1
|
Bazzazzadehgan S, Shariat-Madar Z, Mahdi F. Distinct Roles of Common Genetic Variants and Their Contributions to Diabetes: MODY and Uncontrolled T2DM. Biomolecules 2025; 15:414. [PMID: 40149950 PMCID: PMC11940602 DOI: 10.3390/biom15030414] [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: 12/30/2024] [Revised: 01/26/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) encompasses a range of clinical manifestations, with uncontrolled diabetes leading to progressive or irreversible damage to various organs. Numerous genes associated with monogenic diabetes, exhibiting classical patterns of inheritance (autosomal dominant or recessive), have been identified. Additionally, genes involved in complex diabetes, which interact with environmental factors to trigger the disease, have also been discovered. These genetic findings have raised hopes that genetic testing could enhance diagnostics, disease surveillance, treatment selection, and family counseling. However, the accurate interpretation of genetic data remains a significant challenge, as variants may not always be definitively classified as either benign or pathogenic. Research to date, however, indicates that periodic reevaluation of genetic variants in diabetes has led to more consistent findings, with biases being steadily eliminated. This has improved the interpretation of variants across diverse ethnicities. Clinical studies suggest that genetic risk information may motivate patients to adopt behaviors that promote the prevention or management of T2DM. Given that the clinical features of certain monogenic diabetes types overlap with T2DM, and considering the significant role of genetic variants in diabetes, healthcare providers caring for prediabetic patients should consider genetic testing as part of the diagnostic process. This review summarizes current knowledge of the most common genetic variants associated with T2DM, explores novel therapeutic targets, and discusses recent advancements in the pharmaceutical management of uncontrolled T2DM.
Collapse
Affiliation(s)
- Shadi Bazzazzadehgan
- Department of Pharmacy Administration, School of Pharmacy, University of Mississippi, University, MS 38677, USA;
| | - Zia Shariat-Madar
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA;
| | - Fakhri Mahdi
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA;
| |
Collapse
|
2
|
Zhang C, Yu J. FTO gene polymorphism and susceptibility to polycystic ovary syndrome: A meta-analysis. J Obstet Gynaecol Res 2024; 50:1703-1712. [PMID: 39143730 DOI: 10.1111/jog.16047] [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: 01/21/2024] [Accepted: 07/25/2024] [Indexed: 08/16/2024]
Abstract
AIM To explore the correlation between Fat mass and objectivity associated gene (FTO) rs9939609 polymorphism and susceptibility to polycystic ovary syndrome. METHODS Case-control studies on the relationship between FTO rs9939609 A/T polymorphism and PCOS were searched in PubMed, EMBASE, and Web of Science according to inclusion and exclusion criteria. STATA 12.0 software was conducted for Meta-analysis. RESULTS Nine case-control studies were included, including 1410 cases in PCOS group and 1223 cases in healthy control group. The results of meta-analysis showed that FTO rs9939609 gene polymorphism was associated with PCOS susceptibility, and the risk of developing PCOS was 1.19 times higher for T alleles carriers than for A alleles carriers, and some similar associations were observed in Asian populations. CONCLUSIONS In summary, FTO rs9939609 gene polymorphism is significantly associated with PCOS susceptibility, especially in Asian populations.
Collapse
Affiliation(s)
- Chuanhua Zhang
- Department of Gynecology, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, China
| | - Jiali Yu
- Department of Gynecology and Obstetrics, Yiwu Central Hospital, Yiwu, China
| |
Collapse
|
3
|
Stephen SB, Pauline R, Velmurugan S, Subbaraj GK. An association between fat mass and obesity-associated (FTO) (rs9939609) and kisspeptin-1 (KISS-1) (rs4889, rs372790354) gene polymorphisms with polycystic ovary syndrome: an updated meta-analysis and power analysis. J Assist Reprod Genet 2024; 41:2457-2475. [PMID: 39126595 PMCID: PMC11405594 DOI: 10.1007/s10815-024-03213-7] [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: 03/08/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
INTRODUCTION The present meta-analysis aimed to investigate FTO rs9939609 and KISS1 rs4889, rs372790354 gene polymorphisms and its association with PCOS in Asian population. METHODS The studies included in this article were obtained by using online databases. We searched databases such as Scopus, PubMed, Embase, and Web of Science for case-control articles related to FTO and KISS1 gene polymorphism with PCOS. Metagenyo software was used to determine the 95% confidence interval (CI) and odds ratio (OR). RESULTS A total of 13 articles was included in this meta-analysis for FTO (rs9939609) and KISS1 (rs4889; rs372790354) gene polymorphisms related with PCOS in the Asian population. According to the findings of this study, people with FTO rs9939609 show an association with PCOS risk in dominant model. On contradictory, KISS1 gene polymorphism specifically, rs4889 show an association with PCOS risk in allelic, recessive, and dominant models whereas rs372790354 show an association with PCOS risk in allelic and dominant models. Power analysis was performed and PPI is > 0.04. The sting analysis network for FTO and KISS1 gene estimated 12 nodes and 23 edges. DISCUSSION The FTO rs9939609 variant exhibits an association with an increased risk of PCOS in the dominant model. KISS1 gene polymorphism, particularly rs4889, shows a significant association with PCOS risk in allelic, recessive, and dominant models. Similarly, KISS1 rs372790354 gene is associated with PCOS risk in both allelic and dominant models. Researches were focused only on the Asian population so; it is imperative to conduct further research across diverse populations.
Collapse
Affiliation(s)
- Sharon Benita Stephen
- Faculty of Allied Health Science, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Rashmi Pauline
- Faculty of Allied Health Science, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Saranya Velmurugan
- Faculty of Allied Health Science, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Gowtham Kumar Subbaraj
- Faculty of Allied Health Science, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India.
| |
Collapse
|
4
|
Li X, Jiang B, Gao T, Nian Y, Bai X, Zhong J, Qin L, Gao Z, Wang H, Ma X. Effects of inulin on intestinal flora and metabolism-related indicators in obese polycystic ovary syndrome patients. Eur J Med Res 2024; 29:443. [PMID: 39217395 PMCID: PMC11365155 DOI: 10.1186/s40001-024-02034-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
CONTEXT Polycystic ovary syndrome (PCOS), a common endocrine disorder in women of reproductive age, is closely associated with chronic low-grade inflammation and metabolic disturbances. In PCOS mice, dietary inulin has been demonstrated to regulate intestinal flora and inflammation. However, the efficacy of dietary inulin in clinical PCOS remains unclear. OBJECTIVE The intestinal flora and related metabolic indexes of obese patients with polycystic ovary syndrome (PCOS) after 3 months of inulin treatment were analyzed. SETTING AND DESIGN To analyze the intestinal flora and related metabolic indexes in healthy controls and obese patients with polycystic ovary syndrome after 3 months of inulin treatment. RESULTS The results showed that dietary inulin improved sex hormone disorders, reduced BMI and WHR levels in obese women with PCOS. In addition, the inulin intervention reduced plasma TNF-α, IL-1β, IL-6, and MCP-1levels. Inulin intervention increased the abundance of Actinobacteria, Fusobacteria, Lachnospira, and Bifidobacterium, as well as decreased the ratio of F/B and the abundance of proteobacteria, Sutterella, and Enterobacter. Correlation analyses showed a strong relationship among plasma inflammatory factors, sex steroid hormones, and the intestinal flora of patients. CONCLUSIONS Dietary inulin may improve obese PCOS women disease through the gut flora-inflammation-steroid hormone pathway. THE CLINICAL TRIAL REGISTRATION NUMBER ChiCTR-IOR-17012281.
Collapse
Affiliation(s)
- Xiaorong Li
- Center for Reproductive Medicine, General Hospital of Ningxia Medical University, 164, Zhiping Road, Yinchuan, 750004, Ningxia, China
- Key Laboratory of Fertility Maintenance, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
- College of Traditional Chinese Medicine, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
| | - Bo Jiang
- Key Laboratory of Fertility Maintenance, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
- Key Laboratory of Modernization of Hui Medicine, Ministry of Education, School of Traditional Chinese Medicine, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
- College of Traditional Chinese Medicine, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
| | - Ting Gao
- Chengdu Integrated, TCM&Western Medicine Hospital, 18, Wanxiang North Road, Chengdu, 610095, Sichuan, China
| | - Yan Nian
- Center for Reproductive Medicine, General Hospital of Ningxia Medical University, 164, Zhiping Road, Yinchuan, 750004, Ningxia, China
| | - Xing Bai
- Key Laboratory of Modernization of Hui Medicine, Ministry of Education, School of Traditional Chinese Medicine, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
- College of Traditional Chinese Medicine, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
| | - Jiawen Zhong
- Key Laboratory of Fertility Maintenance, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
- Key Laboratory of Modernization of Hui Medicine, Ministry of Education, School of Traditional Chinese Medicine, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
- College of Traditional Chinese Medicine, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
| | - Ling Qin
- College of Traditional Chinese Medicine, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
| | - Zhengzheng Gao
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
| | - Hao Wang
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, 1160, Shengli Street, Yinchuan, 750004, Ningxia, China
| | - Xiaohong Ma
- Center for Reproductive Medicine, General Hospital of Ningxia Medical University, 164, Zhiping Road, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
5
|
Zhang Q, Ke W, Ye J, Zhang P, Yang Q, Pan F, Wang K, Zha B. Is thyroid function associated with polycystic ovary syndrome? A bidirectional Mendelian randomization study. Endocrine 2024; 85:380-391. [PMID: 38472621 DOI: 10.1007/s12020-024-03756-w] [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: 10/27/2023] [Accepted: 02/18/2024] [Indexed: 03/14/2024]
Abstract
OBJECTIVE Some observational studies have suggested the association between thyroid function and polycystic ovary syndrome (PCOS). However, it remains to be determined whether these associations are causal or not. The aim of this study was to investigate the underlying causal association between different thyroid function status and PCOS. METHODS Bidirectional Mendelian randomization (MR) analysis was conducted to explore the impact of different thyroid function statuses on PCOS. The study included 10,074 individuals with PCOS and 103,164 controls for the primary analysis, with validation analysis repeated in the FinnGen R9 and EstBB PCOS cohorts. Female-specific thyroid function GWAS data were obtained from European population, including Hyperthyroidism (22,383 cases and 54,288 controls) and Hypothyroidism (27,383 cases and 54,288 controls) from the UK Biobank, and TSH (54,288 cases and 72,167 controls) and FT4 (49,269 cases and 72,167 controls) within the reference range from the ThyroidOmics Consortium. Inverse variance weighting (IVW) was chosen as the principal method, and sensitivity analysis was conducted to test for the presence of horizontal pleiotropy or heterogeneity. RESULTS The IVW analysis indicated nominal significance between normal TSH levels and PCOS after adjusted for age and BMI [OR (95% CI) = 0.78(0.62,0.97), P = 0.029], suggesting that maintaining normal TSH levels might act as a protective factor against the pathogenesis of PCOS. Besides, in order to increase the statistical power, we pooled PCOS GWAS above together by meta-analysis and found PCOS contributed to the occurrence of hyperthyroidism [OR(95%CI) = 1.37(0.73,2.57), P = 0.012]. However, no causal relationship was found after Bonferroni correction (P-value < 0.0031). CONCLUSION Although the MR analysis didn't indicate genetic causal association between thyroid function and PCOS after Bonferroni correction. Further efforts are needed to interpret the potential causal relationship between thyroid function and PCOS in different age and BMI subgroup.
Collapse
Affiliation(s)
- Qinnan Zhang
- Department of Endocrinology, Fifth People's Hospital of Shanghai Fudan University, Shanghai, China
| | - Wencai Ke
- Department of Clinical Laboratory Medicine, Fifth People's Hospital of Shanghai Fudan University, Shanghai, China
| | - Jun Ye
- Department of Obstetrics and Gynecology, Fifth People's Hospital of Shanghai Fudan University, Shanghai, China
| | - Panpan Zhang
- Department of Endocrinology, Fifth People's Hospital of Shanghai Fudan University, Shanghai, China
| | - Qian Yang
- Department of Endocrinology, Fifth People's Hospital of Shanghai Fudan University, Shanghai, China
| | - Fanfan Pan
- Department of Endocrinology, Fifth People's Hospital of Shanghai Fudan University, Shanghai, China
| | - Kai Wang
- Department of Endocrinology, Fifth People's Hospital of Shanghai Fudan University, Shanghai, China.
| | - Bingbing Zha
- Department of Endocrinology, Fifth People's Hospital of Shanghai Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Xu Q, Zhang J, Lu Y, Wu L. Association of metabolic-dysfunction associated steatotic liver disease with polycystic ovary syndrome. iScience 2024; 27:108783. [PMID: 38292434 PMCID: PMC10825666 DOI: 10.1016/j.isci.2024.108783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), which has a prevalence of over 25% in adults, encompasses a wide spectrum of liver diseases. Metabolic-dysfunction associated steatotic liver disease (MASLD), the new term for NAFLD, is characterized by steatotic liver disease accompanied by cardiometabolic criteria, showing a strong correlation with metabolic diseases. Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disease affecting 4-21% of women of reproductive age. Numerous studies have indicated that NAFLD and PCOS often occur together. However, as MASLD is a new term, there is still a lack of reports describing the effects of MASLD on the development of PCOS. In this review article, we have summarized the complex and multifaceted connections between MASLD and PCOS. Understanding the pathogenesis and treatment methods could not only guide the clinical prevention, diagnosis, and treatment of PCOS in patients with MASLD, but also increase the clinical attention of reproductive doctors to MASLD.
Collapse
Affiliation(s)
- Qiuyu Xu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Metabolism and Regenerative Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhang
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Lu
- Institute of Metabolism and Regenerative Medicine, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Wu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Kim M, Lee S, Cho E, Hong KW, You SJ, Choi HJ. CETP and APOA2 polymorphisms are associated with weight loss and healthy eating behavior changes in response to digital lifestyle modifications. Sci Rep 2023; 13:21615. [PMID: 38062157 PMCID: PMC10703771 DOI: 10.1038/s41598-023-48823-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Response to digital healthcare lifestyle modifications is highly divergent. This study aimed to examine the association between single nucleotide polymorphism (SNP) genotypes and clinical efficacy of a digital healthcare lifestyle modification. We genotyped 97 obesity-related SNPs from 45 participants aged 18-39 years, who underwent lifestyle modification via digital cognitive behavioral therapy for obesity for 8 weeks. Anthropometric, eating behavior phenotypes, and psychological measures were analyzed before and after the intervention to identify their clinical efficacy. CETP (rs9939224) SNP significantly predict "super-responders" with greater body mass index (BMI) reduction (p = 0.028; GG - 2.91%, GT - 9.94%), while APOA2 (rs5082) appeared to have some potential for predicting "poor-responders" with lower BMI reduction (p = 0.005; AA - 6.17%, AG + 2.05%, and GG + 5.11%). These SNPs was also associated with significant differences in eating behavior changes, healthy diet proportions, health diet diversity, emotional and restrained eating behavior changes. Furthermore, classification using gene-gene interactions between rs9939224 and rs5082 significantly predicted the best response, with a greater decrease in BMI (p = 0.038; - 11.45% for the best response group (CEPT GT/TT × APOA2 AA) vs. + 2.62% for the worst response group (CEPT GG × APOA2 AG/GG)). CETP and APOA2 SNPs can be used as candidate markers to predict the efficacy of digital healthcare lifestyle modifications based on genotype-based precision medicine.Trial registration: NCT03465306, ClinicalTrials.gov. Registered March, 2018.
Collapse
Affiliation(s)
- Meelim Kim
- Department of Biomedical Science, Seoul National University College of Medicine, 28 Yungun-Dong, Chongno-Gu, Seoul, 110-799, Korea
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA, USA
- Center for Wireless and Population Health Systems, Calit2's Qualcomm Institute, University of California, San Diego, La Jolla, CA, USA
- The Design Lab, University of California, San Diego, La Jolla, CA, USA
| | - Seolha Lee
- Department of Biomedical Science, Seoul National University College of Medicine, 28 Yungun-Dong, Chongno-Gu, Seoul, 110-799, Korea
- Department of Global Medical Science, Sungshin Women's University, Seoul, Korea
| | - Eun Cho
- Department of Biomedical Science, Seoul National University College of Medicine, 28 Yungun-Dong, Chongno-Gu, Seoul, 110-799, Korea
| | | | | | - Hyung Jin Choi
- Department of Biomedical Science, Seoul National University College of Medicine, 28 Yungun-Dong, Chongno-Gu, Seoul, 110-799, Korea.
| |
Collapse
|
8
|
Almawi WY, Nemr R, Atazhanova T, Malalla ZH, Sarray S, Mustafa FE, Mahmood NA. Differential Association of FTO Gene variants and Haplotypes with the Susceptibility to Polycystic Ovary Syndrome According To Obesity in Women with PCOS. Reprod Sci 2023; 30:2166-2176. [PMID: 36602653 DOI: 10.1007/s43032-022-01149-w] [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: 09/21/2022] [Accepted: 12/11/2022] [Indexed: 01/06/2023]
Abstract
We explored the relation between FTO single gene variants (rs1861868, rs9939973, rs1421085, rs1121980, rs17817449, rs8050136, rs9939609, rs9930506, and rs8044769) and polycystic ovary syndrome (PCOS), in particular, according to the obesity status. This retrospective population-based case-control study involved women with PCOS (583) and 713 eumenorrheic control women; genotyping was done by real-time PCR. Significantly higher minor allele frequency (MAF) of rs9939973, rs17817449, rs9939609, and rs9930506 and lower MAF of rs1121980 were seen in PCOS cases. Lower risk of PCOS was associated with rs1121980 and rs8050136 heterozygous and minor allele-homozygous genotypes, while an elevated risk of PCOS was seen with minor allele-homozygous rs9939973, rs17817449, and r9939609 heterozygous and genotypes and minor allele-homozygous rs9930506 and rs8044769 genotype. While none of the tested FTO SNPs variants was associated with PCOS in normal body weight/lean subjects, rs9939973, rs9939609, and rs9930506 were negatively associated with PCOS in overweight subjects. In comparison, rs1861868 was negatively, while rs8044769 was positively associated with PCOS in obese subjects. Haplotype analysis identified haplotypes GACCTCTAT, AACCTCTAT, AACCTATAT and AGTTGCAGC, and GACCTCTAC to be positively associated with PCOS, while haplotypes GGTTGAAGC, GACCTATAT, GGTTGCAGC, and GATCTATAT were negatively associated with PCOS. Apart from GGTTGAAGC, these haplotypes remained associated with altered risk of PCOS after adjusting for covariates. In addition to rs17817449, rs9939609, rs9930506, and rs1121980, this study is the first to demonstrate association of rs9939973 and rs8044769 with altered risk of PCOS and the first to confirm the BMI dependency on the association of FTO variants with PCOS. This underscores the role of FTO gene variants as predisposing factors of PCOS.
Collapse
Affiliation(s)
- Wassim Y Almawi
- School of Medicine, Nazarbayev University, Astana, Kazakhstan.
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| | - Rita Nemr
- School of Medicine, Lebanese American University, Beirut, Lebanon
| | | | - Zainab H Malalla
- Department of Medical Biochemistry, Arabian Gulf University, Manama, Bahrain
| | - Sameh Sarray
- Department of Medical Biochemistry, Arabian Gulf University, Manama, Bahrain
| | - Fekria E Mustafa
- Department of Obstetrics and Gynecology, Salmaniya Medical Complex, Manama, Bahrain
| | - Naeema A Mahmood
- Department of Obstetrics and Gynecology, Salmaniya Medical Complex, Manama, Bahrain
| |
Collapse
|
9
|
van Baal L, Tan S. [Polycystic ovary syndrome as a gender-specific cardiometabolic risk factor]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2023:10.1007/s00108-023-01529-7. [PMID: 37291369 DOI: 10.1007/s00108-023-01529-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/10/2023]
Abstract
With a prevalence of 15%, polycystic ovary syndrome (PCOS) is the most common endocrinopathy in fertile-aged women. Insulin resistance and obesity play a pivotal role in the pathophysiology of PCOS, modulate the severity of symptoms and are associated with an increased risk for cardiometabolic sequelae such as diabetes, non-alcoholic fatty liver disease and atherosclerotic cardiovascular disease. PCOS should be considered as a gender-specific cardiovascular risk factor. Therefore, if traits indicative for PCOS are present, affected women should undergo PCOS diagnostics as a first step, thereby making it possible to initiate cardiovascular primary prevention strategies in this population of young women at high cardiometabolic risk. In women with known PCOS, screening and treatment of cardiometabolic risk factors and/or diseases should be routinely integrated into the concept of PCOS care. The close link between insulin resistance/obesity and PCOS can be used to improve PCOS-specific symptoms and enhance cardiometabolic health.
Collapse
Affiliation(s)
- Lukas van Baal
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen, Universität Duisburg-Essen, Hufelandstr. 55, 45721, Essen, Deutschland
| | - Susanne Tan
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen, Universität Duisburg-Essen, Hufelandstr. 55, 45721, Essen, Deutschland.
| |
Collapse
|
10
|
Bhandary P, Shetty PK, Manjeera L, Patil P. Hormonal, genetic, epigenetic and environmental aspects of polycystic ovarian syndrome. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
11
|
Hoseini Tavassol Z, Mousavi SM, Molaei B, Bandarian F, Ejtahed HS, Khalagi K, Ghannadi S, Larijani B, Hasani-Ranjbar S. Association of fat mass and obesity-associated ( FTO) gene polymorphisms with non-communicable diseases (NCDs) in the Iranian population: A systematic review of observational studies. J Diabetes Metab Disord 2022; 21:1975-1989. [PMID: 36404828 PMCID: PMC9672241 DOI: 10.1007/s40200-022-01139-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 10/03/2022] [Indexed: 10/31/2022]
Abstract
Background Single nucleotide polymorphisms have been implicated in various diseases, most notably non-communicable diseases (NCDs). The aim of this study was to review available evidence regarding associations between FTO polymorphisms and NCDs in the Iranian population. Methods A comprehensive search was conducted through PubMed/Medline and Scopus databases up to December 2021, as well as reference lists of pertinent articles and key journals. All observational studies that examined the association between FTO gene polymorphisms and NCDs in the Iranian population were included. There was no limitation on the publication year. The Newcastle-Ottawa Scale (NOS) was used to assess the study's quality. Results The initial search yielded 95 studies, of which 30 studies were included in the current systematic review. The underlying disorders were obesity, type 2 diabetes, breast and colorectal cancers, depression, and metabolic syndrome. These studies found an association between FTO gene polymorphisms and obesity in the Iranian population, but the relationship with other NCDs was debatable. Even though, other diseases such as diabetes and metabolic syndrome, which are closely related to obesity, may also be associated with FTO gene polymorphisms. Conclusion FTO gene polymorphism appears to play a role in the occurrence of NCDs. Some of the study results may be misleading due to ethnic differences and the effect of other genetic factors on disease onset, which needs to be investigated further. Finally, FTO gene polymorphisms can be studied as a preventive or therapeutic target. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-022-01139-4.
Collapse
Affiliation(s)
- Zahra Hoseini Tavassol
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Mousavi
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Molaei
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bandarian
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institutes, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Kazem Khalagi
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Ghannadi
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Hasani-Ranjbar
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Dapas M, Dunaif A. Deconstructing a Syndrome: Genomic Insights Into PCOS Causal Mechanisms and Classification. Endocr Rev 2022; 43:927-965. [PMID: 35026001 PMCID: PMC9695127 DOI: 10.1210/endrev/bnac001] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Indexed: 01/16/2023]
Abstract
Polycystic ovary syndrome (PCOS) is among the most common disorders in women of reproductive age, affecting up to 15% worldwide, depending on the diagnostic criteria. PCOS is characterized by a constellation of interrelated reproductive abnormalities, including disordered gonadotropin secretion, increased androgen production, chronic anovulation, and polycystic ovarian morphology. It is frequently associated with insulin resistance and obesity. These reproductive and metabolic derangements cause major morbidities across the lifespan, including anovulatory infertility and type 2 diabetes (T2D). Despite decades of investigative effort, the etiology of PCOS remains unknown. Familial clustering of PCOS cases has indicated a genetic contribution to PCOS. There are rare Mendelian forms of PCOS associated with extreme phenotypes, but PCOS typically follows a non-Mendelian pattern of inheritance consistent with a complex genetic architecture, analogous to T2D and obesity, that reflects the interaction of susceptibility genes and environmental factors. Genomic studies of PCOS have provided important insights into disease pathways and have indicated that current diagnostic criteria do not capture underlying differences in biology associated with different forms of PCOS. We provide a state-of-the-science review of genetic analyses of PCOS, including an overview of genomic methodologies aimed at a general audience of non-geneticists and clinicians. Applications in PCOS will be discussed, including strengths and limitations of each study. The contributions of environmental factors, including developmental origins, will be reviewed. Insights into the pathogenesis and genetic architecture of PCOS will be summarized. Future directions for PCOS genetic studies will be outlined.
Collapse
Affiliation(s)
- Matthew Dapas
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Andrea Dunaif
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
13
|
Wang Z, Zhang M, Seery S, Zheng G, Wang W, Zhao Y, Wang X, Zhang Y. Construction and validation of an m6A RNA methylation regulator prognostic model for early-stage clear cell renal cell carcinoma. Oncol Lett 2022; 24:250. [PMID: 35761938 PMCID: PMC9214704 DOI: 10.3892/ol.2022.13370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/27/2022] [Indexed: 11/18/2022] Open
Abstract
N6-methyladenosine (m6A) is the most common type of RNA methylation and is considered to participate in various biological and pathological processes, specifically in the regulation of tumorigenesis and metastasis. However, the exact prognostic role of m6A methylation regulators in early-stage clear cell renal cell carcinoma (ccRCC) is currently unknown. In the present study, a prognostic model consisting of m6A RNA methylation regulators in early stage ccRCC was constructed and the reliability of the signature was assessed by proteomics and immunohistochemistry. Additionally, the relationship between the prognostic model and tumor infiltrating immune cells within the tumor microenvironment was investigated. Gene mutation and RNA sequencing data of 19 m6A methylation regulators for early-stage ccRCC patients were extracted from The Cancer Genome Atlas (TCGA) database with the corresponding clinical information. Univariate and multivariate Cox regression analysis were applied to construct a prognostic model and the proteomic data as well as immunohistochemistry were used to validate the result. The correlations between the prognostic model and tumor infiltrating immune cells were assessed using Spearman's rank correlation analysis. A total of 192 early stage ccRCC gene mutation data as well as 261 RNA sequencing data with relative clinical data were extracted from the TCGA. The overall mutation frequency of the 19 m6A RNA methylation regulators was relatively low with 4.69%. The transcriptome data revealed that 11 genes were differentially expressed between cancer tissues and relatively normal tissues. Survival analysis highlighted four specific genes as having a significant influence on overall survival. An established model with four genes demonstrated the best predictability for early-stage ccRCC. After integrating clinical characteristics into the multivariate analysis, the model remained effective at predicting ccRCC prognosis. Spearman's rank analysis suggested several tumor infiltrating immune cells such as dendric cells, CD4+ cells, CD8+ T cells and macrophages were significantly correlated with the model. Proteomic data analysis as well as immunohistochemistry from the Human Protein Atlas showed that all the genes used to construct the model were differentially expressed between ccRCC and normal tissues. In conclusion, a novel m6A methylation regulators-based prognostic signature was established and validated with proteomics and immunohistochemistry. In addition, the model was significantly correlated with multiple infiltrating immune cells in tumor microenvironment.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Mingxin Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Samuel Seery
- School of Humanities and Social Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China.,Faculty of Health and Medicine, Division of Health Research, Lancaster University, Lancaster LA1 4YW, United Kingdom
| | - Guoyang Zheng
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Wenda Wang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yang Zhao
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Xu Wang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yushi Zhang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
14
|
Hu L, Ma L, Xia X, Ying T, Zhou M, Zou S, Yu H, Yin J. Efficacy of Bariatric Surgery in the Treatment of Women With Obesity and Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2022; 107:e3217-e3229. [PMID: 35554540 PMCID: PMC9282367 DOI: 10.1210/clinem/dgac294] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Indexed: 01/31/2023]
Abstract
CONTEXT The comparative effectiveness of drugs and surgical therapy for women with obesity and polycystic ovary syndrome (PCOS) has not been systematically compared. OBJECTIVE We aimed to determine the difference in efficacy between drug and bariatric surgery therapy for women with obesity and PCOS. METHODS This prospective nonrandomized trial enrolled 90 women aged 18 to 40 years with body mass index (BMI) ≥ 27.5 kg/m2 and waist circumference ≥ 85 cm and fulfilling the 2011 Chinese diagnostic criteria for PCOS; 81 subjects completed the study. In the drug group, patients were administered metformin and an oral contraceptive containing ethinyl-estradiol and cyproterone acetate for the first 6 months, and metformin alone for the second 6 months. In the surgical group, patients underwent laparoscopic sleeve gastrectomies. The follow-up period was 12 months. The main outcome was the complete remission of PCOS, requiring 6 consecutive regular menstruation cycles or spontaneous pregnancy. RESULTS Median BMI at endpoint was 30.1 kg/m2 in the drug group and 23.7 kg/m2 in the surgical group; complete remission rate was 15% and 78%, respectively. Except endpoint BMI, no difference was observed in free androgen index, ovarian morphology, homeostasis model assessment for insulin resistance, and total weight loss between remission and nonremission patients. Logistic regression analyses also revealed that the final BMI was the major factor influencing the remission of PCOS. The cutoff points for the final BMI were 27.5 kg/m2 for the drug group and 26 kg/m2 for the surgical group. Overall, nearly 95% of patients with an endpoint BMI below the cutoff values achieved complete remission. CONCLUSION Complete remission of PCOS in patients with obesity depends on the final BMI after weight loss. Thus, bariatric surgery should be prioritized for these patients.
Collapse
Affiliation(s)
| | | | | | - Tao Ying
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Minzhi Zhou
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Shuhua Zou
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Haoyong Yu
- Haoyong Yu, M.D., Department of Endocrinology and Metabolism, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, #600 Yishan Road, Shanghai 200233, China.
| | - Jun Yin
- Correspondence: Jun Yin, M.D., Ph.D., Department of Endocrinology and Metabolism, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People’s Hospital, #600 Yishan Road, Shanghai 200233, China.
| |
Collapse
|
15
|
Sun X, Lu J, Li H, Huang B. The Role of m 6A on Female Reproduction and Fertility: From Gonad Development to Ovarian Aging. Front Cell Dev Biol 2022; 10:884295. [PMID: 35712673 PMCID: PMC9197073 DOI: 10.3389/fcell.2022.884295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/21/2022] [Indexed: 11/20/2022] Open
Abstract
The growth and maturation of oocyte is accompanied by the accumulation of abundant RNAs and posttranscriptional regulation. N6-methyladenosine (m6A) is the most prevalent epigenetic modification in mRNA, and precisely regulates the RNA metabolism as well as gene expression in diverse physiological processes. Recent studies showed that m6A modification and regulators were essential for the process of ovarian development and its aberrant manifestation could result in ovarian aging. Moreover, the specific deficiency of m6A regulators caused oocyte maturation disorder and female infertility with defective meiotic initiation, subsequently the oocyte failed to undergo germinal vesicle breakdown and consequently lost the ability to resume meiosis by disrupting spindle organization as well as chromosome alignment. Accumulating evidence showed that dysregulated m6A modification contributed to ovarian diseases including polycystic ovarian syndrome (PCOS), primary ovarian insufficiency (POI), ovarian aging and other ovarian function disorders. However, the complex and subtle mechanism of m6A modification involved in female reproduction and fertility is still unknown. In this review, we have summarized the current findings of the RNA m6A modification and its regulators in ovarian life cycle and female ovarian diseases. And we also discussed the role and potential clinical application of the RNA m6A modification in promoting oocyte maturation and delaying the reproduction aging.
Collapse
Affiliation(s)
- Xiaoyan Sun
- State Key Laboratory of Reproductive Medicine, Gusu School, Suzhou Municipal Hospital, Suzhou Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
| | - Jiafeng Lu
- State Key Laboratory of Reproductive Medicine, Gusu School, Suzhou Municipal Hospital, Suzhou Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
| | - Hong Li
- State Key Laboratory of Reproductive Medicine, Gusu School, Suzhou Municipal Hospital, Suzhou Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
| | - Boxian Huang
- State Key Laboratory of Reproductive Medicine, Gusu School, Suzhou Municipal Hospital, Suzhou Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
| |
Collapse
|
16
|
Alnafjan AA, Alkhuriji AF, Alobaid HM, Babay ZA, Khalil MI. Association of FTO gene variants rs9939609 and rs1421085 with polycystic ovary syndrome. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00263-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Polycystic ovary syndrome (PCOS) is among the most common complex genetic endocrinopathy, and its etiology and pathophysiology remain controversial. FTO is a large highly polymorphic gene and was coined as the first locus associated with adiposity. The association of the intronic variant FTO rs9939609 or FTO rs1421085 with PCOS has been controversial and unclear, mainly due to ethnic differences among populations. The present study aims to investigate the association of FTO rs9939609 or FTO rs1421085 polymorphisms with PCOS in Saudi Arabian women.
Results
A total of 98 PCOS patients and 99 healthy females were included in this study. PCR and genotyping (TaqMan®SNP Genotyping Assay) were employed. For FTO rs9939609, the genotype TA and the recessive model (TA + AA) in PCOS patients were significantly different compared with control subjects (p = 0.008 and p = 0.007, respectively). The allele frequency of the FTO rs9939609 gene variant was associated significantly (p = 0.027) with PCOS, suggesting that the A allele is a risk factor for PCOS susceptibility. However, for the FTO rs1421085 variant, the genotype and allele distributions did not differ significantly between PCOS patients and controls (p > 0.05).
Conclusions
This is the first report to study the association of FTO rs9939609 and FTO rs1421085 with PCOS in Saudi women. Results suggest that the FTO rs9939609 gene variant could be a genetic predisposing factor for PCOS Saudi women.
Collapse
|
17
|
Nautiyal H, Imam SS, Alshehri S, Ghoneim MM, Afzal M, Alzarea SI, Güven E, Al-Abbasi FA, Kazmi I. Polycystic Ovarian Syndrome: A Complex Disease with a Genetics Approach. Biomedicines 2022; 10:biomedicines10030540. [PMID: 35327342 PMCID: PMC8945152 DOI: 10.3390/biomedicines10030540] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/13/2022] [Accepted: 02/16/2022] [Indexed: 01/27/2023] Open
Abstract
Polycystic ovarian syndrome (PCOS) is a complex endocrine disorder affecting females in their reproductive age. The early diagnosis of PCOS is complicated and complex due to overlapping symptoms of this disease. The most accepted diagnostic approach today is the Rotterdam Consensus (2003), which supports the positive diagnosis of PCOS when patients present two out of the following three symptoms: biochemical and clinical signs of hyperandrogenism, oligo, and anovulation, also polycystic ovarian morphology on sonography. Genetic variance, epigenetic changes, and disturbed lifestyle lead to the development of pathophysiological disturbances, which include hyperandrogenism, insulin resistance, and chronic inflammation in PCOS females. At the molecular level, different proteins and molecular and signaling pathways are involved in disease progression, which leads to the failure of a single genetic diagnostic approach. The genetic approach to elucidate the mechanism of pathogenesis of PCOS was recently developed, whereby four phenotypic variances of PCOS categorize PCOS patients into classic, ovulatory, and non-hyperandrogenic types. Genetic studies help to identify the root cause for the development of this PCOS. PCOS genetic inheritance is autosomal dominant but the latest investigations revealed it as a multigene origin disease. Different genetic loci and specific genes have been identified so far as being associated with this disease. Genome-wide association studies (GWAS) and related genetic studies have changed the scenario for the diagnosis and treatment of this reproductive and metabolic condition known as PCOS. This review article briefly discusses different genes associated directly or indirectly with disease development and progression.
Collapse
Affiliation(s)
- Himani Nautiyal
- Siddhartha Institute of Pharmacy, Near IT-Park, Sahastradhara Road, Dehradun 248001, India;
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.S.I.); (S.A.)
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.S.I.); (S.A.)
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Muhammad Afzal
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia;
- Correspondence: (M.A.); (I.K.)
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia;
| | - Emine Güven
- Biomedical Engineering Department, Faculty of Engineering, Düzce University, Düzce 81620, Turkey;
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Correspondence: (M.A.); (I.K.)
| |
Collapse
|
18
|
Tyrmi JS, Arffman RK, Pujol-Gualdo N, Kurra V, Morin-Papunen L, Sliz E, Piltonen TT, Laisk T, Kettunen J, Laivuori H. Leveraging Northern European population history: novel low-frequency variants for polycystic ovary syndrome. Hum Reprod 2022; 37:352-365. [PMID: 34791234 PMCID: PMC8804330 DOI: 10.1093/humrep/deab250] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/07/2021] [Indexed: 12/21/2022] Open
Abstract
STUDY QUESTION Can we identify novel variants associated with polycystic ovary syndrome (PCOS) by leveraging the unique population history of Northern Europe? SUMMARY ANSWER We identified three novel genome-wide significant associations with PCOS, with two putative independent causal variants in the checkpoint kinase 2 (CHEK2) gene and a third in myosin X (MYO10). WHAT IS KNOWN ALREADY PCOS is a common, complex disorder with unknown aetiology. While previous genome-wide association studies (GWAS) have mapped several loci associated with PCOS, the analysis of populations with unique population history and genetic makeup has the potential to uncover new low-frequency variants with larger effects. STUDY DESIGN, SIZE, DURATION A population-based case-control GWAS was carried out. PARTICIPANTS/MATERIALS, SETTING, METHODS We identified PCOS cases from national registers by ICD codes (ICD-10 E28.2, ICD-9 256.4, or ICD-8 256.90), and all remaining women were considered controls. We then conducted a three-stage case-control GWAS: in the discovery phase, we had a total of 797 cases and 140 558 controls from the FinnGen study. For validation, we used an independent dataset from the Estonian Biobank, including 2812 cases and 89 230 controls. Finally, we performed a joint meta-analysis of 3609 cases and 229 788 controls from both cohorts. Additionally, we reran the association analyses including BMI as a covariate, with 2169 cases and 160 321 controls from both cohorts. MAIN RESULTS AND THE ROLE OF CHANCE Two out of the three novel genome-wide significant variants associating with PCOS, rs145598156 (P = 3.6×10-8, odds ratio (OR) = 3.01 [2.02-4.50] minor allele frequency (MAF) = 0.005) and rs182075939 (P = 1.9×10-16, OR = 1.69 [1.49-1.91], MAF = 0.04), were found to be enriched in the Finnish and Estonian populations and are tightly linked to a deletion c.1100delC (r2 = 0.95) and a missense I157T (r2 = 0.83) in CHEK2. The third novel association is a common variant near MYO10 (rs9312937, P = 1.7 × 10-8, OR = 1.16 [1.10-1.23], MAF = 0.44). We also replicated four previous reported associations near the genes Erb-B2 Receptor Tyrosine Kinase 4 (ERBB4), DENN Domain Containing 1A (DENND1A), FSH Subunit Beta (FSHB) and Zinc Finger And BTB Domain Containing 16 (ZBTB16). When adding BMI as a covariate only one of the novel variants remained genome-wide significant in the meta-analysis (the EstBB lead signal in CHEK2 rs182075939, P = 1.9×10-16, OR = 1.74 [1.5-2.01]) possibly owing to reduced sample size. LARGE SCALE DATA The age- and BMI-adjusted GWAS meta-analysis summary statistics are available for download from the GWAS Catalog with accession numbers GCST90044902 and GCST90044903. LIMITATIONS, REASONS FOR CAUTION The main limitation was the low prevalence of PCOS in registers; however, the ones with the diagnosis most likely represent the most severe cases. Also, BMI data were not available for all (63% for FinnGen, 76% for EstBB), and the biobank setting limited the accessibility of PCOS phenotypes and laboratory values. WIDER IMPLICATIONS OF THE FINDINGS This study encourages the use of isolated populations to perform genetic association studies for the identification of rare variants contributing to the genetic landscape of complex diseases such as PCOS. STUDY FUNDING/COMPETING INTEREST(S) This work has received funding from the European Union's Horizon 2020 research and innovation programme under the MATER Marie Skłodowska-Curie grant agreement No. 813707 (N.P.-G., T.L., T.P.), the Estonian Research Council grant (PRG687, T.L.), the Academy of Finland grants 315921 (T.P.), 321763 (T.P.), 297338 (J.K.), 307247 (J.K.), 344695 (H.L.), Novo Nordisk Foundation grant NNF17OC0026062 (J.K.), the Sigrid Juselius Foundation project grants (T.L., J.K., T.P.), Finska Läkaresällskapet (H.L.) and Jane and Aatos Erkko Foundation (H.L.). The funders had no role in study design, data collection and analysis, publishing or preparation of the manuscript. The authors declare no conflicts of interest.
Collapse
Affiliation(s)
- Jaakko S Tyrmi
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Riikka K Arffman
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Natàlia Pujol-Gualdo
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Venla Kurra
- Department of Clinical Genetics, Faculty of Medicine and Health Technology, Tampere University Hospital and Tampere University, Tampere, Finland
| | - Laure Morin-Papunen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Eeva Sliz
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Johannes Kettunen
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Hannele Laivuori
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Technology, Tampere University Hospital and Tampere University, Tampere, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
19
|
Association between melatonin receptor gene polymorphisms and polycystic ovarian syndrome: a systematic review and meta-analysis. Biosci Rep 2021; 40:225044. [PMID: 32463080 PMCID: PMC7317604 DOI: 10.1042/bsr20200824] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/21/2020] [Accepted: 05/27/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Polycystic ovarian syndrome (PCOS) is a kind of common gynecological endocrine disorder. And the mutations of melatonin receptor (MTNR) genes are related to the occurrence of PCOS. But previous researches have shown opposite results. So, the object of our systematic review and meta-analysis is to investigate the relationship between MTNR 1A/B polymorphisms and PCOS. METHODS PubMed, Embase, Ovid, the Cochrane Library, Web of Science and three Chinese databases (VIP, CNKI and Wanfang) were used to retrieve eligible articles published between January 1980 and February 2020. And we used the odds ratio (OR) and its 95% confidence interval (CI) to investigate the strength of the association by six genetic models, allelic, codominant (homozygous and heterozygous), dominant, recessive and superdominant models. Review Manager 5.3, IBM SPSS statistics 25 and Stata MP 16.0 software were used to do this meta-analysis. RESULTS Our meta-analysis involved 2553 PCOS patients and 3152 controls, for two single nucleotide polymorphisms (rs10830963 C> G in MTNR1B and rs2119882 T> C in MTNR1A) and significant associations were found in some genetic models of these single nucleotide polymorphisms (SNPs). For rs10830963, strongly significant was found in the heterozygote model (GC vs. CC, P=0.02). Additionally, a slight trend was detected in the allelic (G vs. C), homozygote (GG vs. CC) and dominant (GG+GC vs. CC) model of rs10830963 (P=0.05). And after further sensitivity analysis, a study with high heterogeneity was removed. In the allelic (P=0.000), homozygote (P=0.001), dominant (P=0.000) and recessive (GG vs. GC+CC, P=0.001) model, strong associations between rs10830963 and PCOS were found. Moreover, for rs2119882, five genetic models, allelic (C vs. T, P=0.000), codominant (the homozygote (CC vs. TT, P=0.000) and heterozygote model (CT vs. TT, P=0.02), dominant (CC + CT vs. TT, P=0.03) and recessive model (CC vs. CT + TT, P=0.000) showed significant statistical associations with PCOS. CONCLUSION MTNR1B rs10830963 and MTNR1B rs2119882 polymorphisms are associated with PCOS risk. However, the above conclusions still require being confirmed by much larger multi-ethnic studies.
Collapse
|
20
|
Manti M, Stener-Victorin E, Benrick A. Skeletal Muscle Immunometabolism in Women With Polycystic Ovary Syndrome: A Meta-Analysis. Front Physiol 2020; 11:573505. [PMID: 33192572 PMCID: PMC7642984 DOI: 10.3389/fphys.2020.573505] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/25/2020] [Indexed: 12/25/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine and metabolic disorder affecting up to 15% of women at reproductive age. The main features of PCOS are hyperandrogenism and irregular menstrual cycles together with metabolic dysfunctions including hyperinsulinemia and insulin resistance and a 4-fold increased risk of developing type 2 diabetes. Despite the high prevalence the pathophysiology of the syndrome is unclear. Insulin resistance in women with PCOS likely affect the skeletal muscle and recently it was demonstrated that changes in DNA methylation affects the gene expression in skeletal muscle that in part can explain their metabolic abnormalities. The objective of this work was to combine gene expression array data from different datasets to improve statistical power and thereby identify novel biomarkers that can be further explored. In this narrative review, we performed a meta-analysis of skeletal muscle arrays available from Gene Expression Omnibus and from publications. The eligibility criteria were published articles in English, and baseline (no treatment) skeletal muscle samples from women with PCOS and controls. The R package Metafor was used for integration of the datasets. One hundred and fourteen unique transcripts were differentially expressed in skeletal muscle from women with PCOS vs. controls (q < 0.05), 87% of these transcripts have not been previously identified as altered in PCOS muscle. ING2, CDKAL1, and AKTIP had the largest differential increase in expression, and TSHZ2, FKBP2, and OCEL1 had the largest decrease in expression. Two genes, IRX3 and CDKAL1 were consistently upregulated (q < 0.05) in the individual analyses and meta-analysis. Based on the meta-analysis, we identified several dysregulated immunometabolic pathways as a part of the molecular mechanisms of insulin resistance in the skeletal muscle of women with PCOS. The transcriptomic data need to be verified by functional analyses as well as proteomics to advance our understanding of PCOS specific insulin resistance in skeletal muscle.
Collapse
Affiliation(s)
- Maria Manti
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Anna Benrick
- Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,School of Health Sciences, University of Skövde, Skövde, Sweden
| |
Collapse
|
21
|
Metastatin as a Marker for Hyperandrogenemia in Iraqi Women with Polycystic Ovary Syndrome. Obstet Gynecol Int 2020; 2020:5216903. [PMID: 33061987 PMCID: PMC7533004 DOI: 10.1155/2020/5216903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/03/2020] [Accepted: 09/13/2020] [Indexed: 11/30/2022] Open
Abstract
Objective Kisspeptin 1 might reflect increased androgen level in polycystic ovarian syndrome instead of other markers. Study Design. A case control study was performed in Al-Yarmouk Teaching Hospital from 1st of July 2016 to 1st of July 2017; it involved 87 women divided into two groups: 44 women diagnosed as PCOS, 22 women with BMI ≥ 25 kg/m2 and 22 women with BMI < 25 kg/m2, and another 43 women without PCOS, 22 women with BMI ≥ 25 kg/m2 and 21 women with BMI < 25 kg/m2. Hormonal, metabolic profiles, and hirsutism scores, as well as serum kisspeptin level, were assessed by using Human Kisspeptin 1(KISS-1) ELISA Kit. The blood samples between days 2 and 5 of menstrual cycle were drawn by an disposable sterile syringe and collected in EDTA containing tubes (as anticoagulant), and the hormonal profile was measured using a biotech ELISA reader. Result Serum level of kisspeptin was significantly higher in PCOS compared to control (322.4 vs. 235.3 ng/L, respectively). There was no significant difference in age, BMI, and parity between control and PCOS; the frequency of hirsutism, acne, elevated LH, and increased free testosterone (fTT) were significantly higher in PCOS compared to control. Kisspeptin shows a direct significant correlation with hirsutism and fTT (r = 0.648, 0.238, respectively). In ROC analysis, kisspeptin had AUC (95% CI) = 0.874 (0.785–0.935) for predicting PCOS. Conclusion Kisspeptin levels might be used as a marker for hyperandrogenemia in polycystic ovarian syndrome.
Collapse
|
22
|
Quantile-dependent heritability of computed tomography, dual-energy x-ray absorptiometry, anthropometric, and bioelectrical measures of adiposity. Int J Obes (Lond) 2020; 44:2101-2112. [PMID: 32665611 PMCID: PMC7530941 DOI: 10.1038/s41366-020-0636-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 06/07/2020] [Accepted: 07/03/2020] [Indexed: 12/18/2022]
Abstract
Background/Objectives: Quantile-dependent expressivity occurs when a gene’s
phenotypic expression depends upon whether the trait (e.g., BMI) is high or
low relative to its distribution. We have previously shown that the obesity
effects of a genetic risk score (GRSBMI) increased significantly
with increasing quantiles of BMI. However, BMI is an inexact adiposity
measure and GRSBMI explains <3% of the BMI variance. The
purpose of this paper is to test BMI for quantile-dependent expressivity
using a more inclusive genetic measure
(h2, heritability in
the narrow sense), extend the result to other adiposity measures, and
demonstrate its consistency with purported gene-environment
interactions. Subjects/Methods: Quantile-specific offspring-parent regression slopes
(βOP) were obtained from quantile regression for
height (ht) and computed tomography (CT), dual-energy x-ray absorptiometry
(DXA), anthropometric, and bioelectrical impedance (BIA) adiposity measures.
Heritability was estimated by 2βOP/(1+rspouse)
in 6,227 offspring-parent pairs from the Framingham Heart Study, where
rspouse is the spouse correlation. Results: Compared to h2 at the
10th percentile, genetic heritability was significantly
greater at the 90th population percentile for BMI (3.14-fold
greater, P<10−15), waist girth/ht (3.27-fold,
P<10−15), hip girth/ht (3.12-fold,
P=6.3×10−14), waist-to-hip ratio (1.75-fold,
P=0.01), sagittal diameter/ht (3.89-fold,
P=3.7×10−7), DXA total fat/ht2
(3.62-fold, P=0.0002), DXA leg fat/ht2 (3.29-fold,
P=2.0×10−11), DXA arm fat/ht2
(4.02-fold, P=0.001), CT-visceral fat/ht2 (3.03-fold, P=0.002),
and CT-subcutaneous fat/ht2 (3.54-fold, P=0.0004). External
validity was suggested by the phenomenon’s consistency with numerous
published reports. Quantile-dependent expressivity potentially explains
precision medicine markers for weight gain from overfeeding or antipsychotic
medications, and the modifying effects of physical activity, sleep, diet,
polycystic ovary syndrome, socioeconomic status, and depression on gene-BMI
relationships. Conclusion: Genetic heritabilities of anthropometric, CT, and DXA adiposity
measures increase with increasing adiposity. Some gene-environment
interactions may arise from analyzing subjects by characteristics that
distinguish high vs. low adiposity rather than the effects of environmental
stimuli on transcriptional and epigenetic processes.
Collapse
|
23
|
Zhao Y, Xu Y, Wang X, Xu L, Chen J, Gao C, Wu C, Pan D, Zhang Q, Zhou J, Chen R, Wang Z, Zhao H, You L, Cao Y, Li Z, Shi Y. Body Mass Index and Polycystic Ovary Syndrome: A 2-Sample Bidirectional Mendelian Randomization Study. J Clin Endocrinol Metab 2020; 105:5804107. [PMID: 32163573 DOI: 10.1210/clinem/dgaa125] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/06/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Observational studies have shown a link between elevated body mass index (BMI) and the risk of polycystic ovary syndrome (PCOS). While Mendelian randomization (MR) studies in Europeans have suggested a causal role of increased BMI in PCOS, whether the same role is suggested in Asians has yet to be investigated. We used MR studies to infer causal effects using genetic data from East Asian populations. METHODS AND FINDINGS We performed a 2-sample bidirectional MR analysis using summary statistics from genome-wide association studies (GWAS) of BMI (with up to 173 430 individuals) and PCOS (4386 cases and 8017 controls) in East Asian populations. Seventy-eight single nucleotide polymorphisms (SNPs) correlated with BMI were selected as genetic instrumental variables to estimate the causal effect of BMI on PCOS using the inverse-variance weighted (IVW) method. To test the reliability of the results, further sensitivity analyses included MR-Egger regression, weighted median estimates, and leave-one-out analysis. The IVW analysis indicated a significant association between high BMI and the risk of PCOS (odds ratio per standard deviation higher BMI, 2.208; 95% confidence interval 1.537 to 3.168, P = 1.77 × 10-5). In contrast, the genetic risk of PCOS had no significant effect on BMI. CONCLUSIONS The results of our bidirectional MR study showed that an increase in BMI causes PCOS, while PCOS does not cause an increased BMI. This study provides further genetic support for a link between BMI and PCOS. Further research is needed to interpret the potential mechanisms of this association.
Collapse
Affiliation(s)
- Yalin Zhao
- School of Basic Medicine, Qingdao University, China
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Yuping Xu
- School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, Anhui, China
| | - Xiaomeng Wang
- Department of Obstetrics, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lin Xu
- Department of Obstetrics, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianhua Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Chengwen Gao
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Chuanhong Wu
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Dun Pan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Zhang
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Juan Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Ruirui Chen
- School of Basic Medicine, Qingdao University, China
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
| | - Zhuo Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Han Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, China
| | - Li You
- Center for Reproductive Medicine, Shandong University, Jinan, China
| | - Yunxia Cao
- School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, Anhui, China
| | - Zhiqiang Li
- School of Basic Medicine, Qingdao University, China
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
- Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongyong Shi
- School of Basic Medicine, Qingdao University, China
- The Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
- Institute of Social Cognitive and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, China
- Institute of Neuropsychiatric Science and Systems Biological Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
24
|
Douma Z, Dallel M, Bahia W, Ben Salem A, Hachani Ben Ali F, Almawi WY, Lautier C, Haydar S, Grigorescu F, Mahjoub T. Association of estrogen receptor gene variants (ESR1 and ESR2) with polycystic ovary syndrome in Tunisia. Gene 2020; 741:144560. [PMID: 32169631 DOI: 10.1016/j.gene.2020.144560] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/08/2020] [Indexed: 02/07/2023]
Abstract
SNV (single nucleotide variation) in estrogen receptor (ESR1 and ESR2) genes are susceptibility markers for complex diseases, such as cancer, metabolic disorders and women infertility. We explored six widely used SNVs in ESR1 (rs2234693, rs9340799, rs3798577, rs3020314) and ESR2 (rs1256049, rs4986938) in polycystic ovary syndrome (PCOS) in women from Tunisia (n = 254) compared to controls (n = 170). Genotyping was performed by RFLP-PCR or real-time PCR and analyzed in GoldenHelix statistical package. Logistic regression revealed association of rs2234693, rs3798577 and rs3020314 (ESR1) and rs1256049 (ESR2), the association of rs2234693 (C/T) being the strongest with P < 4.81 × 10-6, 2.88 × 10-5 after Bonferroni correction, OR 0.31, 95%CI (0.18-0.53)). Correlations were found with LH, LH/FSH or hyperandrogenism and even more significant with metabolic syndrome (rs9340799) and hyperglycemia (rs3798577). Among 14 haplotypes reconstructed in ESR1gene, four haplotypes (H1 to H4) were associated with PCOS the strongest being that of H1 (P < 0.002) supported by Bonferroni (P < 0.033) and permutation tests (P < 4 x10-4). In haplotype trend regression, concordant correlations were found with insulin resistance (P < 0.033) for H2 and with high blood pressure for H3 (P < 0.048). While these data revealed influential role on metabolic rather and hormonal features of PCOS, the association of rs2234693 was the strongest among all ethnic populations studied thus far giving a new insight on estrogen receptor gene variation in distant North African populations and their role in metabolic alteration of PCOS.
Collapse
Affiliation(s)
- Zeineb Douma
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Meriem Dallel
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Weal Bahia
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Assila Ben Salem
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | | | - Wassim Y Almawi
- School of Medicine, Nazarbayev University, Astana, Kazakhstan and Faculty of Sciences, El-Manar University, Tunis, Tunisia
| | - Corinne Lautier
- University of Montpellier, UMR204 NUTRIPASS (IRD, UM, SupAgro), Montpellier, France
| | - Sara Haydar
- University of Montpellier, UMR204 NUTRIPASS (IRD, UM, SupAgro), Montpellier, France
| | - Florin Grigorescu
- University of Montpellier, UMR204 NUTRIPASS (IRD, UM, SupAgro), Montpellier, France; Institut Convergences Migrations, Collège de France, Paris, France.
| | - Touhemi Mahjoub
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| |
Collapse
|
25
|
Khan MJ, Ullah A, Basit S. Genetic Basis of Polycystic Ovary Syndrome (PCOS): Current Perspectives. Appl Clin Genet 2019; 12:249-260. [PMID: 31920361 PMCID: PMC6935309 DOI: 10.2147/tacg.s200341] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/05/2019] [Indexed: 12/18/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common infertility disorder affecting a significant proportion of the global population. It is the main cause of anovulatory infertility in women and is the most common endocrinopathy affecting reproductive-aged women, with a prevalence of 8-13% depending on the criteria used and population studied. The disease is multifactorial and complex and, therefore, often difficult to diagnose due to overlapping symptoms. Multiple etiological factors have been implicated in PCOS. Due to the complex pathophysiology involving multiple pathways and proteins, single genetic diagnostic tests cannot be determined. Progress has been achieved in the management and diagnosis of PCOS; however, not much is known about the molecular players and signaling pathways underlying it. Conclusively PCOS is a polygenic and multifactorial syndromic disorder. Many genes have been associated with PCOS, which affect fertility either directly or indirectly. However, studies conducted on PCOS patients from multiple families failed to find a fully penetrant variant(s). The present study was designed to review the current genetic understanding of the disease. In the present review, we have discussed the clinical spectrum, the genetics, and the variants identified as being associated with PCOS. The mechanisms by which variants in the genes confer risk to PCOS and the nature of the physical and genetic interaction between the genetic elements underlying PCOS remain to be determined. Elucidation of genetic players and cellular pathways underlying PCOS will certainly increase our understanding of the pathophysiology of this syndrome. The study also discusses the current status of the treatment modalities for PCOS, which is important to find new ways of treatment.
Collapse
Affiliation(s)
- Muhammad Jaseem Khan
- Institute of Paramedical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Anwar Ullah
- Institute of Paramedical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Sulman Basit
- Center for Genetics and Inherited Diseases, Taibah University Almadinah Almunawwarrah, Peshawar, Saudi Arabia
| |
Collapse
|
26
|
Meta-analysis of association of FTO genetic variation with PCOS must account for obesity. Genomics 2019; 112:2164-2165. [PMID: 31857220 DOI: 10.1016/j.ygeno.2019.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/12/2019] [Indexed: 01/01/2023]
|
27
|
Glueck CJ, Goldenberg N. Characteristics of obesity in polycystic ovary syndrome: Etiology, treatment, and genetics. Metabolism 2019; 92:108-120. [PMID: 30445140 DOI: 10.1016/j.metabol.2018.11.002] [Citation(s) in RCA: 248] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/22/2018] [Accepted: 11/05/2018] [Indexed: 01/01/2023]
Abstract
Polycystic ovary syndrome (PCOS) has multiple etiologies including ovarian and adrenal hyperandrogenism, neuro-endocrine and hypothalamic-pituitary dysfunction, and disorders of peripheral insulin resistance. Obesity is neither necessary nor sufficient for the PCOS phenotype, and the association of PCOS with obesity is not universal, with national, cultural, and ethnic differences. Obesity, particularly visceral adiposity which is common in obese and non-obese women with PCOS, amplifies and worsens all metabolic and reproductive outcomes in PCOS. Obesity increases insulin resistance and compensatory hyperinsulinemia, which in turn increases adipogenesis and decreases lipolysis. Obesity sensitizes thecal cells to LH stimulation and amplifies functional ovarian hyperandrogenism by upregulating ovarian androgen production. Obesity increases inflammatory adipokines which, in turn, increase insulin resistance and adipogenesis. Lifestyle interventions focused on diet-weight loss and concurrent exercise are central to therapy which also commonly subsequently needs to include pharmacologic therapy. PCOS symptoms commonly improve with 5% to 10% weight loss, but 25% to 50% weight loss, usually achievable only through bariatric surgery, may be required for morbid obesity unresponsive to lifestyle-medical treatment. Bariatric surgery is a valuable approach to weight loss in PCOS where BMI is ≥40 kg/m2 when non-surgical treatment and/or induction of pregnancy have failed, and can be an initial treatment when BMI is ≥50 kg/m2. Further research in PCOS is needed to better understand the fundamental basis of the disorder, to ameliorate obesity, to correct hyperandrogenism, ovulation, hyperinsulinemia, and to optimize metabolic homeostasis.
Collapse
Affiliation(s)
- Charles J Glueck
- The Cholesterol, Metabolism, and Thrombosis Research Center, 3906 Middleton Avenue, Cincinnati, OH 45220, United States of America.
| | - Naila Goldenberg
- The Cholesterol, Metabolism, and Thrombosis Research Center, 3906 Middleton Avenue, Cincinnati, OH 45220, United States of America
| |
Collapse
|
28
|
Brower MA, Hai Y, Jones MR, Guo X, Chen YDI, Rotter JI, Krauss RM, Legro RS, Azziz R, Goodarzi MO. Bidirectional Mendelian randomization to explore the causal relationships between body mass index and polycystic ovary syndrome. Hum Reprod 2019; 34:127-136. [PMID: 30496407 PMCID: PMC6295958 DOI: 10.1093/humrep/dey343] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 10/17/2018] [Accepted: 11/01/2018] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION What are the causal relationships between polycystic ovary syndrome (PCOS) and body mass index (BMI)? SUMMARY ANSWER Bidirectional Mendelian randomization analyses suggest that increased BMI is causal for PCOS while the reverse is not the case. WHAT IS KNOWN ALREADY The contribution of obesity to the pathogenesis of PCOS is controversial. To date, published genetic studies addressing this question have generated conflicting results and have not utilized the full extent of known single nucleotide polymorphisms associated with body mass index (BMI). STUDY DESIGN, SIZE, DURATION This cross-sectional Mendelian randomization (MR) and genetic association study was conducted in 750 individuals of European origin and with PCOS and 1567 BMI-matched controls. PARTICIPANTS/MATERIALS, SETTING, METHODS Cases and controls were matched for BMI as well as for distribution of weight categories (normal weight, overweight, obese). Two-sample MR using inverse variance weighting (IVW) was conducted using a 92-SNP instrument variable for BMI with PCOS as the outcome, followed by two-sample MR with a 16-SNP instrument variable for PCOS with BMI as the outcome. Sensitivity analyses included MR-Egger and maximum likelihood methods. Secondary analyses assessed associations of genetic risk scores and individual SNPs with PCOS, BMI and quantitative androgen-related and glucose homeostasis-related traits. MAIN RESULTS AND THE ROLE OF CHANCE Each standard deviation genetically higher BMI was associated with a 4.89 (95% CI 1.46-16.32) higher odds of PCOS. Conversely, genetic risk of PCOS did not influence BMI. Sensitivity analyses yielded directionally consistent results. The genetic risk score of 92 BMI SNPs was associated with the diagnosis of PCOS (OR 1.043, 95% CI 1.009-1.078, P = 0.012). Of the 92 BMI risk variants evaluated, none were associated individually with PCOS after considering multiple testing. The association of FTO SNP rs1421085 with BMI was stronger in women with PCOS (β = 0.071, P = 0.0006) than in controls (β = 0.046, P = 0.065). LIMITATIONS, REASONS FOR CAUTION The current sample size, while providing good power for MR and genetic risk score analyses, had limited power to demonstrate association of individual SNPs with PCOS. Cases and controls were not matched for age; however, this was mitigated by adjusting analyses for age. Dietary and lifestyle data, which could have been used to explore the greater association of the FTO SNP with BMI in women with PCOS, was not available. WIDER IMPLICATIONS OF THE FINDINGS Increasing BMI appears to be causal for PCOS but having PCOS does not appear to affect BMI. This study used the most comprehensive set of SNPs for BMI currently available. Prior studies using fewer SNPs had yielded conflicting results and may have been confounded because cases and controls were not matched for weight categories. The current results highlight the potential utility of weight management in the prevention and treatment of PCOS. STUDY FUNDING/COMPETING INTEREST(S) National Institutes of Health Grants R01-HD29364 and K24-HD01346 (to R.A.), Grant R01-DK79888 (to M.O.G.), Grant U54-HD034449 (to R.S.L.), Grant U19-HL069757 (to R.M.K.). The funders had no influence on the data collection, analyses or conclusions of the study. No conflict of interests to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- M A Brower
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Y Hai
- Department of Statistics, University of Auckland, Auckland, New Zealand
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - M R Jones
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - X Guo
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Y -D I Chen
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - J I Rotter
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - R M Krauss
- Children’s Hospital of Oakland Research Institute, Oakland, CA, USA
| | - R S Legro
- Department of Obstetrics and Gynecology, Pennsylvania State College of Medicine, Hershey, PA, USA
| | - R Azziz
- Departments of Obstetrics and Gynecology and Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - M O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
29
|
Heshmati J, Farsi F, Yosaee S, Razavi M, Rezaeinejad M, Karimie E, Sepidarkish M. The Effects of Probiotics or Synbiotics Supplementation in Women with Polycystic Ovarian Syndrome: a Systematic Review and Meta-Analysis of Randomized Clinical Trials. Probiotics Antimicrob Proteins 2018; 11:1236-1247. [DOI: 10.1007/s12602-018-9493-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
30
|
Jacewicz-Święcka M, Kowalska I. Polycystic ovary syndrome and the risk of cardiometabolic complications in longitudinal studies. Diabetes Metab Res Rev 2018; 34:e3054. [PMID: 30089337 DOI: 10.1002/dmrr.3054] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 06/18/2018] [Accepted: 07/29/2018] [Indexed: 12/11/2022]
Abstract
The purpose of this study was to perform a review of the longitudinal studies to determine whether polycystic ovary syndrome is associated with higher prevalence of metabolic complications and cardiovascular morbidity and mortality. The primary outcomes included body mass index, metabolic syndrome and its components (waist circumference, lipid profile, arterial hypertension, abnormal glucose metabolism (impaired fasting glucose, impaired glucose tolerance, type 2 diabetes), insulin resistance, and cardiovascular diseases like stroke, angina, and coronary heart disease. Complications in pregnant women were beyond the scope of this review. PubMed database (1992-2018) was searched to identify proper publications. Finally, data from 47 articles were analysed. Studies differed in the design (prospective, retrospective, cohort, observational), research methods, polycystic ovary syndrome diagnostic criteria, studied populations, race, and ethnicity of the participants. Based on the data collected, it appears that women with polycystic ovary syndrome have higher prevalence of obesity, abdominal fat distribution, dyslipidaemia and deterioration of glucose metabolism, but increased prevalence of cardiovascular diseases is not proven.
Collapse
Affiliation(s)
- Małgorzata Jacewicz-Święcka
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Irina Kowalska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
31
|
Goldrat O, Delbaere A. PCOS: update and diagnostic approach. Clin Biochem 2018; 62:24-31. [PMID: 30195483 DOI: 10.1016/j.clinbiochem.2018.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/26/2018] [Accepted: 09/04/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Oranite Goldrat
- Fertility Clinic, Department of Obstetrics and Gynecology, CUB-Hôpital Erasme, Université Libre de Bruxelles (ULB), Route de Lennik, 808, Brussels, Belgium.
| | - Anne Delbaere
- Fertility Clinic, Department of Obstetrics and Gynecology, CUB-Hôpital Erasme, Université Libre de Bruxelles (ULB), Route de Lennik, 808, Brussels, Belgium.
| |
Collapse
|
32
|
Atanasova Boshku A, Ivanova Panova D, Zafirova Ivanovska B. ADIPONECTIN AS A SERUM MARKER OF ADIPOSE TISSUE DYSFUNCTION IN WOMEN WITH POLYCYSTIC OVARY SYNDROME: CORRELATION WITH INDICATORS OF METABOLIC DISTURBANCES. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2018; 14:346-352. [PMID: 31149282 PMCID: PMC6525779 DOI: 10.4183/aeb.2018.346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Adipose tissue is a major store of energy for the human body. Polycystic ovary syndrome (PCOS) patients are more prone to abnormal production of some regulatory proteins secreted from the adipose tissue. This study aims to investigate serum levels of adiponectin and their correlation with metabolic and endocrine indices in PCOS. PATIENTS AND METHODS This study was conducted on 61 women with PCOS and 17 healthy women whose age and body mass index (BMI) were matched. Adiponectin serum levels were assessed and correlated with parameters of metabolic and hormonal disturbances. RESULTS In PCOS women, serum levels of insulin, HOMA-IR, testosterone, LH, and LH/FSH were significantly higher, while SHBG was lower than in healthy women. Lower adiponectin was observed in both PCOS groups compared to the control group. Serum levels of adiponectin correlated inversely with BMI (r=- 0.56; p<0.001),WC(r = -0.452;p<0.001), insulin levels (r= - 0.409; p<0.001), HOMA-IR (r= -0.368; p<0.001), and free androgen index (FAI) (r= - 0.53; p<0.001). A positive correlation was found between adiponectin and LH (r= 0.35; p<0.001), LH/FSH ratio (r= 0.33; p<0.001) and SHGB (r= 0.51; p<0.001). Serum adiponectin levels are decreased in women with PCOS compared to the control group. The decrease in adiponectin concentration indicates its potential role in metabolic disorders in the pathogenesis of PCOS, as well as in the development and progression of insulin resistance in PCOS patients.
Collapse
Affiliation(s)
- A. Atanasova Boshku
- University Clinic of Gynecology and Obstetrics, Medical Faculty, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - D. Ivanova Panova
- University Clinic of Gynecology and Obstetrics, Medical Faculty, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| | - B. Zafirova Ivanovska
- Institute of Epidemiology and Biostatistics with Medical Informatics, Medical Faculty, Ss Cyril and Methodius University of Skopje, Skopje, Republic of Macedonia
| |
Collapse
|
33
|
Daghestani MH. Evaluation of biochemical, endocrine, and metabolic biomarkers for the early diagnosis of polycystic ovary syndrome among non-obese Saudi women. Int J Gynaecol Obstet 2018; 142:162-169. [DOI: 10.1002/ijgo.12527] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 03/22/2018] [Accepted: 05/08/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Maha H. Daghestani
- Zoology Department; Science College; King Saud University; Riyadh Saudi Arabia
- Central Laboratory; Center for Female Scientific and Medical Colleges; King Saud University; Riyadh Saudi Arabia
| |
Collapse
|
34
|
Albalawi FS, Daghestani MH, Daghestani MH, Eldali A, Warsy AS. rs4889 polymorphism in KISS1 gene, its effect on polycystic ovary syndrome development and anthropometric and hormonal parameters in Saudi women. J Biomed Sci 2018; 25:50. [PMID: 29848339 PMCID: PMC5975709 DOI: 10.1186/s12929-018-0452-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/22/2018] [Indexed: 11/30/2022] Open
Abstract
Background Kisspeptin is involved in female reproduction. This study was designed to i- estimate kisspeptin levels in women with polycystic ovary syndrome (PCOS), in comparison with controls, ii- study the correlations between kisspeptin and PCOS-related reproductive hormones, and iii- investigate the relation between KISS1 gene polymorphisms and hormone levels in women suffering from PCOS. Methods The investigation was a clinically designed study on 28 women with PCOS, and 30 normal, healthy women with no signs of PCOS as controls. Blood samples were collected between day 3 and day 6 of the menstrual cycle in both groups at 8:00 a.m., and circulating levels of LH, FSH and kisspeptin were estimated. DNA was extracted from whole blood and all coding exons of KISS1 gene were sequenced. Results Women with PCOS had higher LH levels and BMI compared to controls. Plasma kisspeptin levels were positively correlated with LH levels. There was no statistically significant difference between the groups in terms of kisspeptin and FSH levels. The SNP rs4889 C/G, a non-synonymous SNP, was investigated in the PCOS group. The frequency of GG genotype was significantly higher in the PCOS compared to the controls. These patients were more obese, had higher kisspeptin and FSH levels. Conclusion The results of the study show that the genetic variation of KISS1 gene may be a factor contributing to PCOS development. The association between the gene and the gene variation and PCOS need further validation in large-scaled and functional studies.
Collapse
|
35
|
Effects of synbiotic supplementation on metabolic parameters and apelin in women with polycystic ovary syndrome: a randomised double-blind placebo-controlled trial. Br J Nutr 2018; 119:398-406. [DOI: 10.1017/s0007114517003920] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
AbstractPolycystic ovary syndrome (PCOS) is one of the most common causes of infertility in women of reproductive age. Insulin resistance is a main pathophysiologic feature in these patients. According to some studies, the intake of probiotic bacteria may improve glucose homoeostasis. The aim of this study was to investigate the effect of synbiotics on metabolic parameters and apelin in PCOS patients. This randomised double-blind placebo-controlled trial was conducted on eighty-eight PCOS women aged 19–37 years old. The participants were randomly assigned to two groups receiving (1) synbiotic supplement (n44), and (2) placebo (n44) for 12 weeks. Fasting blood samples were taken at baseline and after 12 weeks. The two groups showed no difference in fasting blood sugar (adjusted mean difference: 0·60; 95 % CI −3·80, 5·00,P=0·727), plasma glucose fasting 2-h (adjusted mean difference 2·09; 95 % CI −9·96, 14·15,P=0·134), HbA1c (adjusted mean difference 0·06; 95 % CI −0·09, 0·22,P=0·959), homoeostatic model assessment-insulin resistance (HOMA-IR) (adjusted mean difference: 0·02; 95 % CI −0·99, 1·03,P=0·837), quantitative insulin sensitivity check index (QUICKI) (adjusted mean difference: −0·02; 95 % CI −0·33, 0·29,P=0·940) and C-reactive protein (CRP) (adjusted mean difference: 0·24; 95 % CI −1·61, 2·08,P=0·141) by the end of the intervention. A significant difference was observed in the mean apelin 36 before and after the intervention between synbiotic and placebo groups (adjusted mean difference: −4·05; 95 % CI −7·15, −0·96,P=0·004). A 12-week synbiotic supplementation has no significant beneficial effects on HOMA-IR and CRP in PCOS patients, whereas the level of apelin 36 significantly decreased.
Collapse
|
36
|
Qasim A, Turcotte M, de Souza RJ, Samaan MC, Champredon D, Dushoff J, Speakman JR, Meyre D. On the origin of obesity: identifying the biological, environmental and cultural drivers of genetic risk among human populations. Obes Rev 2018; 19:121-149. [PMID: 29144594 DOI: 10.1111/obr.12625] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/28/2017] [Accepted: 09/08/2017] [Indexed: 12/12/2022]
Abstract
Genetic predisposition to obesity presents a paradox: how do genetic variants with a detrimental impact on human health persist through evolutionary time? Numerous hypotheses, such as the thrifty genotype hypothesis, attempt to explain this phenomenon yet fail to provide a justification for the modern obesity epidemic. In this critical review, we appraise existing theories explaining the evolutionary origins of obesity and explore novel biological and sociocultural agents of evolutionary change to help explain the modern-day distribution of obesity-predisposing variants. Genetic drift, acting as a form of 'blind justice,' may randomly affect allele frequencies across generations while gene pleiotropy and adaptations to diverse environments may explain the rise and subsequent selection of obesity risk alleles. As an adaptive response, epigenetic regulation of gene expression may impact the manifestation of genetic predisposition to obesity. Finally, exposure to malnutrition and disease epidemics in the wake of oppressive social systems, culturally mediated notions of attractiveness and desirability, and diverse mating systems may play a role in shaping the human genome. As an important first step towards the identification of important drivers of obesity gene evolution, this review may inform empirical research focused on testing evolutionary theories by way of population genetics and mathematical modelling.
Collapse
Affiliation(s)
- A Qasim
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - M Turcotte
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - R J de Souza
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - M C Samaan
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada.,Department of Pediatrics, McMaster University, Hamilton, ON, Canada.,Division of Pediatric Endocrinology, McMaster Children's Hospital, Hamilton, ON, Canada
| | - D Champredon
- Department of Biology, McMaster University, Hamilton, ON, Canada.,Agent-Based Modelling Laboratory, York University, Toronto, ON, Canada
| | - J Dushoff
- Department of Biology, McMaster University, Hamilton, ON, Canada
| | - J R Speakman
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - D Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
37
|
Mykhalchenko K, Lizneva D, Trofimova T, Walker W, Suturina L, Diamond MP, Azziz R. Genetics of polycystic ovary syndrome. Expert Rev Mol Diagn 2017; 17:723-733. [DOI: 10.1080/14737159.2017.1340833] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
| | - Daria Lizneva
- Department of OB/GYN, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Reproductive Health Protection, Scientific Center of Family Health and Human Reproduction, Irkutsk, Russian Federation
| | - Tatiana Trofimova
- Department of Reproductive Health Protection, Scientific Center of Family Health and Human Reproduction, Irkutsk, Russian Federation
| | - Walidah Walker
- Department of OB/GYN, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Larisa Suturina
- Department of Reproductive Health Protection, Scientific Center of Family Health and Human Reproduction, Irkutsk, Russian Federation
| | - Michael P. Diamond
- Department of OB/GYN, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ricardo Azziz
- Chief Officer of Academic Health and Hospital Affairs, State University Plaza, The State University of New York, Albany, NY, USA
| |
Collapse
|
38
|
Zaki M, Hassan N, El-Bassyouni HT, Kamal S, Basha W, Azmy O, Amr K. Association of the Pro12Ala Polymorphism with the Metabolic Parameters in Women with Polycystic Ovary Syndrome. Open Access Maced J Med Sci 2017; 5:275-280. [PMID: 28698741 PMCID: PMC5503721 DOI: 10.3889/oamjms.2017.088] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 04/21/2017] [Accepted: 04/22/2017] [Indexed: 11/17/2022] Open
Abstract
AIM: To investigate the association of peroxisome proliferator-activated receptor gamma (PPARG) Pro12Ala polymorphism with polycystic ovary syndrome (PCOS) and its effect on the metabolic parameters in PCOS women. METHODS: The study used PCR to identify the presence of the PPARG Pro12Ala polymorphism in 100 PCOS women and 120 age-matched healthy women. All participants were subjected to anthropometry, biochemical and metabolic evaluation. RESULTS: Significant difference in the genotypes distributions of PPARG Pro12Ala polymorphism was observed among PCOS women and controls (p = 0.03). The frequency of the polymorphic allele Ala was significantly higher in PCOS cases than that in the controls (OR = 2.01, p = 0.01). The carries of the variant allele Ala in PCOS women showed significant higher values in body mass index (BMI), systolic and diastolic blood pressure, waist circumference, waist to hip ratio, sum of skin folds, fasting blood glucose, fasting blood insulin, HOMA-IR, fasting triglycerides, total cholesterol and low-density lipoprotein than non-carriers. CONCLUSION: The PPARG Pro12Ala polymorphism might contribute to the risk of PCOS and abnormal metabolic parameters and could be considered as a biomarker for early diagnosis and clinic prediction of metabolic complications.
Collapse
Affiliation(s)
- Moushira Zaki
- Biological Anthropology Department, National Research Centre, Cairo, Egypt
| | - Naglaa Hassan
- Biological Anthropology Department, National Research Centre, Cairo, Egypt
| | | | - Sanaa Kamal
- Biological Anthropology Department, National Research Centre, Cairo, Egypt
| | - Walaa Basha
- Biological Anthropology Department, National Research Centre, Cairo, Egypt
| | - Osama Azmy
- Reproductive Health Research Department, National Research Centre, Cairo, Egypt
| | - Khalda Amr
- Molecular Genetics Department, National Research Centre, Cairo, Egypt
| |
Collapse
|
39
|
Jafari Nedooshan J, Kargar S, Neamatzadeh H, Haghighi F, Dehghani Mohammad Abadi R, Seddighi N. Lack of Association of the Fat Mass and Obesity Associated (FTO) Gene rs9939609 Polymorphism with Breast Cancer Risk: a Systematic Review and Meta-Analysis Based on Case - Control Studies. Asian Pac J Cancer Prev 2017; 18:1031-1037. [PMID: 28547937 PMCID: PMC5494212 DOI: 10.22034/apjcp.2017.18.4.1031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background: Previously published data on any association of the fat mass and obesity-associated (FTO) gene with
breast cancer risk remain inconclusive. Therefore, we conducted the present meta-analysis of links between breast cancer
and the FTO rs9939609 polymorphism. Methods: We have conducted a systematic review of the English literature by
searching PubMed, Google Scholar and ISI Web of Knowledge databases for studies on associations between the FTO
rs9939609 polymorphism and breast cancer risk. Crude odds ratios (ORs) with 95% confidence intervals (CIs) were
calculated to estimate the strength of the association using fixed- or random-effects model. Results: We included five
studies with 1134 cases and 1453 controls. Overall, no significant association between the FTO rs9939609 polymorphism
and risk of breast cancer was found. On subgroup analysis by ethnicity, there was still no significant association detected.
Conclusions: To our knowledge, this is the first meta-analysis of the FTO rs9939609 polymorphism and risk of breast
cancer. However, the present meta-analysis suggested that only there might be a significant association of the CXCL12
rs1801157 polymorphisms with breast cancer risk.
Collapse
Affiliation(s)
- Jamal Jafari Nedooshan
- Department of General Surgery, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | | | | | | | | | | |
Collapse
|
40
|
Knebel B, Lehr S, Janssen OE, Hahn S, Jacob S, Nitzgen U, Müller-Wieland D, Kotzka J. Association between copy-number variation on metabolic phenotypes and HDL-C levels in patients with polycystic ovary syndrome. Mol Biol Rep 2016; 44:51-61. [DOI: 10.1007/s11033-016-4080-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/09/2016] [Indexed: 01/08/2023]
|
41
|
Chedraui P, Pérez-López FR, Escobar GS, Espinoza-Caicedo JA, Montt-Guevara M, Genazzani AR, Simoncini T. Polymorphisms of the FTO and MTHFR genes and vascular, inflammatory and metabolic marker levels in postmenopausal women. J Endocrinol Invest 2016; 39:885-90. [PMID: 26902996 DOI: 10.1007/s40618-016-0443-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/03/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To determine the prevalence of three single nucleotide polymorphisms (SNPs) in postmenopausal women with and without the metabolic syndrome (METS) and to explore levels of circulating biomarkers of inflammation, vascular and metabolic dysfunction according to SNP genotypes. METHODS DNA was extracted from the whole blood of 192 natural postmenopausal women (40 to 65 years) screened for the METS and tested for three gene SNPs related to obesity: the fat mass obesity (FTO: rs9939609) and the methylenetetrahydrofolate reductase (MTHFR: C677T and A1298C). Blood levels of angiopoietin, IL-8, sFASL, IL-6, TNF-α, sCD40L, PAI-1, u-PA, leptin, adiponectin, resistin, ghrelin, visfatin, adipsin and insulin were measured in a subgroup, with and without the METS, using multiplex technology (n = 100) and compared according to SNP genotypes. RESULTS Genotype frequency of the three studied SNPs did not differ in relation to the presence of the METS. However, genotypes CT+TT (C677T) and AT (rs9939609) were more prevalent in women with high triglyceride levels. Pooled sub-analysis (n = 100) found that median sCD40L and visfatin levels were higher in women with genotypes AT+TT (rs9939609) as compared to AA (1178 vs. 937.0 pg/mL and 0.93 vs. 0.43 ng/mL, respectively, p < 0.05). CONCLUSION Two SNP genotypes related to obesity were more prevalent in women with abnormal triglyceride levels and two vascular and inflammatory serum markers were higher in relation to the rs9939609 SNP.
Collapse
Affiliation(s)
- P Chedraui
- Institute of Biomedicine, Research Area for Women's Health, Facultad de Ciencias Médicas, Universidad Católica de Santiago de Guayaquil, PO BOX 09-01-4671, Guayaquil, Ecuador.
| | - F R Pérez-López
- Department of Obstetrics and Gynecology, Facultad de Medicina, Lozano Blesa University Hospital, Universidad de Zaragoza, Zaragoza, Spain
| | - G S Escobar
- Institute of Biomedicine, Research Area for Women's Health, Facultad de Ciencias Médicas, Universidad Católica de Santiago de Guayaquil, PO BOX 09-01-4671, Guayaquil, Ecuador
| | - J A Espinoza-Caicedo
- Institute of Biomedicine, Research Area for Women's Health, Facultad de Ciencias Médicas, Universidad Católica de Santiago de Guayaquil, PO BOX 09-01-4671, Guayaquil, Ecuador
| | - M Montt-Guevara
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - A R Genazzani
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - T Simoncini
- Division of Obstetrics and Gynecology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
42
|
Wood AR, Tyrrell J, Beaumont R, Jones SE, Tuke MA, Ruth KS, Yaghootkar H, Freathy RM, Murray A, Frayling TM, Weedon MN. Variants in the FTO and CDKAL1 loci have recessive effects on risk of obesity and type 2 diabetes, respectively. Diabetologia 2016; 59:1214-21. [PMID: 26961502 PMCID: PMC4869698 DOI: 10.1007/s00125-016-3908-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/10/2016] [Indexed: 11/26/2022]
Abstract
AIMS/HYPOTHESIS Genome-wide association (GWA) studies have identified hundreds of common genetic variants associated with obesity and type 2 diabetes. These studies have usually focused on additive association tests. Identifying deviations from additivity may provide new biological insights and explain some of the missing heritability for these diseases. METHODS We performed a GWA study using a dominance deviation model for BMI, obesity (29,925 cases) and type 2 diabetes (4,040 cases) in 120,286 individuals of British ancestry from the UK Biobank study. We also investigated whether single nucleotide polymorphisms previously shown to be associated with these traits showed any enrichment for departures from additivity. RESULTS Known obesity-associated variants in FTO showed strong evidence of deviation from additivity (p DOMDEV = 3 × 10(-5)) through a recessive effect of the allele associated with higher BMI. The average BMI of individuals carrying zero, one or two BMI-raising alleles was 27.27 (95% CI 27.22, 27.31) kg/m(2), 27.54 (95% CI 27.50, 27.58) kg/m(2) and 28.07 (95% CI 28.00, 28.14) kg/m(2), respectively. A similar effect was observed in 105,643 individuals from the GIANT Consortium (p DOMDEV = 0.003; meta-analysis p DOMDEV = 1 × 10(-7)). For type 2 diabetes, we detected a recessive effect (p DOMDEV = 5 × 10(-4)) at CDKAL1. Relative to homozygous non-risk allele carriers, homozygous risk allele carriers had an OR of 1.48 (95% CI 1.32, 1.65), while the heterozygous group had an OR of 1.06 (95% CI 0.99, 1.14), a result consistent with that of a previous study. We did not identify any novel associations at genome-wide significance. CONCLUSIONS/INTERPRETATION Although we found no evidence of widespread non-additive genetic effects contributing to obesity and type 2 diabetes risk, we did find robust examples of recessive effects at the FTO and CDKAL1 loci. ACCESS TO RESEARCH MATERIALS Summary statistics are available at www.t2diabetesgenes.org and by request (a.r.wood@exeter.ac.uk). All underlying data are available on application from the UK Biobank.
Collapse
Affiliation(s)
- Andrew R Wood
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK.
| | - Jessica Tyrrell
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Robin Beaumont
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Samuel E Jones
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Marcus A Tuke
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Katherine S Ruth
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | | | - Hanieh Yaghootkar
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Rachel M Freathy
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Anna Murray
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Timothy M Frayling
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| | - Michael N Weedon
- Genetics of Complex Traits, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital, Barrack Road, Exeter, EX2 5DW, UK
| |
Collapse
|
43
|
Genetic determinants of polycystic ovary syndrome: progress and future directions. Fertil Steril 2016; 106:25-32. [PMID: 27179787 DOI: 10.1016/j.fertnstert.2016.04.040] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/21/2016] [Accepted: 04/26/2016] [Indexed: 01/05/2023]
Abstract
The field of the genetics of polycystic ovary syndrome (PCOS) has relatively recently moved into the era of genome-wide association studies. This has led to the discovery of 16 robust loci for PCOS. Some loci contain genes with clear roles in reproductive (LHCGR, FSHR, and FSHB) and metabolic (INSR and HMGA2) dysfunction in the syndrome. The next challenge facing the field is the identification of causal variants and genes and the role they play in PCOS pathophysiology. The potential for gene discovery to improve diagnosis and treatment of PCOS is promising, though there is much to be done in the field before the current findings can be translated to the clinic.
Collapse
|
44
|
Tehrani FR, Zarkesh M, Tohidi M, Azizi F, Zadeh-Vakili A. Is the association between insulin resistance and diabetogenic haematopoietically expressed homeobox (HHEX) polymorphism (rs1111875) affected by polycystic ovary syndrome status? Reprod Fertil Dev 2015; 29:670-678. [PMID: 26563606 DOI: 10.1071/rd15157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 09/29/2015] [Indexed: 12/11/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is frequently accompanied by insulin resistance (IR). The aim of the present study was to investigate whether the genetic association between insulin resistance and two single nucleotide polymorphisms (SNPs), namely rs7903146 (C/T) in transcription factor 7-like 2 (TCF7L2) and rs1111875 (A/G) in haematopoietically expressed homeobox (HHEX), is affected by PCOS status in Iranian women. The study participants consisted of 582 women with PCOS (cases) referred to the Reproductive Endocrinology Research Center and 504 subjects without PCOS (controls), randomly selected from the Tehran Lipid and Glucose Study. Cases and controls were further subdivided to two groups according to IR status: those with and without IR. IR was identified on the basis of homeostasis model assessment of insulin resistance (HOMA-IR) ≥2.63. The SNPs in TCF7L2 and HHEX were genotyped by polymerase chain reaction-restriction fragment length polymorphism. There were no significant differences in the distribution of genotypes and alleles between cases and controls (P<0.05). Among cases, the prevalence of the CC, CT and TT genotypes was 37.8%, 46.3% and 15.9%, respectively, whereas the prevalence of the AA, AG and GG genotypes was 13.5%, 46.1% and 40.4%, respectively. In the control group, the prevalence of the CC, CT and TT genotypes was 32.2%, 53.9% and 13.9%, respectively, whereas the prevalence of the AA, AG and GG genotypes was 11.3%, 48.6% and 40.0%, respectively. After adjustment for age and body mass index, the probability of IR was decreased by 49% among carriers of the A allele in the control group (95% confidence interval 0.33-0.78; P=0.002). The findings of the present study suggest that the association between IR and diabetogenic polymorphisms may be affected by PCOS status.
Collapse
Affiliation(s)
- F Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24 Arabi Street - Yemen Street - Velenjak - Tehran, 1985717413, Islamic Republic of Iran
| | - M Zarkesh
- Cellular and Molecular Endocrine Research Center, Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24 Arabi Street - Yemen Street - Velenjak - Tehran, 1985717413, Islamic Republic of Iran
| | - M Tohidi
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24 Arabi Street - Yemen Street - Velenjak - Tehran, 1985717413, Islamic Republic of Iran
| | - F Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24 Arabi Street - Yemen Street - Velenjak - Tehran, 1985717413, Islamic Republic of Iran
| | - A Zadeh-Vakili
- Cellular and Molecular Endocrine Research Center, Obesity Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24 Arabi Street - Yemen Street - Velenjak - Tehran, 1985717413, Islamic Republic of Iran
| |
Collapse
|
45
|
Dumesic DA, Oberfield SE, Stener-Victorin E, Marshall JC, Laven JS, Legro RS. Scientific Statement on the Diagnostic Criteria, Epidemiology, Pathophysiology, and Molecular Genetics of Polycystic Ovary Syndrome. Endocr Rev 2015; 36:487-525. [PMID: 26426951 PMCID: PMC4591526 DOI: 10.1210/er.2015-1018] [Citation(s) in RCA: 594] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous and complex disorder that has both adverse reproductive and metabolic implications for affected women. However, there is generally poor understanding of its etiology. Varying expert-based diagnostic criteria utilize some combination of oligo-ovulation, hyperandrogenism, and the presence of polycystic ovaries. Criteria that require hyperandrogenism tend to identify a more severe reproductive and metabolic phenotype. The phenotype can vary by race and ethnicity, is difficult to define in the perimenarchal and perimenopausal period, and is exacerbated by obesity. The pathophysiology involves abnormal gonadotropin secretion from a reduced hypothalamic feedback response to circulating sex steroids, altered ovarian morphology and functional changes, and disordered insulin action in a variety of target tissues. PCOS clusters in families and both female and male relatives can show stigmata of the syndrome, including metabolic abnormalities. Genome-wide association studies have identified a number of candidate regions, although their role in contributing to PCOS is still largely unknown.
Collapse
Affiliation(s)
- Daniel A Dumesic
- Department of Obstetrics and Gynecology (D.A.D.), David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Division of Pediatric Endocrinology (S.E.O.), Children's Hospital of New York-Presbyterian, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Physiology (E.S.-V.), Karolinska Institutet, 171 77 Stockholm, Sweden; Center for Research in Reproduction and Division of Endocrinology (J.C.M.), Department of Internal Medicine, University of Virginia Health System, Charlottesville, Virginia 22903; Division of Reproductive Medicine (J.S.L.), Department of Obstetrics and Gynecology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; and Department of Obstetrics and Gynecology (R.S.L.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Sharon E Oberfield
- Department of Obstetrics and Gynecology (D.A.D.), David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Division of Pediatric Endocrinology (S.E.O.), Children's Hospital of New York-Presbyterian, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Physiology (E.S.-V.), Karolinska Institutet, 171 77 Stockholm, Sweden; Center for Research in Reproduction and Division of Endocrinology (J.C.M.), Department of Internal Medicine, University of Virginia Health System, Charlottesville, Virginia 22903; Division of Reproductive Medicine (J.S.L.), Department of Obstetrics and Gynecology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; and Department of Obstetrics and Gynecology (R.S.L.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Elisabet Stener-Victorin
- Department of Obstetrics and Gynecology (D.A.D.), David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Division of Pediatric Endocrinology (S.E.O.), Children's Hospital of New York-Presbyterian, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Physiology (E.S.-V.), Karolinska Institutet, 171 77 Stockholm, Sweden; Center for Research in Reproduction and Division of Endocrinology (J.C.M.), Department of Internal Medicine, University of Virginia Health System, Charlottesville, Virginia 22903; Division of Reproductive Medicine (J.S.L.), Department of Obstetrics and Gynecology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; and Department of Obstetrics and Gynecology (R.S.L.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - John C Marshall
- Department of Obstetrics and Gynecology (D.A.D.), David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Division of Pediatric Endocrinology (S.E.O.), Children's Hospital of New York-Presbyterian, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Physiology (E.S.-V.), Karolinska Institutet, 171 77 Stockholm, Sweden; Center for Research in Reproduction and Division of Endocrinology (J.C.M.), Department of Internal Medicine, University of Virginia Health System, Charlottesville, Virginia 22903; Division of Reproductive Medicine (J.S.L.), Department of Obstetrics and Gynecology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; and Department of Obstetrics and Gynecology (R.S.L.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Joop S Laven
- Department of Obstetrics and Gynecology (D.A.D.), David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Division of Pediatric Endocrinology (S.E.O.), Children's Hospital of New York-Presbyterian, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Physiology (E.S.-V.), Karolinska Institutet, 171 77 Stockholm, Sweden; Center for Research in Reproduction and Division of Endocrinology (J.C.M.), Department of Internal Medicine, University of Virginia Health System, Charlottesville, Virginia 22903; Division of Reproductive Medicine (J.S.L.), Department of Obstetrics and Gynecology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; and Department of Obstetrics and Gynecology (R.S.L.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Richard S Legro
- Department of Obstetrics and Gynecology (D.A.D.), David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Division of Pediatric Endocrinology (S.E.O.), Children's Hospital of New York-Presbyterian, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Physiology (E.S.-V.), Karolinska Institutet, 171 77 Stockholm, Sweden; Center for Research in Reproduction and Division of Endocrinology (J.C.M.), Department of Internal Medicine, University of Virginia Health System, Charlottesville, Virginia 22903; Division of Reproductive Medicine (J.S.L.), Department of Obstetrics and Gynecology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; and Department of Obstetrics and Gynecology (R.S.L.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| |
Collapse
|
46
|
Ramos RB, Fabris VC, Brondani LDA, Spritzer PM. Association between rs7903146 and rs12255372 polymorphisms of transcription factor 7-like 2 gene and polycystic ovary syndrome: a systematic review and meta-analysis. Endocrine 2015; 49:635-42. [PMID: 25678248 DOI: 10.1007/s12020-015-0541-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 01/27/2015] [Indexed: 12/11/2022]
Abstract
The present systematic review and meta-analysis of case-control studies examines the associations between rs7903146 and rs12255372 polymorphisms of the TCF7L2 gene and PCOS. A search of the literature published until August 2014 was carried out in PubMed, Cochrane Central Register of Controlled Trials, EMBASE, and LILACS. There were no other limits except for the end date. We included observational studies with women of any age diagnosed with PCOS and healthy controls that analyzed polymorphisms rs7903146 and rs12255372. We included case-control or cross-sectional studies analyzing polymorphism rs7903146 or rs12255372 in women with PCOS and healthy controls. Eighteen studies were identified after the primary literature search and seven articles were included in the meta-analysis. All employed Rotterdam criteria for the diagnosis of PCOS. The genotypic distributions in the control groups were in agreement with Hardy-Weinberg equilibrium in all studies. The pooled population included Asian descendents (Chinese, Korean), Caucasians from southern Brazil, Tunisian, and European populations (British/Irish, Northern Finns, Greeks, Czechs), including 1,892 women with PCOS and 2,695 controls. There were no significant associations between PCOS and TCF7L2 rs7903146 or rs12255372 polymorphisms, irrespective of whether allele contrast, additive, dominant, or recessive models of inheritance were used. Furthermore, no significant associations were found after stratification for ethnicity (Asian or non-Asian). There was no significant heterogeneity between studies and no publication bias. The present results suggest that rs7903146 T allele or rs12255372 is not associated with risk for PCOS in non-Asian or Asian women. This systematic review and meta-analysis are registered in PROSPERO under number CRD42013005930.
Collapse
Affiliation(s)
- Ramon Bossardi Ramos
- Gynecological Endocrinology Unit, Division of Endocrinology, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Porto Alegre, RS, 90035-003, Brazil
| | | | | | | |
Collapse
|
47
|
Yuan H, Zhu G, Wang F, Wang X, Guo H, Shen M. Interaction between common variants of FTO and MC4R is associated with risk of PCOS. Reprod Biol Endocrinol 2015; 13:55. [PMID: 26032905 PMCID: PMC4455322 DOI: 10.1186/s12958-015-0050-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/23/2015] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common and complex endocrine-metabolic disease. One of the well-documented characteristics of PCOS is obesity or overweightness. It is possible to be genetically predisposed to becoming obese or overweight, and several potentially causative single nucleotide polymorphisms (SNPs), such as rs9939609 (A/T) in the fat mass, and obesity-associated gene (FTO) and rs17782313 (T/C) in the melanocortin-4 receptor gene (MC4R), have been investigated. Further investigation of association between obesity-associated SNPs and PCOS susceptibility will contribute to a better understanding of the disease. METHODS In the present study, we enrolled 733 patients with PCOS and 892 control subjects. The common variants FTO rs9939609 and MC4R rs17782313 were genotyped and their relationship with obesity-related traits was evaluated. RESULTS Rs9939609 and rs17782313 are associated with PCOS and obesity-related traits and profiles. The association found between PCOS and FTO rs9939609 (p=0.0302) was attenuated after adjustment for BMI (p=0.187). MC4R rs17782313 did not confer an increased risk for PCOS (p=0.368) even after adjustments (p=0.715). Interestingly, the interaction of FTO and MC4R polymorphisms was more significantly associated with PCOS (p=0.031, adjusted for age and BMI). The FTO variant rs9939609 is associated with Chinese women with PCOS; however, this association is affected by BMI. CONCLUSIONS The combined pathogenic effect of FTO and MC4R polymorphisms indicates a direct role of the interaction between FTO and MC4R polymorphisms in the development of PCOS.
Collapse
Affiliation(s)
- Huiqin Yuan
- Department of Gynaecology and Obstetrics, First Affiliated Hospital, Huzhou Teachers College, the First People's Hospital of Huzhou, Huzhou, 313000, China.
| | - Guoping Zhu
- Department of Gynaecology and Obstetrics, First Affiliated Hospital, Huzhou Teachers College, the First People's Hospital of Huzhou, Huzhou, 313000, China.
| | - Fang Wang
- Department of Gynaecology and Obstetrics, First Affiliated Hospital, Huzhou Teachers College, the First People's Hospital of Huzhou, Huzhou, 313000, China.
| | - Xiang Wang
- Department of Laboratory Medicine, First Affiliated Hospital, Huzhou Teachers College, the First People's Hospital of Huzhou, Huzhou, 313000, China.
| | - Huihui Guo
- Department of Laboratory Medicine, First Affiliated Hospital, Huzhou Teachers College, the First People's Hospital of Huzhou, Huzhou, 313000, China.
| | - Mo Shen
- Department of Laboratory Medicine, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
48
|
Yılmaz SA, Altınkaya SÖ, Kebabçılar A, Seçilmiş Kerimoğlu Ö, Tazegül Pekin A, Abuşoğlu S, Çelik Ç, Ünlü A. The relationship between Polycystic ovary syndrome and vitamin D levels. Turk J Obstet Gynecol 2015; 12:18-24. [PMID: 28913035 PMCID: PMC5558399 DOI: 10.4274/tjod.76148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/17/2014] [Indexed: 12/28/2022] Open
Abstract
Objective: The main aim of this study was to determine the association of serum 25-hydroxyvitamin D (25-OH D) levels with hormonal, clinical and metabolic profile in patients with and without Polycystic ovary syndrome (PCOS). Materials and Methods: Forty-eight normal-weight (body mass index (BMI) of 19-24.99 kg/m2) women with PCOS, 36 overweight (BMI of 25-29.9 kg/m2) women with PCOS and 56 normal-weight controls participated in the study. Blood samples were collected in the early follicular phase (between day 2 and day 5 of the menstrual cycle) at 9:00 am after an overnight fast. Circulating concentrations of 25-OH D, luteinizing hormone (LH), follicle stimulating hormone (FSH), prolactin, TSH, free testosterone, dehydroepiandrosterone sulphate (DHEA-SO4), 17-hydroxyprogesterone, sex hormone-binding globulin (SHBG), fasting insulin, fasting glucose, and lipid profile were assessed. Results: Normal weight (BMI 19-24.99 kg/m2) and overweight (BMI 25-29.99 kg/m2) women with PCOS were compared with normal-weight controls and lower 25-OH D levels were found in both PCOS groups (p<0.05 and p<0.01, respectively 25-OH D significantly negatively correlated with waist circumference (WC), waist-to-hip ratio (WHR), free testosterone and modified Ferriman-Gallwey scores, however, there was a positive correlation between 25-OH D and SHBG levels (p<0.05). Conclusion: Our findings suggest that PCOS is associated with hypovitaminosis D.
Collapse
Affiliation(s)
- Setenay Arzu Yılmaz
- Selçuk University Faculty of Medicine, Department of Obstetrics and Gynecology, Konya, Turkey
| | - Sündüz Özlem Altınkaya
- Adnan Menderes University Faculty of Medicine, Department of Obstetrics and Gynecology, Aydın, Turkey
| | - Ayşegül Kebabçılar
- Selçuk University Faculty of Medicine, Department of Obstetrics and Gynecology, Konya, Turkey
| | | | - Aybike Tazegül Pekin
- Selçuk University Faculty of Medicine, Department of Obstetrics and Gynecology, Konya, Turkey
| | - Sedat Abuşoğlu
- Selçuk University Faculty of Medicine, Department of Biochemistry, Konya, Turkey
| | - Çetin Çelik
- Selçuk University Faculty of Medicine, Department of Obstetrics and Gynecology, Konya, Turkey
| | - Ali Ünlü
- Selçuk University Faculty of Medicine, Department of Biochemistry, Konya, Turkey
| |
Collapse
|
49
|
Gaberšček S, Zaletel K, Schwetz V, Pieber T, Obermayer-Pietsch B, Lerchbaum E. Mechanisms in endocrinology: thyroid and polycystic ovary syndrome. Eur J Endocrinol 2015; 172:R9-21. [PMID: 25422352 DOI: 10.1530/eje-14-0295] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Thyroid disorders, especially Hashimoto's thyroiditis (HT), and polycystic ovary syndrome (PCOS) are closely associated, based on a number of studies showing a significantly higher prevalence of HT in women with PCOS than in controls. However, the mechanisms of this association are not as clear. Certainly, genetic susceptibility contributes an important part to the development of HT and PCOS. However, a common genetic background has not yet been established. Polymorphisms of the PCOS-related gene for fibrillin 3 (FBN3) could be involved in the pathogenesis of HT and PCOS. Fibrillins influence the activity of transforming growth factor beta (TGFβ). Multifunctional TGFβ is also a key regulator of immune tolerance by stimulating regulatory T cells (Tregs), which are known to inhibit excessive immune response. With lower TGFβ and Treg levels, the autoimmune processes, well known in HT and assumed in PCOS, might develop. In fact, lower levels of TGFβ1 were found in HT as well as in PCOS women carrying allele 8 of D19S884 in the FBN3 gene. Additionally, vitamin D deficiency was shown to decrease Tregs. Finally, high estrogen-to-progesterone ratio owing to anovulatory cycles in PCOS women could enhance the immune response. Harmful metabolic and reproductive effects were shown to be more pronounced in women with HT and PCOS when compared with women with HT alone or with controls. In conclusion, HT and PCOS are associated not only with respect to their prevalence, but also with regard to etiology and clinical consequences. However, a possible crosstalk of this association is yet to be elucidated.
Collapse
Affiliation(s)
- Simona Gaberšček
- Department of Nuclear MedicineUniversity Medical Centre Ljubljana, Zaloška 7, 1525 Ljubljana, SloveniaFaculty of MedicineUniversity of Ljubljana, Vrazov trg 2, 1104 Ljubljana, SloveniaDivision of Endocrinology and MetabolismDepartment of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria Department of Nuclear MedicineUniversity Medical Centre Ljubljana, Zaloška 7, 1525 Ljubljana, SloveniaFaculty of MedicineUniversity of Ljubljana, Vrazov trg 2, 1104 Ljubljana, SloveniaDivision of Endocrinology and MetabolismDepartment of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Katja Zaletel
- Department of Nuclear MedicineUniversity Medical Centre Ljubljana, Zaloška 7, 1525 Ljubljana, SloveniaFaculty of MedicineUniversity of Ljubljana, Vrazov trg 2, 1104 Ljubljana, SloveniaDivision of Endocrinology and MetabolismDepartment of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Verena Schwetz
- Department of Nuclear MedicineUniversity Medical Centre Ljubljana, Zaloška 7, 1525 Ljubljana, SloveniaFaculty of MedicineUniversity of Ljubljana, Vrazov trg 2, 1104 Ljubljana, SloveniaDivision of Endocrinology and MetabolismDepartment of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Thomas Pieber
- Department of Nuclear MedicineUniversity Medical Centre Ljubljana, Zaloška 7, 1525 Ljubljana, SloveniaFaculty of MedicineUniversity of Ljubljana, Vrazov trg 2, 1104 Ljubljana, SloveniaDivision of Endocrinology and MetabolismDepartment of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Barbara Obermayer-Pietsch
- Department of Nuclear MedicineUniversity Medical Centre Ljubljana, Zaloška 7, 1525 Ljubljana, SloveniaFaculty of MedicineUniversity of Ljubljana, Vrazov trg 2, 1104 Ljubljana, SloveniaDivision of Endocrinology and MetabolismDepartment of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Elisabeth Lerchbaum
- Department of Nuclear MedicineUniversity Medical Centre Ljubljana, Zaloška 7, 1525 Ljubljana, SloveniaFaculty of MedicineUniversity of Ljubljana, Vrazov trg 2, 1104 Ljubljana, SloveniaDivision of Endocrinology and MetabolismDepartment of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| |
Collapse
|
50
|
Nomair AM, Aref NK, Rizwan F, Ezzo OH, Hassan N. Serum leptin level in obese women with polycystic ovary syndrome, and its relation to insulin resistance. ASIAN PACIFIC JOURNAL OF REPRODUCTION 2014. [DOI: 10.1016/s2305-0500(14)60041-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|