101
|
Liu D, Gao X, Pan XF, Zhou T, Zhu C, Li F, Fan JG, Targher G, Zhao J. The hepato-ovarian axis: genetic evidence for a causal association between non-alcoholic fatty liver disease and polycystic ovary syndrome. BMC Med 2023; 21:62. [PMID: 36800955 PMCID: PMC9940436 DOI: 10.1186/s12916-023-02775-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 02/09/2023] [Indexed: 02/21/2023] Open
Abstract
BACKGROUND Recent studies found associations between non-alcoholic fatty liver disease (NAFLD) and polycystic ovary syndrome (PCOS), but the causal nature of this association is still uncertain. METHODS We performed a bidirectional two-sample Mendelian randomization (MR) analysis to test for the causal association between NAFLD and PCOS using data from a large-scale biopsy-confirmed NAFLD genome-wide association study (GWAS) (1483 cases and 17,781 controls) and PCOS GWAS (10,074 cases and 103,164 controls) in European ancestries. Data from glycemic-related traits GWAS (in up to 200,622 individuals) and sex hormones GWAS (in 189,473 women) in the UK Biobank (UKB) were used in the MR mediation analysis to assess potential mediating roles of these molecules in the causal pathway between NAFLD and PCOS. Replication analysis was conducted using two independent datasets from NAFLD and PCOS GWASs in the UKB and a meta-analysis of data from FinnGen and the Estonian Biobank, respectively. A linkage disequilibrium score regression was conducted to assess genetic correlations between NAFLD, PCOS, glycemic-related traits, and sex hormones using full summary statistics. RESULTS Individuals with higher genetic liability to NAFLD were more likely to develop PCOS (OR per one-unit log odds increase in NAFLD: 1.10, 95% CI: 1.02-1.18; P = 0.013). Indirect causal effects of NAFLD on PCOS via fasting insulin only (OR: 1.02, 95% CI: 1.01-1.03; P = 0.004) and further a suggestive indirect causal effect via fasting insulin in concert with androgen levels were revealed in MR mediation analyses. However, the conditional F statistics of NAFLD and fasting insulin were less than 10, suggesting likely weak instrument bias in the MVMR and MR mediation analyses. CONCLUSIONS Our study suggests that genetically predicted NAFLD was associated with a higher risk of developing PCOS but less evidence for vice versa. Fasting insulin and sex hormones might mediate the link between NAFLD and PCOS.
Collapse
Affiliation(s)
- Dong Liu
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Xue Gao
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiong-Fei Pan
- Ministry of Education Key Laboratory of Birth Defects and Related Diseases in Women and Children, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Shuangliu Institute of Women's and Children's Health, Shuangliu Maternal and Child Health Hospital, Chengdu, Sichuan, China
| | - Tao Zhou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Cairong Zhu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fei Li
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.,Department of Developmental and Behavioral Pediatric & Child Primary Care, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Maternal and Child Health, School of Public Health, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Verona, Verona, Italy
| | - Jian Zhao
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China. .,Department of Maternal and Child Health, School of Public Health, Shanghai Jiao Tong University, Shanghai, China. .,MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
| |
Collapse
|
102
|
Alan Harris R, Archer KJ, Goodarzi MO, York TP, Rogers J, Dunaif A, McAllister JM, Strauss JF. Loci on chromosome 12q13.2 encompassing ERBB3, PA2G4 and RAB5B are associated with polycystic ovary syndrome. Gene 2023; 852:147062. [PMID: 36423778 PMCID: PMC9811427 DOI: 10.1016/j.gene.2022.147062] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
Polycystic ovary syndrome (PCOS) is characterized by hyperandrogenemia of ovarian theca cell origin. We report significant association of androgen production with 15 single nucleotide variants (SNVs) identified by exome sequencing of theca cells from women with PCOS and normal ovulatory women. Ten SNVs are located within a 150 kbp region on 12q13.2 which encompasses loci identified in PCOS genome-wide association studies (GWAS) and contains PCOS candidate genes ERBB3 and RAB5B. The region also contains PA2G4 which encodes a transcriptional corepressor of androgen receptor and androgen receptor-regulated genes. PA2G4 has not previously been recognized as related to PCOS in published GWAS studies. Two of the SNVs are predicted to have functional consequences (ERBB3 missense SNV, PA2G4 promoter SNV). PA2G4 interacts with the ERBB3 cytoplasmic domain containing the missense variant, suggesting a potential signaling pathway disruption that could lead to the PCOS ovarian phenotype. Single cell RNA sequencing of theca cells showed significantly less expression of PA2G4 after forskolin treatment in PCOS cells compared to normal cells (padj = 3.82E-30) and in cells heterozygous for the PA2G4 promoter SNV compared to those without the SNV (padj = 2.16E-11). This is consistent with a functional effect of the PA2G4 promoter SNV. No individual SNV was significantly associated with PCOS in an independent family cohort, but a haplotype with minor alleles of three SNVs was found preferentially in women with PCOS. These findings suggest a functional role for 12q13.2 variants in PCOS and implicate variants in ERBB3 and PA2G4 in the pathophysiology of PCOS.
Collapse
Affiliation(s)
- R Alan Harris
- Human Genome Sequencing Center and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA.
| | - Kellie J Archer
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH 43210 USA.
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.
| | - Timothy P York
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298 USA; Department of Obstetrics and Gynecology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA.
| | - Jeffrey Rogers
- Human Genome Sequencing Center and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA.
| | - Andrea Dunaif
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA.
| | - Jan M McAllister
- Department of Pathology, Penn State Hershey College of Medicine, Hershey, PA 17033 USA.
| | - Jerome F Strauss
- Department of Obstetrics and Gynecology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298 USA; Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104 USA.
| |
Collapse
|
103
|
Insulin Metabolism in Polycystic Ovary Syndrome: Secretion, Signaling, and Clearance. Int J Mol Sci 2023; 24:ijms24043140. [PMID: 36834549 PMCID: PMC9962893 DOI: 10.3390/ijms24043140] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/23/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine and metabolic disorder in women of reproductive age. Its heterogeneous clinical presentation is characterized by hyperandrogenemia, reproductive changes, polycystic ovary morphology, and insulin resistance (IR). The primary pathophysiological process in its multifactorial etiology has not yet been identified. However, the two most proposed core etiologies are the disruption of insulin metabolism and hyperandrogenemia, both of which begin to intertwine and propagate each other in the later stages of the disease. Insulin metabolism can be viewed as the interconnectedness of beta cell function, IR or insulin sensitivity, and insulin clearance. Previous studies of insulin metabolism in PCOS patients have yielded conflicting results, and literature reviews have focused mainly on the molecular mechanisms and clinical implications of IR. In this narrative review, we comprehensively explored the role of insulin secretion, clearance, and decreased sensitivity in target cells as a potential primary insult in PCOS pathogenesis, along with the molecular mechanism behind IR in PCOS.
Collapse
|
104
|
Haddad-Filho H, Tosatti JAG, Vale FM, Gomes KB, Reis FM. Updates in diagnosing polycystic ovary syndrome-related infertility. Expert Rev Mol Diagn 2023; 23:123-132. [PMID: 36856088 DOI: 10.1080/14737159.2023.2177536] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is a condition that affects approximately 13% of reproductive age women and is characterized by androgen excess, menstrual irregularity and altered ovarian morphology. PCOS presents a complex etiology and pathophysiology, which still requires a detailed investigation of biochemical signatures to identify the molecules and mechanisms that govern it. AREAS COVERED This narrative review summarizes the main molecular alterations found in the ovarian follicular fluid, endometrium and placenta of women with PCOS, and the genotypes potentially associated with the outcome of infertility treatments in PCOS. EXPERT OPINION PCOS is associated with multiple alterations in growth factors, sex steroid hormones, reactive oxygen species, proinflammatory cytokines and adipokines, which contribute to follicle arrest/ anovulation or suboptimal corpus luteum function, and ultimately to menstrual irregularity and hyperandrogenic symptoms. A panel of PCOS biomarkers should include, besides ovarian products, markers of adipose tissue function, insulin resistance, vascular health, and low-grade chronic inflammation. The effects of ovarian stimulation drugs on infertile women with PCOS are likely to be modified by genetic factors, but the available evidence is heterogeneous; therefore, future studies should evaluate standard treatments and pre-specified outcomes of interest to provide more conclusive answers.
Collapse
Affiliation(s)
- Hélio Haddad-Filho
- Graduate Program in Surgery, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Department of Medicine, Universidade Federal de Lavras, Lavras, Brazil
| | - Jéssica A G Tosatti
- Department of Clinical and Toxicological Analyzes - Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda M Vale
- Department of Clinical and Toxicological Analyzes - Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Karina B Gomes
- Department of Clinical and Toxicological Analyzes - Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando M Reis
- Division of Human Reproduction, Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
105
|
Woolner AM, Bhattacharya S. Intergenerational trends in reproduction: Infertility and pregnancy loss. Best Pract Res Clin Obstet Gynaecol 2023; 86:102305. [PMID: 36639284 DOI: 10.1016/j.bpobgyn.2022.102305] [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: 08/31/2022] [Revised: 11/25/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
This review article summarises the evidence for intergenerational trends observed to date within infertility and pregnancy loss. There appears to be evidence of intergenerational trends between mothers and daughters for the age at menopause, endometriosis, polycystic ovarian syndrome (PCOS), male factor infertility and miscarriage. At present, there is no evidence for a predisposition to stillbirth between mothers and daughters. One study found an association with familial predisposition for ectopic pregnancy. Very few studies have considered the potential for paternal transmission of risk of infertility or pregnancy loss. The majority of studies to date have significant limitations because of their observational design, risk of recall bias and risk of confounding. Therefore, high-quality well-designed research, with multi-centre collaboration and utilisation of registry-based data sources and individual patient data, is needed to understand whether infertility and pregnancy loss may have heritable factors. Epidemiological findings need to be followed up and investigated with translational research to determine the possible causalities as well as any implications for clinical practice.
Collapse
Affiliation(s)
- Andrea Mf Woolner
- Aberdeen Centre for Women's Health Research, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, United Kingdom.
| | - Siladitya Bhattacharya
- Aberdeen Centre for Women's Health Research, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, United Kingdom.
| |
Collapse
|
106
|
Lyle SM, Ahmed S, Elliott JE, Stener-Victorin E, Nachtigal MW, Drögemöller BI. Transcriptome-wide association analyses identify an association between ARL14EP and polycystic ovary syndrome. J Hum Genet 2023; 68:347-353. [PMID: 36720993 DOI: 10.1038/s10038-023-01120-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 02/02/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder, which is accompanied by a variety of comorbidities including metabolic, reproductive, and psychiatric disorders. Genome-wide association studies have identified several genetic variants that are associated with PCOS. However, these variants often occur outside of coding regions and require further investigation to understand their contribution to PCOS. A transcriptome-wide association study (TWAS) was performed to uncover heritable gene expression profiles that are associated with PCOS in two independent cohorts. Causal gene prioritization was subsequently performed and expression of genes prioritized through these analyses was examined in 49 PCOS patients and 30 controls. TWAS analyses revealed that increased expression of ARL14EP was significantly associated with PCOS risk in the discovery (P = 1.6 × 10-6) and replication cohorts (P = 2.0 × 10-13). Gene prioritization pipelines provided further evidence that ARL14EP is the most likely causal gene at this locus. ARL14EP gene expression was shown to be significantly different between PCOS cases and controls, after adjusting for body mass index, age and testosterone levels (P = 1.2 × 10-13). This study has provided evidence for the role of ARL14EP in PCOS. Given that ARL14EP has been reported to play an important role in chromatin remodeling, variants affecting the expression of ARL14EP may also affect the expression of other genes that contribute to PCOS pathogenesis.
Collapse
Affiliation(s)
- Sarah M Lyle
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Samah Ahmed
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Jason E Elliott
- Department of Obstetrics, Gynecology and Reproductive Sciences, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.,Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | | | - Mark W Nachtigal
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.,Department of Obstetrics, Gynecology and Reproductive Sciences, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.,CancerCare Manitoba Research Institute, Winnipeg, MB, Canada
| | - Britt I Drögemöller
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada. .,Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada. .,CancerCare Manitoba Research Institute, Winnipeg, MB, Canada.
| |
Collapse
|
107
|
Pei Y, Risal S, Jiang H, Lu H, Lindgren E, Stener-Victorin E, Deng Q. Transcriptomic survey of key reproductive and metabolic tissues in mouse models of polycystic ovary syndrome. Commun Biol 2023; 6:69. [PMID: 36653487 PMCID: PMC9849269 DOI: 10.1038/s42003-022-04362-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/09/2022] [Indexed: 01/19/2023] Open
Abstract
Excessive androgen production and obesity are key to polycystic ovary syndrome (PCOS) pathogenesis. Prenatal androgenized (PNA), peripubertal androgenized, and overexpression of nerve growth factor in theca cells (17NF) are commonly used PCOS-like mouse models and diet-induced maternal obesity model is often included for comparsion. To reveal the molecular features of these models, we have performed transcriptome survey of the hypothalamus, adipose tissue, ovary and metaphase II (MII) oocytes. The largest number of differentially expressed genes (DEGs) is found in the ovaries of 17NF and in the adipose tissues of peripubertal androgenized models. In contrast, hypothalamus is most affected in PNA and maternal obesity models suggesting fetal programming effects. The Ms4a6e gene, membrane-spanning 4-domains subfamily A member 6E, a DEG identified in the adipose tissue in all mouse models is also differently expressed in adipose tissue of women with PCOS, highlighting a conserved disease function. Our comprehensive transcriptomic profiling of key target tissues involved in PCOS pathology highlights the effects of developmental windows for androgen exposure and maternal obesity, and provides unique resource to investigate molecular mechanisms underlying PCOS pathogenesis.
Collapse
Affiliation(s)
- Yu Pei
- grid.4714.60000 0004 1937 0626Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Center for molecular medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Sanjiv Risal
- grid.4714.60000 0004 1937 0626Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Hong Jiang
- grid.4714.60000 0004 1937 0626Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Haojiang Lu
- grid.4714.60000 0004 1937 0626Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Eva Lindgren
- grid.4714.60000 0004 1937 0626Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Elisabet Stener-Victorin
- grid.4714.60000 0004 1937 0626Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Qiaolin Deng
- grid.4714.60000 0004 1937 0626Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Center for molecular medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
108
|
Piltonen T, Morin-Papunen L, Ollila MM, Tapanainen J, Arffman R, Järvelin MR, Franks S. Women self-reporting PCOS symptoms should not be overlooked. Hum Reprod 2023; 38:189-190. [PMID: 36433765 DOI: 10.1093/humrep/deac251] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Affiliation(s)
- Terhi Piltonen
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, PEDEGO Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Laure Morin-Papunen
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, PEDEGO Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Meri-Maija Ollila
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, PEDEGO Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Juha Tapanainen
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, PEDEGO Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland.,Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Riikka Arffman
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, PEDEGO Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Marjo-Riitta Järvelin
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland.,Unit of Primary Care, Oulu University Hospital, Oulu, Finland.,MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.,Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| | - Stephen Franks
- Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
| |
Collapse
|
109
|
Abruzzese GA, Velazquez ME, Cerrone GE, Motta AB. Polycystic ovary syndrome in Latin American populations: What is known and what remains unresolved. J Steroid Biochem Mol Biol 2023; 225:106195. [PMID: 36183993 DOI: 10.1016/j.jsbmb.2022.106195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 02/01/2023]
Abstract
Polycystic ovary syndrome (PCOS) is one of the main endocrine and reproductive disorders affecting women in their reproductive age. The syndrome is considered a multifactorial pathology. Therefore, genetic susceptibility and environmental factors contribute to PCOS development and phenotypic manifestation. Ethnicity and socioeconomic factors influence the development of PCOS and could affect the possibility of its diagnosis. Latin America is a unique case of study because of the heterogeneity within the region, complex socioeconomic status, and the mixed ancestry found in these populations. Up-to-date, most studies have focused on developed countries' populations, and there is a lack of evidence regarding Latin-American countries. We propose to review the state of the art of PCOS knowledge regarding Latin American populations, including the metabolic and reproductive aspects of the syndrome and the different influencing factors, and suggest future directions to deepen the study of PCOS.
Collapse
Affiliation(s)
- Giselle Adriana Abruzzese
- Laboratorio de Fisio-Patología Ovárica, Centro de Estudios Farmacológicos y Botánicos, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Mariela Edith Velazquez
- Laboratorio de Fisio-Patología Ovárica, Centro de Estudios Farmacológicos y Botánicos, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gloria Edith Cerrone
- Universidad de Buenos Aires, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Cátedra de Genética, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo (INIGEM), Laboratorio de Diabetes y Metabolismo, Buenos Aires, Argentina
| | - Alicia Beatriz Motta
- Laboratorio de Fisio-Patología Ovárica, Centro de Estudios Farmacológicos y Botánicos, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
110
|
Tamaoka S, Saito K, Yoshida T, Nakabayashi K, Tatsumi K, Kawamura T, Matsuzaki T, Matsubara K, Ogata‐Kawata H, Hata K, Kato‐Fukui Y, Fukami M. Exome-based genome-wide screening of rare variants associated with the risk of polycystic ovary syndrome. Reprod Med Biol 2023; 22:e12504. [PMID: 36845002 PMCID: PMC9947624 DOI: 10.1002/rmb2.12504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/28/2022] [Accepted: 01/12/2023] [Indexed: 02/25/2023] Open
Abstract
Purpose Genetic factors associated with the risk of polycystic ovary syndrome (PCOS) remain largely unknown. Here, we conducted an optimal sequence kernel association test (SKAT-O), an exome-based rare variant association study, to clarify whether rare variants in specific genes contribute to the development of PCOS. Methods SKAT-O was performed using exome data of 44 Japanese patients with PCOS and 301 control women. We analyzed frequencies of rare probably damaging variants in the genome. Results Rare variants of GSTO2 were more commonly identified in the patient group than in the control group (6/44 vs. 1/301; Bonferroni-corrected p-value, 0.028), while the frequencies of variants in other genes were comparable between the two groups. The identified GSTO2 variants were predicted to affect the function, structure, stability, hydrophobicity, and/or the formation of intrinsically disordered regions of the protein. GSTO2 encodes a glutathione transferase that mediates the oxidative stress response and arsenic metabolism. Previously, common variants in GSTO2 and its paralog GSTO1 were associated with the risk of PCOS. Conclusions The results indicate that there are no genes whose rare variants account for a large fraction of the etiology of PCOS, although rare damaging variants in GSTO2 may constitute a risk factor in some cases.
Collapse
Affiliation(s)
- Satoshi Tamaoka
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Kazuki Saito
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
- Department of Perinatal and Maternal Medicine (Ibaraki), Graduate SchoolTokyo Medical and Dental UniversityTokyoJapan
| | - Tomoko Yoshida
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Kazuhiko Nakabayashi
- Department of Maternal‐Fetal BiologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| | | | | | - Toshiya Matsuzaki
- Department of Obstetrics and GynecologyYoshinogawa Medical CenterTokushimaJapan
| | - Keiko Matsubara
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Hiroko Ogata‐Kawata
- Department of Maternal‐Fetal BiologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Kenichiro Hata
- Department of Maternal‐Fetal BiologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Yuko Kato‐Fukui
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| | - Maki Fukami
- Department of Molecular EndocrinologyNational Research Institute for Child Health and DevelopmentTokyoJapan
| |
Collapse
|
111
|
Pruett JE, Romero DG, Yanes Cardozo LL. Obesity-associated cardiometabolic complications in polycystic ovary syndrome: The potential role of sodium-glucose cotransporter-2 inhibitors. Front Endocrinol (Lausanne) 2023; 14:951099. [PMID: 36875461 PMCID: PMC9974663 DOI: 10.3389/fendo.2023.951099] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 01/26/2023] [Indexed: 02/17/2023] Open
Abstract
Polycystic Ovary Syndrome (PCOS) is the most common endocrine disorder in reproductive-age women. PCOS is characterized by androgen excess, oligo/anovulation, and polycystic appearance of the ovaries. Women with PCOS have an increased prevalence of multiple cardiovascular risk factors such as insulin resistance, hypertension, renal injury, and obesity. Unfortunately, there is a lack of effective, evidence-based pharmacotherapeutics to target these cardiometabolic complications. Sodium-glucose cotransporter-2 (SGLT2) inhibitors provide cardiovascular protection in patients with and without type 2 diabetes mellitus. Although the exact mechanisms of how SGLT2 inhibitors confer cardiovascular protection remains unclear, numerous mechanistic hypotheses for this protection include modulation of the renin-angiotensin system and/or the sympathetic nervous system and improvement in mitochondrial function. Data from recent clinical trials and basic research show a potential role for SGLT2 inhibitors in treating obesity-associated cardiometabolic complications in PCOS. This narrative review discusses the mechanisms of the beneficial effect of SGLT2 inhibitors in cardiometabolic diseases in PCOS.
Collapse
Affiliation(s)
- Jacob E. Pruett
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Damian G. Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, United States
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS, United States
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS, United States
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS, United States
| | - Licy L. Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, United States
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS, United States
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS, United States
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, MS, United States
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
- *Correspondence: Licy L. Yanes Cardozo,
| |
Collapse
|
112
|
Fujii S, Oguchi T. Shapes of the uterine cavity are different in women with polycystic ovary syndrome. Reprod Med Biol 2023; 22:e12508. [PMID: 36845000 PMCID: PMC9949362 DOI: 10.1002/rmb2.12508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/25/2023] Open
Abstract
Purpose A cross-sectional study was conducted to evaluate differences in uterine morphology between women with or without polycystic ovary syndrome. Methods The authors recruited 333 infertile reproductive-age women including 93 with polycystic ovary syndrome diagnosed using the criteria of the Japanese Society of Obstetrics Gynecology-2007. Shapes of uterine cavity were measured by transvaginal three-dimensional ultrasound. Results The polycystic ovary syndrome group had a significantly deeper indentation (2.2 ± 0.4 mm vs. 0.0 ± 0.2 mm, p < 0.0001) and a significantly more acute indentation angle (162.9 ± 2.2 deg vs. 175.2 ± 1.3 deg, p < 0.0001) than the control group. Conclusion The depth and the apical angle of fundal indentation of uterine cavity are different in women with polycystic ovary syndrome.
Collapse
|
113
|
Zhang N, Liao Y, Zhao H, Chen T, Jia F, Yu Y, Zhu S, Wang C, Zhang W, Liu X. Polycystic ovary syndrome and 25-hydroxyvitamin D: A bidirectional two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2023; 14:1110341. [PMID: 36967791 PMCID: PMC10034407 DOI: 10.3389/fendo.2023.1110341] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/07/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Accumulating observational studies have indicated that vitamin D deficiency (serum 25-hydroxyvitamin D (25OHD) < 50 nmol/L) is common in women with polycystic ovary syndrome (PCOS). However, the direction and causal nature remain unclear. In this study, we aimed to investigate the causal association between PCOS and 25OHD. METHODS A bidirectional two-sample Mendelian randomization (MR) study was used to evaluate the causal association between PCOS and 25OHD. From the publicly available European-lineage genome-wide association studies (GWAS) summary statistics for PCOS (4,890 cases of PCOS and 20,405 controls) and 25OHD (n = 417,580), we selected 11 and 102 single nucleotide polymorphisms (SNPs) as instrumental variables (IVs), respectively. In univariate MR (uvMR) analysis, inverse-variance weighted (IVW) method was employed in the primary MR analysis and multiple sensitivity analyses were implemented. Additionally, a multivariable MR (mvMR) design was carried to adjust for obesity and insulin resistance (IR) as well. RESULTS UvMR demonstrated that genetically determined PCOS was negatively associated with 25OHD level (IVW Beta: -0.02, P = 0.008). However, mvMR found the causal effect disappeared when adjusting the influence of obesity and IR. Both uvMR and mvMR analysis didn't support the causal effect of 25OHD deficiency on risk of PCOS (IVW OR: 0.86, 95% CI: 0.66 ~ 1.12, P = 0.280). CONCLUSION Our findings highlighted that the casual effect of PCOS on 25OHD deficiency might be mediated by obesity and IR, and failed to find substantial causal effect of 25OHD deficiency on risk of PCOS. Further observational studies and clinical trials are necessary.
Collapse
Affiliation(s)
- Nana Zhang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yan Liao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongyu Zhao
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tong Chen
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fan Jia
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Yu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shiqin Zhu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chaoying Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wufan Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinmin Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Xinmin Liu,
| |
Collapse
|
114
|
Translational Bioinformatics for Human Reproductive Biology Research: Examples, Opportunities and Challenges for a Future Reproductive Medicine. Int J Mol Sci 2022; 24:ijms24010004. [PMID: 36613446 PMCID: PMC9819745 DOI: 10.3390/ijms24010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/16/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Since 1978, with the first IVF (in vitro fertilization) baby birth in Manchester (England), more than eight million IVF babies have been born throughout the world, and many new techniques and discoveries have emerged in reproductive medicine. To summarize the modern technology and progress in reproductive medicine, all scientific papers related to reproductive medicine, especially papers related to reproductive translational medicine, were fully searched, manually curated and reviewed. Results indicated whether male reproductive medicine or female reproductive medicine all have made significant progress, and their markers have experienced the progress from karyotype analysis to single-cell omics. However, due to the lack of comprehensive databases, especially databases collecting risk exposures, disease markers and models, prevention drugs and effective treatment methods, the application of the latest precision medicine technologies and methods in reproductive medicine is limited.
Collapse
|
115
|
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: 87] [Impact Index Per Article: 43.5] [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
|
116
|
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: 13] [Impact Index Per Article: 6.5] [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
|
117
|
Liu M, Hummitzsch K, Bastian NA, Hartanti MD, Irving-Rodgers HF, Anderson RA, Rodgers RJ. Expression of PCOS candidate genes in bovine fetal and adult ovarian somatic cells. REPRODUCTION AND FERTILITY 2022; 3:RAF-22-0068. [PMID: 36346793 PMCID: PMC9782414 DOI: 10.1530/raf-22-0068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 11/08/2022] [Indexed: 11/09/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine metabolic disorder that appears to have a genetic predisposition and a fetal origin. The fetal ovary has two major somatic cell types shown previously to be of different cellular origins, different morphologies and to differentially express 15 genes. We isolated the somatic gonadal ridge epithelial-like (GREL) cells (n = 7) and ovarian fetal fibroblasts (n = 6) by clonal expansion. Using qRT-PCR, we compared the gene expression levels of PCOS candidate genes with previous data on the expression levels in whole fetal ovaries across gestation. We also compared these levels with those in bovine adult ovarian cells including fibroblasts (n = 4), granulosa cells (n = 5) and surface epithelial cells (n = 5). Adult cell types exhibited clear differences in the expression of most genes. In fetal ovarian cells, DENND1A and ERBB3 had significantly higher expression in GREL cells. HMGA2 and TGFB1I1 tended to have higher expression in fetal fibroblasts than GREL cells. Another 19 genes did not exhibit differences between GREL cells and fetal fibroblasts and FBN3, FSHB, LHCGR, FSHR and ZBTB16 were very lowly expressed in GREL cells and fibroblasts. The culture of fetal fibroblasts in EGF-containing medium resulted in lower expression of NEIL2, but higher expression of MAPRE1 compared to culture in the absence of EGF. Thus, the two fetal ovarian somatic cell types mostly lacked differential expression of PCOS candidate genes.
Collapse
Affiliation(s)
- Menghe Liu
- School of Biomedicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Katja Hummitzsch
- School of Biomedicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Nicole A Bastian
- School of Biomedicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Monica D Hartanti
- School of Biomedicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Faculty of Medicine, Universitas Trisakti, Jakarta, Indonesia
- National Research and Innovation Agency, Jakarta, Indonesia
| | - Helen F Irving-Rodgers
- School of Biomedicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- School of Medical Science, Griffith University, Gold Coast Campus, QLD, Australia
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Raymond J Rodgers
- School of Biomedicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
118
|
Leone M, Kuja-Halkola R, Leval A, Butwicka A, Skov J, Zhang R, Liu S, Larsson H, Bergen SE. Genetic and Environmental Contribution to the Co-Occurrence of Endocrine-Metabolic Disorders and Depression: A Nationwide Swedish Study of Siblings. Am J Psychiatry 2022; 179:824-832. [PMID: 36128682 DOI: 10.1176/appi.ajp.21090954] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Depression is common in individuals with endocrine-metabolic disorders and vice versa, and a better understanding of the underlying factors contributing to the comorbidity of these disorders is needed. This study investigated the familial coaggregation of depression and endocrine-metabolic disorders and estimated the contribution of genetic and environmental factors to their co-occurrence. METHODS This population-based cohort study included 2.2 million individuals born in Sweden between 1973 and 1996, with follow-up through 2013. Participants were linked to their biological parents, allowing identification of full siblings, maternal half siblings, and paternal half siblings. Diagnoses of depression and endocrine-metabolic conditions were investigated, with the latter grouped into autoimmune disorders (autoimmune hypothyroidism, Graves' disease, and type 1 diabetes) and non-autoimmune disorders (type 2 diabetes, obesity, and polycystic ovary syndrome). Logistic regression and Cox regression were used to estimate the associations between endocrine-metabolic disorders and depression within the same individual and across siblings. Quantitative genetic modeling was performed to investigate the relative contribution of genetic and environmental influences. RESULTS Individuals with endocrine-metabolic disorders had a significantly higher risk of depression, with odds ratios ranging from 1.43 (95% CI=1.30, 1.57) for Graves' disease to 3.48 (95% CI=3.25, 3.72) for type 2 diabetes. Increased risks extended to full and half siblings. These correlations were mainly explained by shared genetic influences for non-autoimmune conditions, and by nonshared environmental factors for autoimmune disorders, especially for type 1 diabetes. CONCLUSIONS These findings provide phenotypic and etiological insights into the co-occurrence of depression and various endocrine-metabolic conditions, which could guide future research aiming at identifying pathophysiological mechanisms and intervention targets.
Collapse
Affiliation(s)
- Marica Leone
- Janssen Pharmaceutical Companies of Johnson & Johnson, Solna, Sweden (Leone, Leval); Department of Medical Epidemiology and Biostatistics (Leone, Kuja-Halkola, Leval, Butwicka, Zhang, Liu, Larsson, Bergen) and Department of Molecular Medicine and Surgery (Skov), Karolinska Institutet, Solna, Sweden; Child and Adolescent Psychiatry Stockholm, Stockholm Health Care Services, Region Stockholm, Sweden (Butwicka); Department of Child Psychiatry, Medical University of Warsaw, Warsaw (Butwicka); Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland (Butwicka); Department of Medicine, Karlstad Central Hospital, Karlstad, Sweden (Skov); School of Medical Sciences, Örebro University, Örebro, Sweden (Larsson)
| | - Ralf Kuja-Halkola
- Janssen Pharmaceutical Companies of Johnson & Johnson, Solna, Sweden (Leone, Leval); Department of Medical Epidemiology and Biostatistics (Leone, Kuja-Halkola, Leval, Butwicka, Zhang, Liu, Larsson, Bergen) and Department of Molecular Medicine and Surgery (Skov), Karolinska Institutet, Solna, Sweden; Child and Adolescent Psychiatry Stockholm, Stockholm Health Care Services, Region Stockholm, Sweden (Butwicka); Department of Child Psychiatry, Medical University of Warsaw, Warsaw (Butwicka); Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland (Butwicka); Department of Medicine, Karlstad Central Hospital, Karlstad, Sweden (Skov); School of Medical Sciences, Örebro University, Örebro, Sweden (Larsson)
| | - Amy Leval
- Janssen Pharmaceutical Companies of Johnson & Johnson, Solna, Sweden (Leone, Leval); Department of Medical Epidemiology and Biostatistics (Leone, Kuja-Halkola, Leval, Butwicka, Zhang, Liu, Larsson, Bergen) and Department of Molecular Medicine and Surgery (Skov), Karolinska Institutet, Solna, Sweden; Child and Adolescent Psychiatry Stockholm, Stockholm Health Care Services, Region Stockholm, Sweden (Butwicka); Department of Child Psychiatry, Medical University of Warsaw, Warsaw (Butwicka); Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland (Butwicka); Department of Medicine, Karlstad Central Hospital, Karlstad, Sweden (Skov); School of Medical Sciences, Örebro University, Örebro, Sweden (Larsson)
| | - Agnieszka Butwicka
- Janssen Pharmaceutical Companies of Johnson & Johnson, Solna, Sweden (Leone, Leval); Department of Medical Epidemiology and Biostatistics (Leone, Kuja-Halkola, Leval, Butwicka, Zhang, Liu, Larsson, Bergen) and Department of Molecular Medicine and Surgery (Skov), Karolinska Institutet, Solna, Sweden; Child and Adolescent Psychiatry Stockholm, Stockholm Health Care Services, Region Stockholm, Sweden (Butwicka); Department of Child Psychiatry, Medical University of Warsaw, Warsaw (Butwicka); Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland (Butwicka); Department of Medicine, Karlstad Central Hospital, Karlstad, Sweden (Skov); School of Medical Sciences, Örebro University, Örebro, Sweden (Larsson)
| | - Jakob Skov
- Janssen Pharmaceutical Companies of Johnson & Johnson, Solna, Sweden (Leone, Leval); Department of Medical Epidemiology and Biostatistics (Leone, Kuja-Halkola, Leval, Butwicka, Zhang, Liu, Larsson, Bergen) and Department of Molecular Medicine and Surgery (Skov), Karolinska Institutet, Solna, Sweden; Child and Adolescent Psychiatry Stockholm, Stockholm Health Care Services, Region Stockholm, Sweden (Butwicka); Department of Child Psychiatry, Medical University of Warsaw, Warsaw (Butwicka); Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland (Butwicka); Department of Medicine, Karlstad Central Hospital, Karlstad, Sweden (Skov); School of Medical Sciences, Örebro University, Örebro, Sweden (Larsson)
| | - Ruyue Zhang
- Janssen Pharmaceutical Companies of Johnson & Johnson, Solna, Sweden (Leone, Leval); Department of Medical Epidemiology and Biostatistics (Leone, Kuja-Halkola, Leval, Butwicka, Zhang, Liu, Larsson, Bergen) and Department of Molecular Medicine and Surgery (Skov), Karolinska Institutet, Solna, Sweden; Child and Adolescent Psychiatry Stockholm, Stockholm Health Care Services, Region Stockholm, Sweden (Butwicka); Department of Child Psychiatry, Medical University of Warsaw, Warsaw (Butwicka); Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland (Butwicka); Department of Medicine, Karlstad Central Hospital, Karlstad, Sweden (Skov); School of Medical Sciences, Örebro University, Örebro, Sweden (Larsson)
| | - Shengxin Liu
- Janssen Pharmaceutical Companies of Johnson & Johnson, Solna, Sweden (Leone, Leval); Department of Medical Epidemiology and Biostatistics (Leone, Kuja-Halkola, Leval, Butwicka, Zhang, Liu, Larsson, Bergen) and Department of Molecular Medicine and Surgery (Skov), Karolinska Institutet, Solna, Sweden; Child and Adolescent Psychiatry Stockholm, Stockholm Health Care Services, Region Stockholm, Sweden (Butwicka); Department of Child Psychiatry, Medical University of Warsaw, Warsaw (Butwicka); Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland (Butwicka); Department of Medicine, Karlstad Central Hospital, Karlstad, Sweden (Skov); School of Medical Sciences, Örebro University, Örebro, Sweden (Larsson)
| | - Henrik Larsson
- Janssen Pharmaceutical Companies of Johnson & Johnson, Solna, Sweden (Leone, Leval); Department of Medical Epidemiology and Biostatistics (Leone, Kuja-Halkola, Leval, Butwicka, Zhang, Liu, Larsson, Bergen) and Department of Molecular Medicine and Surgery (Skov), Karolinska Institutet, Solna, Sweden; Child and Adolescent Psychiatry Stockholm, Stockholm Health Care Services, Region Stockholm, Sweden (Butwicka); Department of Child Psychiatry, Medical University of Warsaw, Warsaw (Butwicka); Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland (Butwicka); Department of Medicine, Karlstad Central Hospital, Karlstad, Sweden (Skov); School of Medical Sciences, Örebro University, Örebro, Sweden (Larsson)
| | - Sarah E Bergen
- Janssen Pharmaceutical Companies of Johnson & Johnson, Solna, Sweden (Leone, Leval); Department of Medical Epidemiology and Biostatistics (Leone, Kuja-Halkola, Leval, Butwicka, Zhang, Liu, Larsson, Bergen) and Department of Molecular Medicine and Surgery (Skov), Karolinska Institutet, Solna, Sweden; Child and Adolescent Psychiatry Stockholm, Stockholm Health Care Services, Region Stockholm, Sweden (Butwicka); Department of Child Psychiatry, Medical University of Warsaw, Warsaw (Butwicka); Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland (Butwicka); Department of Medicine, Karlstad Central Hospital, Karlstad, Sweden (Skov); School of Medical Sciences, Örebro University, Örebro, Sweden (Larsson)
| |
Collapse
|
119
|
Stener-Victorin E. Update on Animal Models of Polycystic Ovary Syndrome. Endocrinology 2022; 163:bqac164. [PMID: 36201611 PMCID: PMC9631972 DOI: 10.1210/endocr/bqac164] [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: 07/02/2022] [Indexed: 11/19/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a complex disease affecting up to 15% of women of reproductive age. Women with PCOS suffer from reproductive dysfunctions with excessive androgen secretion and irregular ovulation, leading to reduced fertility and pregnancy complications. The syndrome is associated with a wide range of comorbidities including type 2 diabetes, obesity, and psychiatric disorders. Despite the high prevalence of PCOS, its etiology remains unclear. To understand the pathophysiology of PCOS, how it is inherited, and how to predict PCOS, and prevent and treat women with the syndrome, animal models provide an important approach to answering these fundamental questions. This minireview summarizes recent investigative efforts on PCOS-like rodent models aiming to define underlying mechanisms of the disease and provide guidance in model selection. The focus is on new genetic rodent models, on a naturally occurring rodent model, and provides an update on prenatal and peripubertal exposure models.
Collapse
|
120
|
Sharma A, Krick B, Li Y, Summers SA, Playdon MC, Welt C. The Use of Ceramides to Predict Metabolic Response to Metformin in Women With PCOS. J Endocr Soc 2022; 6:bvac131. [PMID: 36249411 PMCID: PMC9557973 DOI: 10.1210/jendso/bvac131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Indexed: 01/26/2023] Open
Abstract
Context Polycystic ovarian syndrome (PCOS) is a complex disorder in which metabolic abnormalities are associated with reproductive dysfunction. Women with PCOS have increased ceramide concentrations. Previous studies demonstrated that treating metabolic abnormalities of PCOS with metformin improved glucose effectiveness after 12 weeks. Objective We evaluated whether, in women with PCOS, lower baseline ceramide, diacylglycerol (DAG), and triacylglycerol (TAG) concentrations were associated with improved metabolic response to metformin. Methods Women (n = 29), aged 29 ± 5 years and diagnosed with PCOS by the NIH criteria underwent an intravenous glucose tolerance test (IVGTT) before and after 12-week treatment with metformin (1500 mg per day). Metabolic responders were defined by improved glucose effectiveness, specifically, the ability of glucose to stimulate uptake and suppress production, after metformin treatment. Results Twelve weeks of metformin resulted in weight loss (-1.7 ± 2.6 kg, P < 0.01) and a reduction in BMI (-0.6 ± 0.9 kg/m2, P < 0.01) with no change in HbA1c. The concentrations of Cer(d18:1/22:0), Cer(d18:1/24:0), total ceramides, total Cer(d16:0), total Cer(d18:2), DAG, dihydrosphingomyelin (DHSM), and TAG decreased after metformin treatment (P < 0.05). Baseline total Cer(d16:0) concentration <204.1 pmol/mL was 82% sensitive (AUC 0.72, P = 0.03) and total DHSM concentration <32237 pmol/mL was 100% specific (AUC 0.73, P = 0.03) in predicting improved metabolic response to metformin, as measured by IVGTT. Conclusion Lower total Cer(16:0) and DHSM concentrations are associated with a beneficial metabolic response to metformin in women with PCOS. Based on the known association between higher ceramide levels and type 2 diabetes, the data suggest that metformin improves metabolic parameters in women with mild metabolic derangements.
Collapse
Affiliation(s)
- Anu Sharma
- Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Benjamin Krick
- Cancer Control and Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Ying Li
- Department of Nutrition & Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Scott A Summers
- Department of Nutrition & Integrative Physiology, University of Utah, Salt Lake City, UT, USA.,Diabetes and Metabolism Research Center, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Mary C Playdon
- Cancer Control and Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT, USA.,Department of Nutrition & Integrative Physiology, University of Utah, Salt Lake City, UT, USA.,Diabetes and Metabolism Research Center, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Corrine Welt
- Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
121
|
Dalibalta S, Abukhaled Y, Samara F. Factors influencing the prevalence of polycystic ovary syndrome (PCOS) in the United Arab Emirates. REVIEWS ON ENVIRONMENTAL HEALTH 2022; 37:311-319. [PMID: 35538690 DOI: 10.1515/reveh-2021-0036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
Statistics indicate that at least 20-25% of women suffer from PCOS in the Gulf region. Despite its prevalence and negative implications on reproductive, metabolic, and physiological heath the exact cause of PCOS is unknown, in part due to the diversity of symptoms manifested by this disorder. In this review, we investigate causes of PCOS globally and draw on these studies, to determine the potential contributing factors for PCOS pathogenesis in the UAE population. The most frequently identified factors promoting PCOS pathogenesis that may be pertinent to this population include physiological factors such as insulin resistance, vitamin D deficiency, genetic factors, obesity, and anti-mullerian hormone (AMH) levels in the body as well as environmental factors such as air pollution, endocrine disrupting chemicals, and pesticide use. This evidence will help inform healthcare workers and government agencies to set up optimal guidelines for control and awareness of PCOS in the UAE.
Collapse
Affiliation(s)
- Sarah Dalibalta
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah, United Arab Emirates
| | - Yara Abukhaled
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah, United Arab Emirates
| | - Fatin Samara
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
122
|
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is a common condition characterized by reproductive, hyperandrogenic and dysmetabolic features, and often becomes clinically manifest during adolescence, particularly with weight-gain. SOURCES OF DATA Pubmed search. AREAS OF AGREEMENT PCOS is heritable and closely associates with obesity (based on data from both epidemiological and genetic studies). Furthermore, insulin resistance forms a central cornerstone of the pathogenesis of PCOS and mediates a close association between obesity and the severity of the phenotypic features of PCOS. AREAS OF CONTROVERSY Our understanding of the pathogenesis of PCOS remains incomplete, especially regarding its missing heritability (with only a small fraction having been identified from the genome-wide association studies reported to date), and its developmental origins. GROWING POINTS A challenge for the future is to explore a role for epigenetic modifications in the development of PCOS, and implications for the in utero environment and novel therapeutic opportunities.
Collapse
Affiliation(s)
- T M Barber
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Clinical Sciences Research Laboratories, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
| |
Collapse
|
123
|
Liu Q, Tang B, Zhu Z, Kraft P, Deng Q, Stener-Victorin E, Jiang X. A genome-wide cross-trait analysis identifies shared loci and causal relationships of type 2 diabetes and glycaemic traits with polycystic ovary syndrome. Diabetologia 2022; 65:1483-1494. [PMID: 35771237 PMCID: PMC9345824 DOI: 10.1007/s00125-022-05746-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/06/2022] [Indexed: 12/20/2022]
Abstract
AIMS/HYPOTHESIS The link underlying abnormal glucose metabolism, type 2 diabetes and polycystic ovary syndrome (PCOS) that is independent of BMI remains unclear in observational studies. We aimed to clarify this association using a genome-wide cross-trait approach. METHODS Summary statistics from the hitherto largest genome-wide association studies conducted for type 2 diabetes, type 2 diabetes mellitus adjusted for BMI (T2DMadjBMI), fasting glucose, fasting insulin, 2h glucose after an oral glucose challenge (all adjusted for BMI), HbA1c and PCOS, all in populations of European ancestry, were used. We quantified overall and local genetic correlations, identified pleiotropic loci and expression-trait associations, and made causal inferences across traits. RESULTS A positive overall genetic correlation between type 2 diabetes and PCOS was observed, largely influenced by BMI (rg=0.31, p=1.63×10-8) but also independent of BMI (T2DMadjBMI-PCOS: rg=0.12, p=0.03). Sixteen pleiotropic loci affecting type 2 diabetes, glycaemic traits and PCOS were identified, suggesting mechanisms of association that are independent of BMI. Two shared expression-trait associations were found for type 2 diabetes/T2DMadjBMI and PCOS targeting tissues of the cardiovascular, exocrine/endocrine and digestive systems. A putative causal effect of fasting insulin adjusted for BMI and type 2 diabetes on PCOS was demonstrated. CONCLUSIONS/INTERPRETATION We found a genetic link underlying type 2 diabetes, glycaemic traits and PCOS, driven by both biological pleiotropy and causal mediation, some of which is independent of BMI. Our findings highlight the importance of controlling fasting insulin levels to mitigate the risk of PCOS, as well as screening for and long-term monitoring of type 2 diabetes in all women with PCOS, irrespective of BMI.
Collapse
Affiliation(s)
- Qianwen Liu
- Department of Clinical Neuroscience, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Bowen Tang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Zhaozhong Zhu
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter Kraft
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Stockholm, Sweden
| | | | - Xia Jiang
- Department of Clinical Neuroscience, Karolinska Institutet, Solna, Stockholm, Sweden.
| |
Collapse
|
124
|
Li X, Xiao H, Ma Y, Zhou Z, Chen D. Identifying novel genetic loci associated with polycystic ovary syndrome based on its shared genetic architecture with type 2 diabetes. Front Genet 2022; 13:905716. [PMID: 36105080 PMCID: PMC9464923 DOI: 10.3389/fgene.2022.905716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified several common variants associated with polycystic ovary syndrome (PCOS). However, the etiology behind PCOS remains incomplete. Available evidence suggests a potential genetic correlation between PCOS and type 2 diabetes (T2D). The publicly available data may provide an opportunity to enhance the understanding of the PCOS etiology. Here, we quantified the polygenic overlap between PCOS and T2D using summary statistics of PCOS and T2D and then identified the novel genetic variants associated with PCOS behind this phenotypic association. A bivariate causal mixture model (MiXeR model) found a moderate genetic overlap between PCOS and T2D (Dice coefficient = 44.1% and after adjusting for body mass index, 32.1%). The conditional/conjunctional false discovery rate method identified 11 potential risk variants of PCOS conditional on associations with T2D, 9 of which were novel and 6 of which were jointly associated with two phenotypes. The functional annotation of these genetic variants supports a significant role for genes involved in lipid metabolism, immune response, and the insulin signaling pathway. An expression quantitative trait locus functionality analysis successfully repeated that 5 loci were significantly associated with the expression of candidate genes in many tissues, including the whole blood, subcutaneous adipose, adrenal gland, and cerebellum. We found that SCN2A gene is co-localized with PCOS in subcutaneous adipose using GWAS-eQTL co-localization analyses. A total of 11 candidate genes were differentially expressed in multiple tissues of the PCOS samples. These findings provide a new understanding of the shared genetic architecture between PCOS and T2D and the underlying molecular genetic mechanism of PCOS.
Collapse
|
125
|
He R, Liu R, Wu H, Yu J, Jiang Z, Huang H. The Causal Evidence of Birth Weight and Female-Related Traits and Diseases: A Two-Sample Mendelian Randomization Analysis. Front Genet 2022; 13:850892. [PMID: 36035116 PMCID: PMC9412024 DOI: 10.3389/fgene.2022.850892] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives: A large meta-analysis indicated a more pronounced association between lower birth weight (BW) and diseases in women but less concern about the causality between BW and female-related phenotypes and diseases. Methods: Mendelian randomization (MR) analysis was used to estimate the causal relationship between two traits or diseases using summary datasets from genome-wide association studies. Exposure instrumental variables are variants that are strongly associated with traits and are tested using four different statistical methods, including the inverse variance weighting, MR-Egger, weighted median, and weighted mode in MR analysis. Next, sensitivity analysis and horizontal pleiotropy were assessed using leave-one-out and MR-PRESSO packages. Results: The body mass index (BMI) in adulthood was determined by BW (corrected β = 0.071, p = 3.19E-03). Lower BW could decrease the adult sex hormone-binding globulin (SHBG) level (β = −0.081, p = 2.08E-06), but it resulted in increased levels of bioavailable testosterone (bio-T) (β = 0.105, p = 1.25E-05). A potential inverse effect was observed between BW and menarche (corrected β = −0.048, p = 4.75E-03), and no causal association was confirmed between BW and the risk of endometriosis, leiomyoma, and polycystic ovary syndrome. Conclusion: Our results suggest that BW may play an important role and demonstrates a significant direct influence on female BMI, SHBG and bio-T levels, and menarche.
Collapse
Affiliation(s)
- Renke He
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Rui Liu
- Department of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haiyan Wu
- Department of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiaen Yu
- Department of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhaoying Jiang
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Hefeng Huang
- Department of Reproductive Endocrinology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, School of Medicine, Zhejiang University, Hangzhou, China
- Shanghai Frontiers Science Center of Reproduction and Development, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China
- *Correspondence: Hefeng Huang,
| |
Collapse
|
126
|
Peña AS, Codner E, Witchel S. Criteria for Diagnosis of Polycystic Ovary Syndrome during Adolescence: Literature Review. Diagnostics (Basel) 2022; 12:diagnostics12081931. [PMID: 36010282 PMCID: PMC9406411 DOI: 10.3390/diagnostics12081931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 11/11/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine conditions in women. PCOS may be more challenging to diagnose during adolescence due to an overlap with the physiological events of puberty, which are part of the diagnostic criteria in adult women. This review focuses on the evidence available in relation to PCOS diagnostic criteria for adolescents. Adolescent PCOS should be diagnosed using two main criteria irregular -menstrual cycles (relative to number of years post-menarche) and hyperandrogenism (clinical and/or biochemical); after excluding other conditions that mimic PCOS. Accurate definitions of the two main criteria will decrease challenges/controversies with the diagnosis and provide timely diagnosis during adolescence to establish early management. Despite the attempts to create accurate diagnostic criteria and definitions, this review highlights the limited research in this area, especially in the follow up of adolescents presenting with one diagnostic feature that are called “at risk of PCOS”. Studies in adolescents continue to use the Rotterdam diagnostic criteria that uses pelvic ultrasound. This is inappropriate, because previous and emerging data that show many healthy adolescents have polycystic ovarian morphology in the early years post-menarche. In the future, anti-Müllerian hormone levels might help support PCOS diagnosis if adolescents meet two main criteria.
Collapse
Affiliation(s)
- Alexia S. Peña
- Discipline of Paediatrics, The University of Adelaide Robinson Research Institute, 72 King William Road, Adelaide, SA 5006, Australia
- Endocrinology and Diabetes Department, Women’s and Children’s Hospital, 72 King William Road, Adelaide, SA 5006, Australia
- Correspondence: ; Tel.: +61-881618134
| | - Ethel Codner
- Institute of Child and Maternal Research, School of Medicine, University of Chile, Santiago 836-0160, Chile
| | - Selma Witchel
- UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA
| |
Collapse
|
127
|
Akre S, Sharma K, Chakole S, Wanjari MB. Recent Advances in the Management of Polycystic Ovary Syndrome: A Review Article. Cureus 2022; 14:e27689. [PMID: 36072214 PMCID: PMC9440853 DOI: 10.7759/cureus.27689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/04/2022] [Indexed: 11/25/2022] Open
Abstract
Polycystic ovarian syndrome (PCOS) is an endocrine disorder. This condition is characterized by chronic anovulation and ovarian dysfunction, unlike other ovulation disorders when the ovaries are non-functional or abnormal. Currently, most therapy is centred on the patient's primary complaint. Treatment focuses on reducing hyperandrogenism symptoms, restoring menstrual regularity, and achieving conception. In treating infertility caused by polycystic ovarian syndrome, letrozole (an aromatase inhibitor) appears to be more successful than clomiphene citrate (an anti-estrogen and a reference infertility drug). When provided by a multidisciplinary team, it can help patients maintain appropriate lifestyle changes, such as reducing body fat, increasing metabolism, and enhancing reproductive health. Compound oral contraceptives are the most common kind of androgen inhibitor and are the preferred therapy for menstrual disruption in PCOS patients who do not want to get pregnant. Weight loss should be prioritized for women with PCOS since a healthy, balanced diet combined with regular exercise can boost metabolism, increase insulin sensitivity, and aid weight loss safely. This will improve their physical health. Other than reproductive symptoms, PCOS symptoms include insulin resistance (IR), metabolic syndrome (MS), and chronic low-grade inflammation. Our understanding of the pathophysiological process, diagnosis, and therapy of PCOS has advanced recently.
Collapse
|
128
|
Joham AE, Piltonen T, Lujan ME, Kiconco S, Tay CT. Challenges in diagnosis and understanding of natural history of polycystic ovary syndrome. Clin Endocrinol (Oxf) 2022; 97:165-173. [PMID: 35593530 PMCID: PMC9541175 DOI: 10.1111/cen.14757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 11/30/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrinopathy affecting 8%-13% of reproductive-aged women. The aetiology of the syndrome is complex, with genetic susceptibility, androgen exposure in early life and adiposity related dysfunction leading to perturbance in hypothalamic-ovarian function. PCOS clinical features are heterogeneous, with manifestations arising even in early adolescence, developing into multisystem reproductive, metabolic and psychological manifestations in adulthood. In this review, we will discuss challenges in the diagnosis of PCOS and understanding of the natural history of PCOS.
Collapse
Affiliation(s)
- Anju E. Joham
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
- Department of Diabetes and Vascular MedicineMonash HealthMelbourneVictoriaAustralia
| | - Terhi Piltonen
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center, Oulu University HospitalUniversity of OuluOuluFinland
| | - Marla E. Lujan
- Division of Nutritional Sciences, Colleges of Human Ecology and Agriculture and Life SciencesCornell UniversityIthacaNew YorkUSA
| | - Sylvia Kiconco
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Chau Thien Tay
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
- Department of Diabetes and Vascular MedicineMonash HealthMelbourneVictoriaAustralia
| |
Collapse
|
129
|
Tay CT, Loxton D, Khomami MB, Teede H, Joham AE. Negative associations of ideal family size achievement with hypertension, obesity and maternal age in women with and without polycystic ovary syndrome. Clin Endocrinol (Oxf) 2022; 97:217-226. [PMID: 35394665 DOI: 10.1111/cen.14736] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To investigate lifetime reproductive outcomes and the relationship of ideal family size (IFS) achievement with metabolic, psychiatric and reproductive history in women with and without polycystic ovary syndrome (PCOS). DESIGN Cross-sectional. PATIENT(S) A total of 9034 women with (n = 778) and without self-reported PCOS (n = 8256) born between 1973 and 1978 in the Australian Longitudinal Study on Women's Health. MEASUREMENTS Self-reported IFS achievement and total number of live births. RESULTS Women with and without PCOS aspired for similar IFS. Compared with women without PCOS, significantly less women with PCOS achieved their IFS (53.08% vs. 60.47%, p < 0.001). Higher proportion of women with PCOS did not achieve a live birth (37.15% vs. 31.64%, p = 0.002) and their median total number of live births was also lower (1 vs. 2, p < 0.001) than women without PCOS. After controlling for sociodemographic factors, negative associations were observed between IFS achievement and PCOS status, various metabolic, psychiatric and reproductive history. However, only hypertension (adjusted odds ratio [OR]: 0.82, 95% confidence interval [CI]: 0.67-1.00), obesity (adjusted OR: 0.79, 95% CI: 0.69-0.90), history of in vitro fertilisation use (IVF) (adjusted OR: 0.49, 95% CI: 0.38-0.63) and maternal age at first childbirth (adjusted OR: 0.92, 95% CI: 0.91-0.93) remained inversely associated with achievement of IFS in further multivariable regression models. CONCLUSION Metabolic conditions and reproductive history of maternal age at first childbirth and history of IVF use, but not psychological conditions, were associated with reduced odds of achieving IFS. Early family planning/initiation and optimisation of metabolic health may help to improve reproductive outcomes.
Collapse
Affiliation(s)
- Chau Thien Tay
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
- Department of Diabetes and Vascular Medicine, Monash Health, Melbourne, Victoria, Australia
| | - Deborah Loxton
- Research Centre for Generational Health and Ageing, School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, New South Wales, Australia
| | - Mahnaz Bahri Khomami
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
- Department of Diabetes and Vascular Medicine, Monash Health, Melbourne, Victoria, Australia
| | - Helena Teede
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
- Department of Diabetes and Vascular Medicine, Monash Health, Melbourne, Victoria, Australia
| | - Anju E Joham
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
- Department of Diabetes and Vascular Medicine, Monash Health, Melbourne, Victoria, Australia
| |
Collapse
|
130
|
Allen LA, Shrikrishnapalasuriyar N, Rees DA. Long-term health outcomes in young women with polycystic ovary syndrome: A narrative review. Clin Endocrinol (Oxf) 2022; 97:187-198. [PMID: 34617616 DOI: 10.1111/cen.14609] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 09/22/2021] [Accepted: 10/03/2021] [Indexed: 11/29/2022]
Abstract
Polycystic ovary syndrome (PCOS) has long been recognized as a common disorder in young women leading to reproductive and cutaneous sequelae. However, the associated health risks are now known to extend beyond these familiar manifestations to a range of longer-term comorbidities. Here we review the evidence for an association of PCOS with adverse long-term health outcomes, discussing the pathophysiological mechanisms involved in addition to opportunities for therapeutic intervention. Cross-sectional and longitudinal studies point to an increased risk of type 2 diabetes, hypertension and dyslipidaemia, with recent data confirming that these translate to an increased risk of cardiovascular events independently of obesity. Obstructive sleep apnoea, nonalcoholic fatty liver disease and endometrial cancer are also more prevalent, while mental health disorders, notably anxiety and depression, are common but under-appreciated associations. Uncertainties remain as to whether these risks are apparent in all patients with PCOS or are confined to particular subtypes, whether risks persist post-menopausally and how risk may be affected by ethnicity. Further work is also needed in establishing if systematic screening and targeted intervention can lead to improved outcomes. Until such data are available, clinicians managing women with PCOS should counsel patients on long-term health risks and invest in strategies that limit progression to metabolic and non-metabolic morbidities.
Collapse
Affiliation(s)
- Lowri A Allen
- Diabetes Research Group, School of Medicine, Cardiff University, Cardiff, UK
| | | | - Dafydd Aled Rees
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
131
|
Gillespie NA, Gentry AE, Kirkpatrick RM, Reynolds CA, Mathur R, Kendler KS, Maes HH, Webb BT, Peterson RE. Determining the stability of genome-wide factors in BMI between ages 40 to 69 years. PLoS Genet 2022; 18:e1010303. [PMID: 35951648 PMCID: PMC9398001 DOI: 10.1371/journal.pgen.1010303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 08/23/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
Genome-wide association studies (GWAS) have successfully identified common variants associated with BMI. However, the stability of aggregate genetic variation influencing BMI from midlife and beyond is unknown. By analysing 165,717 men and 193,073 women from the UKBiobank, we performed BMI GWAS on six independent five-year age intervals between 40 and 72 years. We then applied genomic structural equation modeling to test competing hypotheses regarding the stability of genetic effects for BMI. LDSR genetic correlations between BMI assessed between ages 40 to 73 were all very high and ranged 0.89 to 1.00. Genomic structural equation modeling revealed that molecular genetic variance in BMI at each age interval could not be explained by the accumulation of any age-specific genetic influences or autoregressive processes. Instead, a common set of stable genetic influences appears to underpin genome-wide variation in BMI from middle to early old age in men and women alike.
Collapse
Affiliation(s)
- Nathan A. Gillespie
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Amanda Elswick Gentry
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Robert M. Kirkpatrick
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Chandra A. Reynolds
- Department of Psychology, University of California, Riverside, California, United States of America
| | - Ravi Mathur
- GenOmics, Bioinformatics, and Translational Research Center, Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, North Carolina, United States of America
| | - Kenneth S. Kendler
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Hermine H. Maes
- Virginia Institute for Psychiatric and Behavior Genetics, Departments of Human and Molecular Genetics, Psychiatry, & Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Bradley T. Webb
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
- GenOmics, Bioinformatics, and Translational Research Center, Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, North Carolina, United States of America
| | - Roseann E. Peterson
- Virginia Institute for Psychiatric and Behavior Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
132
|
Berga SL. Brain phenotype in PCOS: androgens, anovulation, and gender. Gynecol Endocrinol 2022; 38:615-616. [PMID: 35971943 DOI: 10.1080/09513590.2022.2106475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2022] Open
Affiliation(s)
- Sarah L Berga
- Department of Obstetrics and Gynecology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo SUNY, Buffalo, NY, USA
| |
Collapse
|
133
|
Pei Z, Lu W, Feng Y, Xu C, Hsueh AJW. Out of step societal and Darwinian adaptation during evolution is the cause of multiple women's health issues. Hum Reprod 2022; 37:1959-1969. [PMID: 35881063 DOI: 10.1093/humrep/deac156] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/18/2022] [Indexed: 11/15/2022] Open
Abstract
During human evolution, major changes in our societal conditions and environment took place without sufficient time for concomitant genetic alterations, leading to out of step adaptation and diseases in women. We first discuss recent societal adaptation mismatch (menstrual bleeding; increases in cancers of reproductive organs, endometriosis; mother's nursing; polycystic ovarian syndrome; transgenerational epigenetic modifications), followed by Darwinian out of step adaptation (labor difficulties; sex chromosomes, human diseases and sex disparity in genomic DNA). We discuss the evolutionary basis of menstrual bleeding, followed by recent increases in cancers of reproductive organs and endometriosis. The importance of breastfeeding by mothers is also emphasized. Earlier onset of menarche, decreased rates of childbirths and breastfeeding resulted in increased number of menstrual cycles in a lifetime, coupled with excess estrogen exposure and incessant ovulation, conditions that increased the susceptibility to mammary and uterine cancers as well as ovarian epithelial cancer and endometriosis. Shorter lactation duration in mothers also contributed to more menstrual cycles. We further discuss the evolutionary basis of the prevalent polycystic ovary syndrome. During the long-term Darwinian evolution, difficulties in childbirth evolved due to a narrowed pelvis, our upright walking and enlarged fetal brain sizes. Because there are 1.5% genomic DNA differences between woman and man, it is of significance to investigate sex-specific human physiology and diseases. In conclusion, understanding out of step adaptation during evolution could allow the prevention and better management of female reproductive dysfunction and diseases.
Collapse
Affiliation(s)
- Zhenle Pei
- Department of Genetics, Shanghai Ji Ai Genetics & IVF Institute, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.,Department of Integrative Medicine and Neurobiology, Institute of Integrative Medicine of Fudan University, Institute of Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wenhan Lu
- Department of Integrative Medicine and Neurobiology, Institute of Integrative Medicine of Fudan University, Institute of Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, Institute of Integrative Medicine of Fudan University, Institute of Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Congjian Xu
- Department of Genetics, Shanghai Ji Ai Genetics & IVF Institute, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Aaron J W Hsueh
- Department of Integrative Medicine and Neurobiology, Institute of Integrative Medicine of Fudan University, Institute of Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Division of Reproductive and Stem Cell Biology, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
134
|
Lidaka L, Bekere L, Lazdane G, Lazovska M, Dzivite-Krisane I, Gailite L. Role of Single Nucleotide Variants in the YAP1 Gene in Adolescents with Polycystic Ovary Syndrome. Biomedicines 2022; 10:biomedicines10071688. [PMID: 35884992 PMCID: PMC9312887 DOI: 10.3390/biomedicines10071688] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Polycystic ovary syndrome (PCOS) is one of the most common endocrinopathies in women. It can manifest in adolescence, affecting up to 8% of adolescents. Long-term health consequences characteristic of PCOS are impaired fertility, increased risk of type 2 diabetes, metabolic disorders and cardiovascular disease. All of these sequelae are exacerbated by increased body weight, a major feature of PCOS. The protein encoded by the YAP1 gene plays a key role in one of the pivotal mechanisms that govern cellular/organismal metabolism and contributes to the pathogenesis of metabolic diseases. Aim: To compare the prevalence of single nucleotide variants (SNVs) in the YAP1 gene among adolescents with PCOS, adolescents at risk of PCOS development and healthy adolescents, and assess their association with the clinical characteristics of PCOS. Results: The frequencies of the five investigated YAP1 gene SNVs (rs11225161, rs11225166, rs3858420, rs11225138 and rs79981660) were not significantly different among adolescents with PCOS, risk group patients and healthy controls. Furthermore, none of the SNVs contributed to the clinical characteristics of adolescents with PCOS and adolescents at risk of PCOS development. Conclusions: No significant associations were found between PCOS in adolescents and the five investigated SNVs in the YAP1 gene.
Collapse
Affiliation(s)
- Lasma Lidaka
- Department of Paediatric Gynaecology, Children’s Clinical University Hospital, LV-1004 Riga, Latvia
- Department of Obstetrics and Gynaecology, Riga Stradins University, LV-1007 Riga, Latvia;
- Correspondence:
| | - Laine Bekere
- Faculty of Residency, Riga Stradins University, LV-1007 Riga, Latvia;
| | - Gunta Lazdane
- Department of Obstetrics and Gynaecology, Riga Stradins University, LV-1007 Riga, Latvia;
| | - Marija Lazovska
- Scientific Laboratory of Molecular Genetics, Riga Stradins University, LV-1007 Riga, Latvia; (M.L.); (L.G.)
| | - Iveta Dzivite-Krisane
- Department of Paediatric Endocrinology, Children’s Clinical University Hospital, LV-1004 Riga, Latvia;
| | - Linda Gailite
- Scientific Laboratory of Molecular Genetics, Riga Stradins University, LV-1007 Riga, Latvia; (M.L.); (L.G.)
| |
Collapse
|
135
|
An Ovarian Steroid Metabolomic Pathway Analysis in Basal and Polycystic Ovary Syndrome (PCOS)-like Gonadotropin Conditions Reveals a Hyperandrogenic Phenotype Measured by Mass Spectrometry. Biomedicines 2022; 10:biomedicines10071646. [PMID: 35884951 PMCID: PMC9313004 DOI: 10.3390/biomedicines10071646] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/29/2022] [Accepted: 07/06/2022] [Indexed: 12/26/2022] Open
Abstract
Prior work has demonstrated that murine ovarian explants and isolated ovarian follicles can recapitulate human-like 28-day cycles in vitro with normal patterns of estradiol and progesterone secretion in response to gonadotropin stimulation. The objective of this study was to manipulate the gonadotropin stimulation protocol to mimic polycystic ovary syndrome (PCOS) and assess the resulting changes in ovarian steroidogenesis. A secondary aim of the study was to develop a high-throughput, sensitive, and specific liquid chromatography with tandem mass spectrometry (LC-MS/MS) assay to measure seven steroid hormones (estrone, estradiol, progesterone, testosterone, androstenedione, dehydroepiandrosterone, and dihydrotestosterone) in conditioned culture media. Ovaries were harvested from 12-day-old CD-1 mice and cultured for 28 days, with ovulation induction on culture day 14. Media were supplemented human chorionic gonadotropin (hCG, a luteinizing hormone analog) and follicle stimulating hormone (FSH) at ratios of 1:0 (standard media), 1:1 (physiologic ratio), and 3:1 (PCOS-like ratio). Ovaries cultured in PCOS-like media displayed hyperandrogenism and impaired ovulation, two key features of a PCOS-like phenotype. Taken together, this first-of-its-kind presentation of hormone levels from single tissues creates a map of the enzymatic steps most acutely affected by gonadotropin dysregulation and may provide opportunities for assessing other potential insults in PCOS pathogenesis.
Collapse
|
136
|
Crespo RP, Rocha TP, Montenegro LR, Nishi MY, Jorge AAL, Maciel GAR, Baracat E, Latronico AC, Mendonca BB, Gomes LG. High Throughput Sequencing to Identify Monogenic Etiologies in a Preselected Polycystic Ovary Syndrome Cohort. J Endocr Soc 2022; 6:bvac106. [PMID: 35898701 PMCID: PMC9309801 DOI: 10.1210/jendso/bvac106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Indexed: 11/19/2022] Open
Abstract
Context Polycystic ovary syndrome (PCOS) etiology remains to be elucidated, but familial clustering and twin studies have shown a strong heritable component. Objective The purpose of this study was to identify rare genetic variants that are associated with the etiology of PCOS in a preselected cohort. Methods This prospective study was conducted among a selected group of women with PCOS. The study’s inclusion criteria were patients with PCOS diagnosed by the Rotterdam criteria with the following phenotypes: severe insulin resistance (IR), normoandrogenic–normometabolic phenotype, adrenal hyperandrogenism, primary amenorrhea, and familial PCOS. Forty-five patients were studied by target sequencing, while 8 familial cases were studied by whole exome sequencing. Results Patients were grouped according to the inclusion criteria with the following distribution: 22 (41.5%) with severe IR, 13 (24.5%) with adrenal hyperandrogenism, 7 (13.2%) with normoandrogenic phenotype, 3 (5.7%) with primary amenorrhea, and 8 (15.1%) familial cases. DNA sequencing analysis identified 1 pathogenic variant in LMNA, 3 likely pathogenic variants in INSR, PIK3R1, and DLK1, and 6 variants of uncertain significance level with interesting biologic rationale in 5 genes (LMNA, GATA4, NR5A1, BMP15, and FSHR). LMNA was the most prevalent affected gene in this cohort (3 variants). Conclusion Several rare variants in genes related to IR were identified in women with PCOS. Although IR is a common feature of PCOS, patients with extreme or atypical phenotype should be carefully evaluated to rule out monogenic conditions.
Collapse
Affiliation(s)
- Raiane P Crespo
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia e Metabologia, Faculdade de Medicina da Universidade de São Paulo , São Paulo, Brasil
| | - Thais P Rocha
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia e Metabologia, Faculdade de Medicina da Universidade de São Paulo , São Paulo, Brasil
| | - Luciana R Montenegro
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia e Metabologia, Faculdade de Medicina da Universidade de São Paulo , São Paulo, Brasil
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina da Universidade de São Paulo , São Paulo, Brasil
| | - Mirian Y Nishi
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia e Metabologia, Faculdade de Medicina da Universidade de São Paulo , São Paulo, Brasil
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina da Universidade de São Paulo , São Paulo, Brasil
| | - Alexander A L Jorge
- Unidade de Endocrinologia Genética (LIM 25), Hospital das Clínicas, Disciplina de Endocrinologia, Faculdade de Medicina da Universidade de São Paulo , São Paulo, Brasil
| | - Gustavo A R Maciel
- Disciplina de Ginecologia, Faculdade de Medicina da Universidade de São Paulo , Brasil
| | - Edmund Baracat
- Disciplina de Ginecologia, Faculdade de Medicina da Universidade de São Paulo , Brasil
| | - Ana Claudia Latronico
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia e Metabologia, Faculdade de Medicina da Universidade de São Paulo , São Paulo, Brasil
| | - Berenice B Mendonca
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia e Metabologia, Faculdade de Medicina da Universidade de São Paulo , São Paulo, Brasil
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina da Universidade de São Paulo , São Paulo, Brasil
| | - Larissa G Gomes
- Unidade de Endocrinologia do Desenvolvimento, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas, Disciplina de Endocrinologia e Metabologia, Faculdade de Medicina da Universidade de São Paulo , São Paulo, Brasil
| |
Collapse
|
137
|
Chen Z, Zhang Z, Wang Z, Zhang Z, Wang Q, Pan Y. Heterozygosity and homozygosity regions affect reproductive success and the loss of reproduction: a case study with litter traits in pigs. Comput Struct Biotechnol J 2022; 20:4060-4071. [PMID: 35983229 PMCID: PMC9364102 DOI: 10.1016/j.csbj.2022.07.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 12/23/2022] Open
Abstract
Runs of heterozygosity (ROHet) and homozygosity (ROH) harbor useful information related to traits of interest. There is a lack of investigating the effect of ROHet and ROH on reproductive success and the loss of reproduction in mammals. Here, we detected and characterized the ROHet and ROH patterns in the genomes of Chinese indigenous pigs (i.e., Jinhua, Chun’an, Longyou Black, and Shengxian Spotted pigs), revealing the similar genetic characteristics of indigenous pigs. Later, we highlighted the underlying litter traits-related ROHet and ROH using association analysis with linear model in these four indigenous pig breeds. To pinpoint the promising candidate genes associated with litter traits, we further in-depth explore the selection patterns of other five pig breeds (i.e., Erhualian, Meishan, Minzhu, Rongchang, and Diqing pigs) with different levels of reproduction performance at the underlying litter traits-related ROHet and ROH using FST and genetic diversity ratio. Then, we identified a set of known and novel candidate genes associated with reproductive performance in pigs. For the novel candidate genes (i.e., CCDC91, SASH1, SAMD5, MACF1, MFSD2A, EPC2, and MBD5), we obtained public available datasets and performed multi-omics analyses integrating transcriptome-wide association studies and comparative single-cell RNA-seq analyses to uncover the roles of them in mammalian reproductive performance. The genes have not been widely reported to be fertility-related genes and can be complementally considered as prior biological information to modify genomic selections models that benefits pig genetic improvement of litter traits. Besides, our findings provide new insights into the function of ROHet and ROH in mammals.
Collapse
|
138
|
De Silva K, Demmer RT, Jönsson D, Mousa A, Teede H, Forbes A, Enticott J. Causality of anthropometric markers associated with polycystic ovarian syndrome: Findings of a Mendelian randomization study. PLoS One 2022; 17:e0269191. [PMID: 35679284 PMCID: PMC9182303 DOI: 10.1371/journal.pone.0269191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/16/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction Using body mass index (BMI) as a proxy, previous Mendelian randomization (MR) studies found total causal effects of general obesity on polycystic ovarian syndrome (PCOS). Hitherto, total and direct causal effects of general- and central obesity on PCOS have not been comprehensively analyzed. Objectives To investigate the causality of central- and general obesity on PCOS using surrogate anthropometric markers. Methods Summary GWAS data of female-only, large-sample cohorts of European ancestry were retrieved for anthropometric markers of central obesity (waist circumference (WC), hip circumference (HC), waist-to-hip ratio (WHR)) and general obesity (BMI and its constituent variables–weight and height), from the IEU Open GWAS Project. As the outcome, we acquired summary data from a large-sample GWAS (118870 samples; 642 cases and 118228 controls) within the FinnGen cohort. Total causal effects were assessed via univariable two-sample Mendelian randomization (2SMR). Genetic architectures underlying causal associations were explored. Direct causal effects were analyzed by multivariable MR modelling. Results Instrumental variables demonstrated no weak instrument bias (F > 10). Four anthropometric exposures, namely, weight (2.69–77.05), BMI (OR: 2.90–4.06), WC (OR: 6.22–20.27), and HC (OR: 6.22–20.27) demonstrated total causal effects as per univariable 2SMR models. We uncovered shared and non-shared genetic architectures underlying causal associations. Direct causal effects of WC and HC on PCOS were revealed by two multivariable MR models containing exclusively the anthropometric markers of central obesity. Other multivariable MR models containing anthropometric markers of both central- and general obesity showed no direct causal effects on PCOS. Conclusions Both and general- and central obesity yield total causal effects on PCOS. Findings also indicated potential direct causal effects of normal weight-central obesity and more complex causal mechanisms when both central- and general obesity are present. Results underscore the importance of addressing both central- and general obesity for optimizing PCOS care.
Collapse
Affiliation(s)
- Kushan De Silva
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Clayton, Australia
- * E-mail:
| | - Ryan T. Demmer
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States of America
| | - Daniel Jönsson
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Malmö, Sweden
- Public Dental Service of Skane, Lund, Sweden
| | - Aya Mousa
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Clayton, Australia
| | - Helena Teede
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Clayton, Australia
| | - Andrew Forbes
- Biostatistics Unit, Division of Research Methodology, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Joanne Enticott
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Clayton, Australia
| |
Collapse
|
139
|
Desroziers E. Unusual suspects: Glial cells in fertility regulation and their suspected role in polycystic ovary syndrome. J Neuroendocrinol 2022; 34:e13136. [PMID: 35445462 PMCID: PMC9489003 DOI: 10.1111/jne.13136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 11/28/2022]
Abstract
Gonadotropin-releasing-hormone (GnRH) neurons sitting within the hypothalamus control the production of gametes and sex steroids by the gonads, therefore ensuring survival of species. As orchestrators of reproductive function, GnRH neurons integrate information from external and internal cues. This occurs through an extensively studied neuronal network known as the "GnRH neuronal network." However, the brain is not simply composed of neurons. Evidence suggests a role for glial cells in controlling GnRH neuron activity, secretion and fertility outcomes, although numerous questions remain. Glial cells have historically been seen as support cells for neurons. This idea has been challenged by the discovery that some neurological diseases originate from glial dysfunction. The prevalence of infertility disorders is increasing worldwide, with one in four couples being affected; therefore, it remains essential to understand the mechanisms by which the brain controls fertility. The "GnRH glial network" could be a major player in infertility disorders and represent a potential therapeutic target. In polycystic ovary syndrome (PCOS), the most common infertility disorder of reproductive aged women worldwide, the brain is considered a prime suspect. Recent studies have demonstrated pathological neuronal wiring of the "GnRH neuronal network" in PCOS-like animal models. However, the role of the "GnRH glial network" remains to be elucidated. In this review, I aim to propose glial cells as unusual suspects in infertility disorders such as PCOS. In the first part, I state our current knowledge about the role of glia in the regulation of GnRH neurons and fertility. In the second part, based on our recent findings, I discuss how glial cells could be implicated in PCOS pathology.
Collapse
Affiliation(s)
- Elodie Desroziers
- Department of Physiology, Centre for NeuroendocrinologyUniversity of OtagoDunedinNew Zealand
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine – Institut de Biologie Paris Seine, Neuroplasticity of Reproductive Behaviours TeamParisFrance
| |
Collapse
|
140
|
Alarcón-Granados MC, Moreno-Ortíz H, Esteban-Pérez CI, Ferrebuz-Cardozo A, Camargo-Villalba GE, Forero-Castro M. Assessment of THADA gene polymorphisms in a sample of Colombian women with polycystic ovary syndrome: A pilot study. Heliyon 2022; 8:e09673. [PMID: 35711992 PMCID: PMC9194581 DOI: 10.1016/j.heliyon.2022.e09673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/20/2021] [Accepted: 05/31/2022] [Indexed: 01/04/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a multifactorial and polygenic endocrine-metabolic disorder in women of reproductive age. SNPs in the THADA gene have been identified as PCOS risk loci. In this study, we evaluated the frequency of five polymorphisms in a sample of Colombian women with PCOS, and their association with clinical and endocrine-metabolic parameters. Forty-nine women with PCOS and forty-nine healthy women were included. Allelic discrimination was performed in the THADA gene by iPLEX and the MassARRAY system (Agena Bioscience). Haploview software was conducted to analyze the linkage disequilibrium (LD) and haplotypes of polymorphisms. There was an association between the genotypes TT of rs12468394, CC + AA of rs12468394, and GG of rs6544661 and an increase in the levels of free testosterone. The CC + AA of rs12468394 genotype also was associated with an increase of androstenedione levels. THADA gene SNPs were not associated with PCOS risk. There was very strong LD among the SNPs. No significant differences in the frequency of haplotypes between groups were observed. The statistical power of this analysis is low because of the small number of samples analyzed. Additional studies involving large populations of Colombian women with PCOS are needed to verify the role of the THADA gene in this disorder.
Collapse
Affiliation(s)
- Maria Camila Alarcón-Granados
- Facultad de Ciencias. Grupo de Investigación en Ciencias Biomédicas (GICBUPTC). Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia
| | | | | | - Atilio Ferrebuz-Cardozo
- Programa de Medicina. Facultad de Ciencias de La Salud. Universidad de Boyacá, Tunja, Colombia
| | | | - Maribel Forero-Castro
- Facultad de Ciencias. Grupo de Investigación en Ciencias Biomédicas (GICBUPTC). Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia
| |
Collapse
|
141
|
Roberts JF, Jeff Huang CC. Bovine models for human ovarian diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:101-154. [PMID: 35595347 DOI: 10.1016/bs.pmbts.2022.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
During early embryonic development, late fetal growth, puberty, adult reproductive years, and advanced aging, bovine and human ovaries closely share molecular pathways and hormonal signaling mechanisms. Other similarities between these species include the size of ovaries, length of gestation, ovarian follicular and luteal dynamics, and pathophysiology of ovarian diseases. As an economically important agriculture species, cattle are a foundational species in fertility research with decades of groundwork using physiologic, genetic, and therapeutic experimental techniques. Many technologies used in modern reproductive medicine, such as ovulation induction using hormonal therapy, were first used in cows before human trials. Human ovarian diseases with naturally occurring bovine correlates include premature ovary insufficiency (POI), polycystic ovarian syndrome (PCOS), and sex-cord stromal tumors (SCSTs). This article presents an overview of bovine ovary research related to causes of infertility, ovarian diseases, diagnostics, and therapeutics, emphasizing where the bovine model can offer advantages over other lab animals for translational applications.
Collapse
Affiliation(s)
- John F Roberts
- Department of Comparative, Diagnostic & Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States.
| | - Chen-Che Jeff Huang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
142
|
Waterbury JS, Teves ME, Gaynor A, Han AX, Mavodza G, Newell J, Strauss JF, McAllister JM. The PCOS GWAS Candidate Gene ZNF217 Influences Theca Cell Expression of DENND1A.V2, CYP17A1, and Androgen Production. J Endocr Soc 2022; 6:bvac078. [PMID: 35668995 PMCID: PMC9155636 DOI: 10.1210/jendso/bvac078] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Indexed: 12/25/2022] Open
Abstract
Polycystic ovary syndrome (PCOS), a common endocrine disorder of women, is characterized by increased ovarian androgen production and anovulatory infertility. Genome-wide association studies (GWAS) have identified more than 20 PCOS candidate loci. One GWAS candidate locus encompasses ZNF217, a zinc finger transcription factor. Immunohistochemical staining of ovarian tissue demonstrated significantly lower staining intensity for ZNF217 protein in PCOS theca interna compared to ovarian tissue from normal ovulatory women. Immunofluorescence staining of normal and PCOS theca cells demonstrated nuclear localization of ZNF217, with lower intensity in PCOS cells. Western blotting showed reduced ZNF217 protein in PCOS theca cells compared to normal theca cells, and that treatment with forskolin, which mimics the action of luteinizing hormone (LH), reduces ZNF217 expression. Lower ZNF217 expression in PCOS theca cells was confirmed by quantitative reverse transcription polymerase chain reaction. Notably, there was an inverse relationship between ZNF217 messenger RNA (mRNA) levels and theca cell androgen (dehydroepiandrosterone; DHEA) synthesis. The abundance of mRNA encoding a splice variant of DENND1A (DENND1A.V2), a PCOS candidate gene that positively regulates androgen biosynthesis, was also inversely related to ZNF217 mRNA levels. This relationship may be driven by increased miR-130b-3p, which targets DENND1A.V2 transcripts and is directly correlated with ZNF217 expression. Forced expression of ZNF217 in PCOS theca cells reduced androgen production, CYP17A1 and DENND1A.V2 mRNA, while increasing mIR-130b-3p. Conversely, knockdown of ZNF217 in normal theca cells with short hairpin RNA-expressing lentivirus particles increased DENND1A.V2 and CYP17A1 mRNA. These observations suggest that ZNF217 is part of a network of PCOS candidate genes regulating thecal cell androgen production involving DENND1A.V2 and miR-130b-3p.
Collapse
Affiliation(s)
- Jamaia S Waterbury
- Department of Pathology, Penn State Hershey College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Maria E Teves
- Department of Obstetrics and Gynecology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, USA
| | - Alison Gaynor
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, USA
| | - Angela X Han
- Department of Pathology, Penn State Hershey College of Medicine, Hershey, Pennsylvania 17033, USA
| | - Grace Mavodza
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, USA
| | - Jordan Newell
- Department of Pathology, Penn State Hershey College of Medicine, Hershey, Pennsylvania 17033, USA
| | | | - Jan M McAllister
- Correspondence: Jan M. McAllister, PhD, Department of Pathology, Penn State Hershey College of Medicine, 500 University Dr, Hershey, PA 17033, USA.
| |
Collapse
|
143
|
Verdiesen RMG, van der Schouw YT, van Gils CH, Verschuren WMM, Broekmans FJM, Borges MC, Gonçalves Soares AL, Lawlor DA, Eliassen AH, Kraft P, Sandler DP, Harlow SD, Smith JA, Santoro N, Schoemaker MJ, Swerdlow AJ, Murray A, Ruth KS, Onland-Moret NC. Genome-wide association study meta-analysis identifies three novel loci for circulating anti-Müllerian hormone levels in women. Hum Reprod 2022; 37:1069-1082. [PMID: 35274129 PMCID: PMC9071229 DOI: 10.1093/humrep/deac028] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 10/22/2021] [Indexed: 12/05/2022] Open
Abstract
STUDY QUESTION Can additional genetic variants for circulating anti-Müllerian hormone (AMH) levels be identified through a genome-wide association study (GWAS) meta-analysis including a large sample of premenopausal women? SUMMARY ANSWER We identified four loci associated with AMH levels at P < 5 × 10-8: the previously reported MCM8 locus and three novel signals in or near AMH, TEX41 and CDCA7. WHAT IS KNOWN ALREADY AMH is expressed by antral stage ovarian follicles in women, and variation in age-specific circulating AMH levels has been associated with disease outcomes. However, the physiological mechanisms underlying these AMH-disease associations are largely unknown. STUDY DESIGN, SIZE, DURATION We performed a GWAS meta-analysis in which we combined summary statistics of a previous AMH GWAS with GWAS data from 3705 additional women from three different cohorts. PARTICIPANTS/MATERIALS, SETTING, METHODS In total, we included data from 7049 premenopausal female participants of European ancestry. The median age of study participants ranged from 15.3 to 48 years across cohorts. Circulating AMH levels were measured in either serum or plasma samples using different ELISA assays. Study-specific analyses were adjusted for age at blood collection and population stratification, and summary statistics were meta-analysed using a standard error-weighted approach. Subsequently, we functionally annotated GWAS variants that reached genome-wide significance (P < 5 × 10-8). We also performed a gene-based GWAS, pathway analysis and linkage disequilibrium score regression and Mendelian randomization (MR) analyses. MAIN RESULTS AND THE ROLE OF CHANCE We identified four loci associated with AMH levels at P < 5 × 10-8: the previously reported MCM8 locus and three novel signals in or near AMH, TEX41 and CDCA7. The strongest signal was a missense variant in the AMH gene (rs10417628). Most prioritized genes at the other three identified loci were involved in cell cycle regulation. Genetic correlation analyses indicated a strong positive correlation among single nucleotide polymorphisms for AMH levels and for age at menopause (rg = 0.82, FDR = 0.003). Exploratory two-sample MR analyses did not support causal effects of AMH on breast cancer or polycystic ovary syndrome risk, but should be interpreted with caution as they may be underpowered and the validity of genetic instruments could not be extensively explored. LARGE SCALE DATA The full AMH GWAS summary statistics will made available after publication through the GWAS catalog (https://www.ebi.ac.uk/gwas/). LIMITATIONS, REASONS FOR CAUTION Whilst this study doubled the sample size of the most recent GWAS, the statistical power is still relatively low. As a result, we may still lack power to identify more genetic variants for AMH and to determine causal effects of AMH on, for example, breast cancer. Also, follow-up studies are needed to investigate whether the signal for the AMH gene is caused by reduced AMH detection by certain assays instead of actual lower circulating AMH levels. WIDER IMPLICATIONS OF THE FINDINGS Genes mapped to the MCM8, TEX41 and CDCA7 loci are involved in the cell cycle and processes such as DNA replication and apoptosis. The mechanism underlying their associations with AMH may affect the size of the ovarian follicle pool. Altogether, our results provide more insight into the biology of AMH and, accordingly, the biological processes involved in ovarian ageing. STUDY FUNDING/COMPETING INTEREST(S) Nurses' Health Study and Nurses' Health Study II were supported by research grants from the National Institutes of Health (CA172726, CA186107, CA50385, CA87969, CA49449, CA67262, CA178949). The UK Medical Research Council and Wellcome (217065/Z/19/Z) and the University of Bristol provide core support for ALSPAC. This publication is the work of the listed authors, who will serve as guarantors for the contents of this article. A comprehensive list of grants funding is available on the ALSPAC website (http://www.bristol.ac.uk/alspac/external/documents/grant-acknowledgements.pdf). Funding for the collection of genotype and phenotype data used here was provided by the British Heart Foundation (SP/07/008/24066), Wellcome (WT092830M and WT08806) and UK Medical Research Council (G1001357). M.C.B., A.L.G.S. and D.A.L. work in a unit that is funded by the University of Bristol and UK Medical Research Council (MC_UU_00011/6). M.C.B.'s contribution to this work was funded by a UK Medical Research Council Skills Development Fellowship (MR/P014054/1) and D.A.L. is a National Institute of Health Research Senior Investigator (NF-0616-10102). A.L.G.S. was supported by the study of Dynamic longitudinal exposome trajectories in cardiovascular and metabolic non-communicable diseases (H2020-SC1-2019-Single-Stage-RTD, project ID 874739). The Doetinchem Cohort Study was financially supported by the Ministry of Health, Welfare and Sports of the Netherlands. The funder had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. Ansh Labs performed the AMH measurements for the Doetinchem Cohort Study free of charge. Ansh Labs was not involved in the data analysis, interpretation or reporting, nor was it financially involved in any aspect of the study. R.M.G.V. was funded by the Honours Track of MSc Epidemiology, University Medical Center Utrecht with a grant from the Netherlands Organization for Scientific Research (NWO) (022.005.021). The Study of Women's Health Across the Nation (SWAN) has grant support from the National Institutes of Health (NIH), DHHS, through the National Institute on Aging (NIA), the National Institute of Nursing Research (NINR) and the NIH Office of Research on Women's Health (ORWH) (U01NR004061; U01AG012505, U01AG012535, U01AG012531, U01AG012539, U01AG012546, U01AG012553, U01AG012554, U01AG012495). The SWAN Genomic Analyses and SWAN Legacy have grant support from the NIA (U01AG017719). The Generations Study was funded by Breast Cancer Now and the Institute of Cancer Research (ICR). The ICR acknowledges NHS funding to the NIHR Biomedical Research Centre. The content of this manuscript is solely the responsibility of the authors and does not necessarily represent official views of the funders. The Sister Study was funded by the Intramural Research Program of the National Institutes of Health (NIH), National Institute of Environmental Health Sciences (Z01-ES044005 to D.P.S.); the AMH assays were supported by the Avon Foundation (02-2012-065 to H.B. Nichols and D.P.S.). The breast cancer genome-wide association analyses were supported by the Government of Canada through Genome Canada and the Canadian Institutes of Health Research, the 'Ministère de l'Économie, de la Science et de l'Innovation du Québec' through Genome Québec and grant PSR-SIIRI-701, The National Institutes of Health (U19 CA148065, X01HG007492), Cancer Research UK (C1287/A10118, C1287/A16563, C1287/A10710) and The European Union (HEALTH-F2-2009-223175 and H2020 633784 and 634935). All studies and funders are listed in Michailidou et al. (Nature, 2017). F.J.M.B. has received fees and grant support from Merck Serono and Ferring BV. D.A.L. has received financial support from several national and international government and charitable funders as well as from Medtronic Ltd and Roche Diagnostics for research that is unrelated to this study. N.S. is scientific consultant for Ansh Laboratories. The other authors declare no competing interests.
Collapse
Affiliation(s)
- Renée M G Verdiesen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Carla H van Gils
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - W M Monique Verschuren
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Frank J M Broekmans
- Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maria C Borges
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ana L Gonçalves Soares
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol NIHR Biomedical Research Centre, Bristol, UK
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Siobán D Harlow
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Nanette Santoro
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Colorado, USA
| | - Minouk J Schoemaker
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Anthony J Swerdlow
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Division of Breast Cancer Research, The Institute of Cancer Research, London, UK
| | - Anna Murray
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Katherine S Ruth
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - N Charlotte Onland-Moret
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
144
|
McCartney CR, Campbell RE, Marshall JC, Moenter SM. The role of gonadotropin-releasing hormone neurons in polycystic ovary syndrome. J Neuroendocrinol 2022; 34:e13093. [PMID: 35083794 PMCID: PMC9232905 DOI: 10.1111/jne.13093] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/21/2021] [Accepted: 01/11/2022] [Indexed: 01/28/2023]
Abstract
Given the critical central role of gonadotropin-releasing hormone (GnRH) neurons in fertility, it is not surprising that the GnRH neural network is implicated in the pathology of polycystic ovary syndrome (PCOS), the most common cause of anovulatory infertility. Although many symptoms of PCOS relate most proximately to ovarian dysfunction, the central reproductive neuroendocrine system ultimately drives ovarian function through its regulation of anterior pituitary gonadotropin release. The typical cyclical changes in frequency of GnRH release are often absent in women with PCOS, resulting in a persistent high-frequency drive promoting gonadotropin changes (i.e., relatively high luteinizing hormone and relatively low follicle-stimulating hormone concentrations) that contribute to ovarian hyperandrogenemia and ovulatory dysfunction. However, the specific mechanisms underpinning GnRH neuron dysfunction in PCOS remain unclear. Here, we summarize several preclinical and clinical studies that explore the causes of aberrant GnRH secretion in PCOS and the role of disordered GnRH secretion in PCOS pathophysiology.
Collapse
Affiliation(s)
- Christopher R. McCartney
- Center for Research in Reproduction and Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVAUSA
| | - Rebecca E. Campbell
- Centre for Neuroendocrinology and Department of PhysiologySchool of Biomedical SciencesUniversity of OtagoDunedinNew Zealand
| | - John C. Marshall
- Center for Research in Reproduction and Department of MedicineUniversity of Virginia School of MedicineCharlottesvilleVAUSA
| | - Suzanne M. Moenter
- Departments of Molecular & Integrative PhysiologyInternal MedicineObstetrics and GynecologyUniversity of MichiganAnn ArborMIUSA
| |
Collapse
|
145
|
Guan C, Zahid S, Minhas AS, Ouyang P, Vaught A, Baker VL, Michos ED. Polycystic ovary syndrome: a "risk-enhancing" factor for cardiovascular disease. Fertil Steril 2022; 117:924-935. [PMID: 35512976 DOI: 10.1016/j.fertnstert.2022.03.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 12/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among women of reproductive age and is hallmarked by hyperandrogenism, oligo-ovulation, and polycystic ovarian morphology. Polycystic ovary syndrome, particularly the hyperandrogenism phenotype, is associated with several cardiometabolic abnormalities, including obesity, dyslipidemia, elevated blood pressure, and prediabetes or type 2 diabetes. Many, but not all, studies have suggested that PCOS is associated with increased risk of cardiovascular disease (CVD), including coronary heart disease and stroke, independent of body mass index and traditional risk factors. Interpretation of the data from these observational studies is limited by the varying definitions and ascertainment of PCOS and CVD across studies. Recent Mendelian randomization studies have challenged the causality of PCOS with coronary heart disease and stroke. Future longitudinal studies with clearly defined PCOS criteria and newer genetic methodologies may help to determine association and causality. Nevertheless, CVD risk screening remains critical in this patient population, as improvements in metabolic profile and reduction in CVD risk are achievable with a combination of lifestyle management and pharmacotherapy. Statin therapy should be implemented in women with PCOS who have elevated atherosclerotic CVD risk. If CVD risk is uncertain, measurement of subclinical atherosclerosis (carotid plaque or coronary artery calcium) may be a useful tool to guide shared decision-making about initiation of statin therapy. Other medications, such as metformin and glucagon-like peptide-1 receptor agonists, also may be useful in reducing CVD risk in insulin-resistant populations. Additional research is needed to determine the best pathways to mitigate PCOS-associated CVD risk.
Collapse
Affiliation(s)
- Carolyn Guan
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Salman Zahid
- Sands-Constellation Heart Institute, Rochester General Hospital, Rochester, New York
| | - Anum S Minhas
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Pamela Ouyang
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Arthur Vaught
- Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Valerie L Baker
- Division of Reproductive Endocrinology and Infertility, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Erin D Michos
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.
| |
Collapse
|
146
|
Welt CK. What Is the Male Polycystic Ovary Syndrome Phenotype? J Clin Endocrinol Metab 2022; 107:e2188-e2189. [PMID: 34905059 DOI: 10.1210/clinem/dgab898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Indexed: 11/19/2022]
Affiliation(s)
- Corrine K Welt
- Division of Endocrinology, Metabolism and Diabetes, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
147
|
Simons PIHG, Cornelissen MEB, Valkenburg O, Onland‐Moret NC, van der Schouw YT, Stehouwer CD, Burgess S, Brouwers MCGJ. Causal relationship between polycystic ovary syndrome and coronary artery disease: A Mendelian randomisation study. Clin Endocrinol (Oxf) 2022; 96:599-604. [PMID: 34524719 PMCID: PMC7612926 DOI: 10.1111/cen.14593] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) has been associated with an increased risk of coronary artery disease (CAD). However, it remains uncertain whether this increased risk is the result of PCOS per se or, alternatively, is explained by obesity, a common feature of PCOS. The aim of this study was to assess the causal association between PCOS and CAD and the role of obesity herein. DESIGN AND METHODS We conducted two-sample Mendelian randomisation analyses in large-scale, female-specific datasets to study the association between genetically predicted (1) risk of PCOS and risk of CAD, (2) body mass index (BMI) and risk of PCOS and (3) BMI and risk of CAD. Primary analyses were conducted with the inverse-variance weighted (IVW) method. Simple median, penalized weighted median and contamination mixture analyses were performed to assess the robustness of the outcomes. RESULTS IVW analyses did not show a statistically significant association between PCOS and CAD (odds ratio [OR]: 0.99, 95% confidence interval [CI]: 0.89, 1.11). In contrast, genetically predicted BMI was statistically significantly associated with an increased odds of PCOS (OR: 3.21, 95% CI: 2.26, 4.56) and CAD (OR: 1.38, 95% CI: 1.14, 1.67). Similar results were obtained when secondary analyses were performed. CONCLUSION These sex-specific analyses show that the genetically predicted risk of PCOS is not associated with the risk of CAD. Instead, the genetically predicted risk of obesity (and its downstream metabolic effects) is the common denominator of both PCOS and CAD risk.
Collapse
Affiliation(s)
- Pomme I. H. G. Simons
- Department of Internal Medicine, Division of Endocrinology and Metabolic DiseasesMaastricht University Medical CenterMaastrichtThe Netherlands
- Laboratory for Metabolism and Vascular MedicineMaastricht UniversityMaastrichtThe Netherlands
- CARIM School for Cardiovascular DiseasesMaastricht UniversityMaastrichtThe Netherlands
| | - Merel E. B. Cornelissen
- Julius Center for Health Sciences and Primary Care, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Olivier Valkenburg
- Department of Reproductive MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
| | - N. Charlotte Onland‐Moret
- Julius Center for Health Sciences and Primary Care, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Yvonne T. van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Coen D.A. Stehouwer
- Laboratory for Metabolism and Vascular MedicineMaastricht UniversityMaastrichtThe Netherlands
- CARIM School for Cardiovascular DiseasesMaastricht UniversityMaastrichtThe Netherlands
- Department of Internal Medicine, Division of General Internal MedicineMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Stephen Burgess
- Department of Public Health and Primary CareUniversity of CambridgeCambridgeUK
- MRC Biostatistics UnitUniversity of CambridgeCambridgeUK
| | - Martijn C. G. J. Brouwers
- Department of Internal Medicine, Division of Endocrinology and Metabolic DiseasesMaastricht University Medical CenterMaastrichtThe Netherlands
- CARIM School for Cardiovascular DiseasesMaastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
148
|
Affiliation(s)
- Anju E Joham
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia.
- Department of Endocrinology and Diabetes, Monash Health, Melbourne, Victoria, Australia.
| | - Helena J Teede
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Endocrinology and Diabetes, Monash Health, Melbourne, Victoria, Australia
| |
Collapse
|
149
|
Silva MSB, Campbell RE. Polycystic Ovary Syndrome and the Neuroendocrine Consequences of Androgen Excess. Compr Physiol 2022; 12:3347-3369. [PMID: 35578968 DOI: 10.1002/cphy.c210025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a major endocrine disorder strongly associated with androgen excess and frequently leading to female infertility. Although classically considered an ovarian disease, altered neuroendocrine control of gonadotropin-releasing hormone (GnRH) neurons in the brain and abnormal gonadotropin secretion may underpin PCOS presentation. Defective regulation of GnRH pulse generation in PCOS promotes high luteinizing hormone (LH) pulsatile secretion, which in turn overstimulates ovarian androgen production. Early and emerging evidence from preclinical models suggests that maternal androgen excess programs abnormalities in developing neuroendocrine circuits that are associated with PCOS pathology, and that these abnormalities are sustained by postpubertal elevation of endogenous androgen levels. This article will discuss experimental evidence, from the clinic and in preclinical animal models, that has significantly contributed to our understanding of how androgen excess influences the assembly and maintenance of neuroendocrine impairments in the female brain. Abnormal central gamma-aminobutyric acid (GABA) signaling has been identified in both patients and preclinical models as a possible link between androgen excess and elevated GnRH/LH secretion. Enhanced GABAergic innervation and drive to GnRH neurons is suspected to contribute to the pathogenesis and early manifestation of neuroendocrine derangement in PCOS. Accordingly, this article also provides an overview of GABA regulation of GnRH neuron function from prenatal development to adulthood to discuss possible avenues for future discovery research and therapeutic interventions. © 2022 American Physiological Society. Compr Physiol 12:3347-3369, 2022.
Collapse
Affiliation(s)
- Mauro S B Silva
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rebecca E Campbell
- Centre for Neuroendocrinology, Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
150
|
Zhu J, Pujol-Gualdo N, Wittemans LBL, Lindgren CM, Laisk T, Hirschhorn JN, Chan YM. Evidence From Men for Ovary-independent Effects of Genetic Risk Factors for Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2022; 107:e1577-e1587. [PMID: 34969092 PMCID: PMC8947237 DOI: 10.1210/clinem/dgab838] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 11/01/2021] [Indexed: 01/17/2023]
Abstract
CONTEXT Polycystic ovary syndrome (PCOS) is characterized by ovulatory dysfunction and hyperandrogenism and can be associated with cardiometabolic dysfunction, but it remains unclear which of these features are inciting causes and which are secondary consequences. OBJECTIVE To determine whether ovarian function is necessary for genetic risk factors for PCOS to produce nonreproductive phenotypes. DESIGN, SETTING, AND PARTICIPANTS Cohort of 176 360 men in the UK Biobank and replication cohort of 37 348 men in the Estonian Biobank. MAIN OUTCOME MEASURES We calculated individual PCOS polygenic risk scores (PRS), tested for association of these PRS with PCOS-related phenotypes using linear and logistic regression and performed mediation analysis. RESULTS For every 1 SD increase in the PCOS PRS, men had increased odds of obesity (odds ratio [OR]: 1.09; 95% CI, 1.08-1.10; P = 1 × 10-49), type 2 diabetes mellitus (T2DM) (OR: 1.08; 95% CI, 1.05-1.10; P = 3 × 10-12), coronary artery disease (CAD) (OR: 1.03; 95% CI, 1.01-1.04; P = 0.0029), and marked androgenic alopecia (OR: 1.03; 95% CI, 1.02-1.05; P = 3 × 10-5). Body mass index (BMI), hemoglobin A1c, triglycerides, and free androgen index increased as the PRS increased, whereas high-density lipoprotein cholesterol and SHBG decreased (all P < .0001). The association between the PRS and CAD appeared to be completely mediated by BMI, whereas the associations with T2DM and marked androgenic alopecia appeared to be partially mediated by BMI. CONCLUSIONS Genetic risk factors for PCOS have phenotypic consequences in men, indicating that they can act independently of ovarian function. Thus, PCOS in women may not always be a primary disorder of the ovaries.
Collapse
Affiliation(s)
- Jia Zhu
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Natàlia Pujol-Gualdo
- Estonian Genome Centre, Institute of Genomics, University of Tartu 51010, Tartu, Estonia
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu FI-90014, Oulu, Finland
| | - Laura B L Wittemans
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 ZFZ, UK
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
| | - Cecilia M Lindgren
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7FZ, UK
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu 51010, Tartu, Estonia
| | - Joel N Hirschhorn
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Yee-Ming Chan
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|