Institute of Nutrition and Food Technology, University of Chile, Casilla 226-3, Santiago 8360160, Chile
| | - Camila Corvalan
- Institute of Nutrition and Food Technology, University of Chile, Casilla 226-3, Santiago 8360160, Chile
| | - Kristian Almstrup
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Veronica Mericq
- Institute of Maternal and Child Research, Faculty of Medicine, University of Chile, Casilla 226-3, Santiago 8360160, Chile
| | - Anders Juul
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
- International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
44
|
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
|
45
|
Mika KM, Lynch VJ. An Ancient Fecundability-Associated Polymorphism Switches a Repressor into an Enhancer of Endometrial TAP2 Expression. Am J Hum Genet 2016; 99:1059-1071. [PMID: 27745831 DOI: 10.1016/j.ajhg.2016.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/02/2016] [Indexed: 12/25/2022] Open
Abstract
Variation in female reproductive traits, such as fertility, fecundity, and fecundability, is heritable in humans, but identifying and functionally characterizing genetic variants associated with these traits has been challenging. Here, we explore the functional significance and evolutionary history of a T/C polymorphism of SNP rs2071473, which we have previously shown is an eQTL for TAP2 and significantly associated with fecundability (time to pregnancy). We replicated the association between the rs2071473 genotype and TAP2 expression by using GTEx data and demonstrated that TAP2 is expressed by decidual stromal cells at the maternal-fetal interface. Next, we showed that rs2071473 is located within a progesterone-responsive cis-regulatory element that functions as a repressor with the T allele and an enhancer with the C allele. Remarkably, we found that this polymorphism arose before the divergence of modern and archaic humans, segregates at intermediate to high frequencies across human populations, and has genetic signatures of long-term balancing selection. This variant has also previously been identified in genome-wide association studies of immune-related disease, suggesting that both alleles are maintained as a result of antagonistic pleiotropy.
Collapse
|
46
|
The expanding and contracting roles of the genome in regulation of the ovarian reserve. J Assist Reprod Genet 2016; 33:1121-2. [PMID: 27535836 DOI: 10.1007/s10815-016-0789-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
47
|
Laisk-Podar T, Lindgren CM, Peters M, Tapanainen JS, Lambalk CB, Salumets A, Mägi R. Ovarian Physiology and GWAS: Biobanks, Biology, and Beyond. Trends Endocrinol Metab 2016; 27:516-528. [PMID: 27221566 PMCID: PMC7610559 DOI: 10.1016/j.tem.2016.04.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/19/2016] [Accepted: 04/26/2016] [Indexed: 12/17/2022]
Abstract
Ovarian function is central to female fertility, and several genome-wide association studies (GWAS) have been carried out to elucidate the genetic background of traits and disorders that reflect and affect ovarian physiology. While GWAS have been successful in reporting numerous genetic associations and highlighting involved pathways relevant to reproductive aging, for ovarian disorders, such as premature ovarian insufficiency and polycystic ovary syndrome, research has lagged behind due to insufficient study sample size. Novel approaches to study design and analysis methods that help to fit GWAS findings into biological context will improve our knowledge about genetics governing ovarian function in fertility and disease, and provide input for clinical tools and better patient management.
Collapse
Affiliation(s)
- Triin Laisk-Podar
- Women's Clinic, University of Tartu, Tartu 51014, Estonia; Competence Centre on Health Technologies, Tartu 50410, Estonia.
| | - Cecilia M Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Big Data Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Maire Peters
- Women's Clinic, University of Tartu, Tartu 51014, Estonia; Competence Centre on Health Technologies, Tartu 50410, Estonia
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki 00014, Finland; Department of Obstetrics and Gynecology, University Hospital of Oulu, University of Oulu, Medical Research Center Oulu and PEDEGO Research Unit, Oulu 90029, Finland
| | - Cornelis B Lambalk
- Department of Obstetrics and Gynecology, VU University Medical Centre, Amsterdam 1007 MB, Netherlands
| | - Andres Salumets
- Women's Clinic, University of Tartu, Tartu 51014, Estonia; Competence Centre on Health Technologies, Tartu 50410, Estonia; Institute of Bio- and Translational Medicine, University of Tartu, Tartu 50411, Estonia
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| |
Collapse
|