Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 383
Filter
1.
Nature ; 633(8030): 608-614, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39261734

ABSTRACT

Human genetic studies of common variants have provided substantial insight into the biological mechanisms that govern ovarian ageing1. Here we report analyses of rare protein-coding variants in 106,973 women from the UK Biobank study, implicating genes with effects around five times larger than previously found for common variants (ETAA1, ZNF518A, PNPLA8, PALB2 and SAMHD1). The SAMHD1 association reinforces the link between ovarian ageing and cancer susceptibility1, with damaging germline variants being associated with extended reproductive lifespan and increased all-cause cancer risk in both men and women. Protein-truncating variants in ZNF518A are associated with shorter reproductive lifespan-that is, earlier age at menopause (by 5.61 years) and later age at menarche (by 0.56 years). Finally, using 8,089 sequenced trios from the 100,000 Genomes Project (100kGP), we observe that common genetic variants associated with earlier ovarian ageing associate with an increased rate of maternally derived de novo mutations. Although we were unable to replicate the finding in independent samples from the deCODE study, it is consistent with the expected role of DNA damage response genes in maintaining the genetic integrity of germ cells. This study provides evidence of genetic links between age of menopause and cancer risk.


Subject(s)
Aging , Genetic Predisposition to Disease , Menopause , Mutation Rate , Neoplasms , Ovary , Adult , Female , Humans , Male , Middle Aged , Aging/genetics , Aging/pathology , DNA Damage/genetics , Fertility/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Genome, Human/genetics , Germ-Line Mutation/genetics , Menarche/genetics , Menopause/genetics , Neoplasms/genetics , Ovary/metabolism , Ovary/pathology , Time Factors , UK Biobank , United Kingdom/epidemiology
3.
Front Endocrinol (Lausanne) ; 15: 1429514, 2024.
Article in English | MEDLINE | ID: mdl-39247918

ABSTRACT

Background: Evidence suggests a connection between DNA methylation (DNAm) aging and reproductive aging. However, the causal relationship between DNAm and age at menopause remains uncertain. Methods: Employing established DNAm epigenetic clocks, such as DNAm Hannum age acceleration (Hannum), Intrinsic epigenetic age acceleration (IEAA), DNAm-estimated granulocyte proportions (Gran), DNAm GrimAge acceleration (GrimAgeAccel), DNAm PhenoAge acceleration (PhenoAgeAccel), and DNAm-estimated plasminogen activator inhibitor-1 levels (DNAmPAIadjAge), a bidirectional Mendelian randomization (MR) study was carried out to explore the potential causality between DNAm and menopausal age. The primary analytical method used was the inverse variance weighted (IVW) estimation model, supplemented by various other estimation techniques. Results: DNAm aging acceleration or deceleration, as indicated by Hannum, IEAA, Gran, GrimAgeAccel, PhenoAgeAccel, and DNAmPAIadjAge, did not exhibit a statistically significant causal effect on menopausal age according to forward MR analysis. However, there was a suggestive positive causal association between age at menopause and Gran (Beta = 0.0010; 95% confidence interval (CI): 0.0004, 0.0020) in reverse MR analysis. Conclusion: The observed increase in granulocyte DNAm levels in relation to menopausal age could potentially serve as a valuable indicator for evaluating the physiological status at the onset of menopause.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Mendelian Randomization Analysis , Menopause , Humans , Female , Menopause/genetics , Middle Aged , Aging/genetics , Adult , Age Factors
4.
Nat Genet ; 56(9): 1804-1810, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39192094

ABSTRACT

Age at menopause (AOM) has a substantial impact on fertility and disease risk. While many loci with variants that associate with AOM have been identified through genome-wide association studies (GWAS) under an additive model, other genetic models are rarely considered1. Here through GWAS meta-analysis under the recessive model of 174,329 postmenopausal women from Iceland, Denmark, the United Kingdom (UK; UK Biobank) and Norway, we study low-frequency variants with a large effect on AOM. We discovered that women homozygous for the stop-gain variant rs117316434 (A) in CCDC201 (p.(Arg162Ter), minor allele frequency ~1%) reached menopause 9 years earlier than other women (P = 1.3 × 10-15). The genotype is present in one in 10,000 northern European women and leads to primary ovarian insufficiency in close to half of them. Consequently, homozygotes have fewer children, and the age at last childbirth is 5 years earlier (P = 3.8 × 10-5). The CCDC201 gene was only found in humans in 2022 and is highly expressed in oocytes. Homozygosity for CCDC201 loss-of-function has a substantial impact on female reproductive health, and homozygotes would benefit from reproductive counseling and treatment for symptoms of early menopause.


Subject(s)
Genome-Wide Association Study , Homozygote , Primary Ovarian Insufficiency , Humans , Female , Primary Ovarian Insufficiency/genetics , Polymorphism, Single Nucleotide , Middle Aged , Menopause/genetics , United Kingdom , Gene Frequency , Iceland , Denmark , Genetic Predisposition to Disease
5.
Climacteric ; 27(5): 501-506, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39051435

ABSTRACT

OBJECTIVES: Observational studies have suggested an association between age at natural menopause (ANM) and ventricular structure and function. Nevertheless, the causal relationship remains unclear. This study aimed to evaluate the causal effects of ANM on ventricular structure and function by Mendelian randomization (MR) analysis. METHODS: Genome-wide association summary statistics for ANM and 16 ventricular structures and functions were obtained. The inverse variance weighted (IVW) method was the primary MR approach for assessing causal associations. In addition, three additional MR methods (MR-Egger, weighted median and weighted mode) were performed to complement the IVW method. Furthermore, various sensitivity tests were conducted to evaluate the reliability of the MR results. RESULTS: The IVW method identified no causal association between ANM and all 16 ventricular structures or functions (p > 0.05). Three additional MR methods yielded parallel results to the IVW approach (p > 0.05). Various sensitivity tests revealed stability of the MR results, indicating no heterogeneity or horizontal pleiotropy. CONCLUSION: The present MR study indicated that ANM would not causally affect ventricular structure or function. Therefore, the correlation between ANM and ventricular characteristics in previous observational studies might be attributed to shared upstream cardiovascular risk factors or unidentified genetic mutations that simultaneously affect both ANM and ventricular structure and function.


Subject(s)
Genome-Wide Association Study , Heart Ventricles , Mendelian Randomization Analysis , Menopause , Humans , Female , Menopause/genetics , Middle Aged , Heart Ventricles/anatomy & histology , Age Factors , Ventricular Function
6.
Horm Behav ; 164: 105596, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38944998

ABSTRACT

In a subset of females, postmenopausal status has been linked to accelerated aging and neurological decline. A complex interplay between reproductive-related factors, mental disorders, and genetics may influence brain function and accelerate the rate of aging in the postmenopausal phase. Using multiple regressions corrected for age, in this preregistered study we investigated the associations between menopause-related factors (i.e., menopausal status, menopause type, age at menopause, and reproductive span) and proxies of cellular aging (leukocyte telomere length, LTL) and brain aging (white and gray matter brain age gap, BAG) in 13,780 females from the UK Biobank (age range 39-82). We then determined how these proxies of aging were associated with each other, and evaluated the effects of menopause-related factors, history of depression (= lifetime broad depression), and APOE ε4 genotype on BAG and LTL, examining both additive and interactive relationships. We found that postmenopausal status and older age at natural menopause were linked to longer LTL and lower BAG. Surgical menopause and longer natural reproductive span were also associated with longer LTL. BAG and LTL were not significantly associated with each other. The greatest variance in each proxy of biological aging was most consistently explained by models with the addition of both lifetime broad depression and APOE ε4 genotype. Overall, this study demonstrates a complex interplay between menopause-related factors, lifetime broad depression, APOE ε4 genotype, and proxies of biological aging. However, results are potentially influenced by a disproportionate number of healthier participants among postmenopausal females. Future longitudinal studies incorporating heterogeneous samples are an essential step towards advancing female health.


Subject(s)
Aging , Apolipoprotein E4 , Menopause , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Aging/genetics , Aging/physiology , Apolipoprotein E4/genetics , Brain/metabolism , Cohort Studies , Depression/genetics , Menopause/genetics , Menopause/physiology , UK Biobank , United Kingdom
7.
Medicine (Baltimore) ; 103(24): e38318, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38875424

ABSTRACT

Observational studies have revealed that several sleep traits can impact ovarian function in women. However, there is no evidence suggesting associations between sleep traits and age at natural menopause (ANM). The objective of this study was to investigate the causal relationship between sleep traits (insomnia, sleep duration, daytime sleepiness) and ANM from the perspective of genetic variation. We selected the single-nucleotide polymorphisms from large-scale genome-wide association studies as instrumental variables and conducted a two-sample Mendelian randomization (MR) analysis on these single-nucleotide polymorphisms, including inverse variance weighting, MR-Egger, weighted median, simple mode and weighted mode. The Steiger test was employed to verify the correct causal directionality. The robustness of the MR analysis was examined through Cochran's Q test, horizontal pleiotropy test, and leave-one-out analysis. The results indicated that insomnia was causally associated with ANM (inverse variance weighting: ß = -0.982; 95% CI: -1.852 to -0.111, P = .027), with other analyses confirming the robustness of this finding. Steiger test and reverse MR Analysis validated the absence of a reverse causal association between the two. However, sleep duration and daytime sleepiness did not exhibit a causal effect on ANM. In summary, this study provides initial evidence that insomnia can contribute to an earlier onset of ANM. Nevertheless, further clinical studies are needed to elucidate these findings.


Subject(s)
Genome-Wide Association Study , Mendelian Randomization Analysis , Menopause , Polymorphism, Single Nucleotide , Sleep , Humans , Menopause/genetics , Female , Sleep/genetics , Sleep/physiology , Sleep Initiation and Maintenance Disorders/genetics , Sleep Initiation and Maintenance Disorders/epidemiology , Age Factors , Middle Aged
8.
Calcif Tissue Int ; 115(1): 41-52, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38743269

ABSTRACT

Previous observational studies have suggested that anti-Müllerian hormone (AMH) and reproductive factors are linked to reduced bone mineral density (BMD) and an increased risk of osteoporosis (OP) in women. However, related studies are limited, and these traditional observational studies may be subject to residual confounders and reverse causation, while also lacking a more comprehensive observation of various reproductive factors. Univariate and multivariate two-sample Mendelian randomization analyses were conducted to determine the causal associations of AMH levels and six reproductive factors with BMD and OP, using the random-effects inverse-variance weighted method. Heterogeneity was assessed using Cochran's Q-statistic, and sensitivity analyses were performed to identify causal correlations. Age at menarche (AAM) was negatively associated with total body BMD (TB-BMD) in females aged 45-60 and over 60 years, as well as with heel bone mineral density (eBMD). Conversely, age at natural menopause (ANM) was positively associated with TB-BMD in the same age ranges and with eBMD. ANM was only causally associated with self-reported OP and showed no significant correlation with definitively diagnosed OP. Neither AMH level nor other reproductive factors were significantly associated with a genetic predisposition to BMD at any age and OP. Later AAM and earlier ANM are significantly genetically causally associated with decreased BMD but not with OP. AMH levels, length of menstrual cycle, age at first birth, age at last birth, and number of live births, in terms of genetic backgrounds, are not causally related to BMD or OP.


Subject(s)
Anti-Mullerian Hormone , Bone Density , Mendelian Randomization Analysis , Osteoporosis , Humans , Anti-Mullerian Hormone/blood , Female , Bone Density/genetics , Bone Density/physiology , Middle Aged , Osteoporosis/genetics , Menopause/genetics , Menopause/blood , Genetic Predisposition to Disease , Menarche/genetics , Adult , Risk Factors
9.
Genome Med ; 16(1): 69, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38802955

ABSTRACT

BACKGROUND: The role of metabolism in the variation of age at menarche (AAM) and age at natural menopause (ANM) in the female population is not entirely known. We aimed to investigate the causal role of circulating metabolites in AAM and ANM using Mendelian randomization (MR). METHODS: We combined MR with genetic colocalization to investigate potential causal associations between 658 metabolites and AAM and between 684 metabolites and ANM. We extracted genetic instruments for our exposures from four genome-wide association studies (GWAS) on circulating metabolites and queried the effects of these variants on the outcomes in two large GWAS from the ReproGen consortium. Additionally, we assessed the mediating role of the body mass index (BMI) in these associations, identified metabolic pathways implicated in AAM and ANM, and sought validation for selected metabolites in the Avon Longitudinal Study of Parents and Children (ALSPAC). RESULTS: Our analysis identified 10 candidate metabolites for AAM, but none of them colocalized with AAM. For ANM, 76 metabolites were prioritized (FDR-adjusted MR P-value ≤ 0.05), with 17 colocalizing, primarily in the glycerophosphocholines class, including the omega-3 fatty acid and phosphatidylcholine (PC) categories. Pathway analyses and validation in ALSPAC mothers also highlighted the role of omega and polyunsaturated fatty acids levels in delaying age at menopause. CONCLUSIONS: Our study suggests that metabolites from the glycerophosphocholine and fatty acid families play a causal role in the timing of both menarche and menopause. This underscores the significance of specific metabolic pathways in the biology of female reproductive longevity.


Subject(s)
Genome-Wide Association Study , Menarche , Mendelian Randomization Analysis , Menopause , Metabolome , Humans , Menarche/genetics , Menarche/metabolism , Female , Menopause/genetics , Age Factors , Metabolomics/methods , Body Mass Index
10.
Int J Mol Sci ; 25(7)2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38612676

ABSTRACT

For much of human evolution, the average lifespan was <40 years, due in part to disease, infant mortality, predators, food insecurity, and, for females, complications of childbirth. Thus, for much of evolution, many females did not reach the age of menopause (45-50 years of age) and it is mainly in the past several hundred years that the lifespan has been extended to >75 years, primarily due to public health advances, medical interventions, antibiotics, and nutrition. Therefore, the underlying biological mechanisms responsible for disease risk following menopause must have evolved during the complex processes leading to Homo sapiens to serve functions in the pre-menopausal state. Furthermore, as a primary function for the survival of the species is effective reproduction, it is likely that most of the advantages of having such post-menopausal risks relate to reproduction and the ability to address environmental stresses. This opinion/perspective will be discussed in the context of how such post-menopausal risks could enhance reproduction, with improved survival of offspring, and perhaps why such risks are preserved. Not all post-menopausal females exhibit risk for this set of diseases, and those who do develop such diseases do not have all of the conditions. The diseases of the post-menopausal state do not operate as a unified complex, but as independent variables, with the potential for some overlap. The how and why there would be such heterogeneity if the risk factors serve essential functions during the reproductive years is also discussed and the concept of sets of reversible epigenetic changes associated with puberty, pregnancy, and lactation is offered to explain the observations regarding the distribution of post-menopausal conditions and their potential roles in reproduction. While the involvement of an epigenetic system with a dynamic "modification-demodification-remodification" paradigm contributing to disease risk is a hypothesis at this point, validation of it could lead to a better understanding of post-menopausal disease risk in the context of reproduction with commonalities may also lead to future improved interventions to control such risk after menopause.


Subject(s)
Menopause , Postmenopause , Infant , Pregnancy , Female , Humans , Middle Aged , Menopause/genetics , Menstrual Cycle , Lactation/genetics , Puberty , Epigenesis, Genetic
11.
Sci Rep ; 14(1): 9413, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38658584

ABSTRACT

Previous studies investigating the relationship between systemic lupus erythematosus (SLE) and primary ovarian failure (POF) generated conflicting results. To data, no mendelian randomization study has been applied to examine this association. In this study, genetic instruments for exposure (SLE) were selected from a GWAS study with 5201 cases and 9066 noncases. Outcome data for POF and three reproductive traits (age at menarche, age at menopause, and age at first live birth) were obtained from other eligible GWASs. To estimate causal association, the inverse-variance weighted (IVW) method (the main analyse), MR Egger test, weighted median, simple mode, and weighted mode were applied. Moreover, sensitivity analyses were conducted to ensure the robustness of the results. Estimated by the IVW method, SLE was suggested to be causally related to the risk of POF (OR = 1.166, 95% CI 1.055-1.289, P = 0.003) and delayed age at first live birth (OR = 1.006, 95% CI 1.002-1.010, P = 0.007), with no evidence of a causal association between SLE and age at menopause or menarche. The estimates were robust according to sensitivity analysis. In conclusion, the two-sample MR study supported a causal association between SLE and POF from a genetic aspect.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Lupus Erythematosus, Systemic , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Primary Ovarian Insufficiency , Humans , Lupus Erythematosus, Systemic/genetics , Primary Ovarian Insufficiency/genetics , Female , Menarche/genetics , Risk Factors , Menopause/genetics , Adult
12.
Maturitas ; 183: 107942, 2024 May.
Article in English | MEDLINE | ID: mdl-38412592

ABSTRACT

OBJECTIVES: Fluctuating estradiol (E2) levels seem to be associated with menopausal symptoms, though not all women suffer from these symptoms to the same extent despite experiencing these hormonal changes. This suggests underlying, interindividual mechanisms, such as single-nucleotide polymorphisms (SNPs) influencing estrogen receptors α and ß, and the g-protein-coupled estrogen receptor (GPER). As research is scarce, we aimed to address this research gap by assessing genetic traits, E2 levels, and menopausal symptoms longitudinally. STUDY DESIGN: 129 perimenopausal women (aged 40-56 years) participated in the 13-month longitudinal Swiss Perimenopause Study. MAIN OUTCOME MEASURES: Menopausal symptoms were assessed fortnightly using the Menopause Rating Scale (MRS II). Salivary E2 levels were assessed 14 times over two non-consecutive months. Blood samples were collected using the dried blood spot (DBS) technique to analyze ESR1 rs2234693, ESR1 rs9340799, ESR2 rs1256049, ESR2 rs4906938, and GPER rs3808350. Group-based trajectory modeling was performed to identify interindividual trajectories of menopausal symptoms. Multinomial logistic regression models were employed to identify factors associated with these trajectories. RESULTS: Four distinct trajectory groups of menopausal symptoms were identified (increase, moderate, rebound, decrease). ER gene polymorphisms and E2 fluctuation were significantly associated with group membership. Furthermore, ER gene polymorphisms modulated the effect of E2 fluctuations on menopausal symptom trajectory. CONCLUSIONS: This study illuminates the multifaceted factors contributing to the individuality of the perimenopausal experience. ER gene polymorphisms emerged as integral factors by modulating the effect of E2 fluctuations on menopausal symptom trajectory. This underscores the intricate interplay of genetic factors, E2 fluctuations, and menopausal symptoms during perimenopause.


Subject(s)
Menopause , Perimenopause , Female , Humans , Switzerland , Menopause/genetics , Estradiol , Polymorphism, Single Nucleotide , Estrogen Receptor alpha/genetics
13.
PLoS One ; 19(2): e0293540, 2024.
Article in English | MEDLINE | ID: mdl-38324609

ABSTRACT

OBJECTIVES: To determine whether the age at menarche (AAM) and the age at menopause (ANM) are causally related to the development of sepsis. METHODS: We performed a two-sample Mendelian randomization (MR) analysis by utilizing summary statistics from genome-wide association study (GWAS) datasets for both the exposure and outcome variables. Single nucleotide polymorphisms (SNPs) that exhibited significant associations with AAM and ANM were chosen as instrumental variables to estimate the causal effects on sepsis. Our study employed a variety of methods, including MR-Egger regression, weighted median estimation, inverse variance weighting, a simple model, and a weighted model. Odds ratios (ORs) along with their corresponding 95% confidence intervals (CIs) were used as the primary indicators for assessing causality. Furthermore, we conducted sensitivity analyses to explore the presence of genetic heterogeneity and validate the robustness of the tools employed. RESULT: Our analysis revealed a significant negative causal relationship between AAM and the risk of sepsis (IVW: OR = 0.870, 95% CI = 0.793-0.955, P = 0.003). However, our Mendelian randomization (MR) analysis did not yield sufficient evidence to support a causal link between ANM and sepsis (IVW: OR = 0.987, 95% CI = 0.971-1.004, P = 0.129). CONCLUSIONS: Our findings suggest that an earlier AAM may be associated with an increased risk of sepsis. However, we did not find sufficient evidence to support a causal relationship between ANM and sepsis.


Subject(s)
Mendelian Randomization Analysis , Sepsis , Female , Humans , Genome-Wide Association Study , Menarche/genetics , Menopause/genetics , Sepsis/genetics
14.
BMC Womens Health ; 24(1): 4, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38166892

ABSTRACT

BACKGROUND: Menopausal status has a known relationship with the levels of estrogen, progesterone, and other sex hormones, potentially influencing the activity of ER, PR, and many other signaling pathways involved in the initiation and progression of breast cancer. However, the differences between premenopausal and postmenopausal breast cancer patients at the molecular level are unclear. METHODS: We retrieved eight datasets from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) associated with menopausal status in breast cancer patients were identified using the MAMA and LIMMA methods. Based on these validated DEGs, we performed Gene Ontology (GO) functional enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Protein-protein interaction (PPI) networks were constructed. We used DrugBank data to investigate which of these validated DEGs are targetable. Survival analysis was performed to explore the influence of these genes on breast cancer patient prognosis. RESULTS: We identified 762 DEGs associated with menopausal status in breast cancer patients. PPI network analysis indicated that these genes are primarily involved in pathways such as the cell cycle, oocyte meiosis and progesterone-mediated oocyte maturation pathways. Notably, several genes played roles in multiple signaling pathways and were associated with patient survival. These genes were also observed to be targetable according to the DrugBank database. CONCLUSION: We identified DEGs associated with menopausal status in breast cancer patients. The association of these genes with several key pathways may promote understanding of the complex characterizations of breast cancer. Our findings offer valuable insights for developing new therapeutic strategies tailored to the menopausal status of breast cancer patients.


Subject(s)
Breast Neoplasms , Menopause , Female , Humans , Algorithms , Breast Neoplasms/genetics , Gene Expression Profiling/methods , Menopause/genetics , Progesterone
15.
Commun Biol ; 7(1): 47, 2024 01 06.
Article in English | MEDLINE | ID: mdl-38184718

ABSTRACT

Age at menarche (AAM) and age at natural menopause (ANM) are highly heritable traits and have been linked to various health outcomes. We aimed to identify circulating proteins associated with altered ANM and AAM using an unbiased two-sample Mendelian randomization (MR) and colocalization approach. By testing causal effects of 1,271 proteins on AAM, we identified 22 proteins causally associated with AAM in MR, among which 13 proteins (GCKR, FOXO3, SEMA3G, PATE4, AZGP1, NEGR1, LHB, DLK1, ANXA2, YWHAB, DNAJB12, RMDN1 and HPGDS) colocalized. Among 1,349 proteins tested for causal association with ANM using MR, we identified 19 causal proteins among which 7 proteins (CPNE1, TYMP, DNER, ADAMTS13, LCT, ARL and PLXNA1) colocalized. Follow-up pathway and gene enrichment analyses demonstrated links between AAM-related proteins and obesity and diabetes, and between AAM and ANM-related proteins and various types of cancer. In conclusion, we identified proteomic signatures of reproductive ageing in women, highlighting biological processes at both ends of the reproductive lifespan.


Subject(s)
Menarche , Mendelian Randomization Analysis , Humans , Female , Menarche/genetics , Proteomics , Biomarkers , Menopause/genetics , HSP40 Heat-Shock Proteins
16.
Am J Obstet Gynecol ; 230(4): 438.e1-438.e15, 2024 04.
Article in English | MEDLINE | ID: mdl-38191017

ABSTRACT

BACKGROUND: Although phenotypic associations between female reproductive characteristics and uterine leiomyomata have long been observed in epidemiologic investigations, the shared genetic architecture underlying these complex phenotypes remains unclear. OBJECTIVE: We aimed to investigate the shared genetic basis, pleiotropic effects, and potential causal relationships underlying reproductive traits (age at menarche, age at natural menopause, and age at first birth) and uterine leiomyomata. STUDY DESIGN: With the use of large-scale, genome-wide association studies conducted among women of European ancestry for age at menarche (n=329,345), age at natural menopause (n=201,323), age at first birth (n=418,758), and uterine leiomyomata (ncases/ncontrols=35,474/267,505), we performed a comprehensive, genome-wide, cross-trait analysis to examine systematically the common genetic influences between reproductive traits and uterine leiomyomata. RESULTS: Significant global genetic correlations were identified between uterine leiomyomata and age at menarche (rg, -0.17; P=3.65×10-10), age at natural menopause (rg, 0.23; P=3.26×10-07), and age at first birth (rg, -0.16; P=1.96×10-06). Thirteen genomic regions were further revealed as contributing significant local correlations (P<.05/2353) to age at natural menopause and uterine leiomyomata. A cross-trait meta-analysis identified 23 shared loci, 3 of which were novel. A transcriptome-wide association study found 15 shared genes that target tissues of the digestive, exo- or endocrine, nervous, and cardiovascular systems. Mendelian randomization suggested causal relationships between a genetically predicted older age at menarche (odds ratio, 0.88; 95% confidence interval, 0.85-0.92; P=1.50×10-10) or older age at first birth (odds ratio, 0.95; 95% confidence interval, 0.90-0.99; P=.02) and a reduced risk for uterine leiomyomata and between a genetically predicted older age at natural menopause and an increased risk for uterine leiomyomata (odds ratio, 1.08; 95% confidence interval, 1.06-1.09; P=2.30×10-27). No causal association in the reverse direction was found. CONCLUSION: Our work highlights that there are substantial shared genetic influences and putative causal links that underlie reproductive traits and uterine leiomyomata. The findings suggest that early identification of female reproductive risk factors may facilitate the initiation of strategies to modify potential uterine leiomyomata risk.


Subject(s)
Genome-Wide Association Study , Leiomyoma , Female , Humans , Phenotype , Menopause/genetics , Risk Factors , Leiomyoma/epidemiology , Leiomyoma/genetics
18.
Epigenomics ; 16(3): 175-188, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38131149

ABSTRACT

People with ovaries experience reproductive aging as their reproductive function and system declines. This has significant implications for both fertility and long-term health, with people experiencing an increased risk of cardiometabolic disorders after menopause. Reproductive aging can be assessed through markers of ovarian reserve, response to fertility treatment or molecular biomarkers, including DNA methylation. Changes in DNA methylation with age associate with poorer reproductive outcomes, and epigenome-wide studies can provide insight into genes and pathways involved. DNA methylation-based epigenetic clocks can quantify biological age in reproductive tissues and systemically. This review provides an overview of hallmarks and theories of aging in the context of the reproductive system, and then focuses on studies of DNA methylation in reproductive tissues.


People with ovaries experience a natural decline in the function of their reproductive system as they age. This decline eventually leads to menopause, and after menopause, people have an increased risk of developing cardiovascular or other chronic diseases. In the clinic, it is hard to measure aging of the reproductive system, so other markers of the ovary's function, like the number of remaining eggs, are used. We can also measure reproductive aging using molecular biomarkers, which can help us determine when a person's molecular age is different from their chronological age. This review focuses on an overview of biological processes and theories associated with aging, and then focuses on what can be learned from molecular biomarkers.


Subject(s)
Aging , DNA Methylation , Female , Humans , Aging/genetics , Reproduction/genetics , Menopause/genetics , Ovary , Epigenesis, Genetic
19.
Mol Biol Rep ; 51(1): 6, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38085363

ABSTRACT

PURPOSE: We aimed to compare the expression levels of anti-apoptotic and proapoptotic genes in the parametrium, sacrouterine and round ligaments with respect to menopausal status in women presenting without any indication of pelvic organ prolapse (POP). We hypothesized that apoptosis related gene expressions in female pelvic tissues may be altered during menopause. METHODS: The study groups consisted of pre-menopausal (n = 10) and menopausal (n = 10) females who did not have POP symptoms. Three different types of tissue samples (Parametrium, Round Ligament and Sacrouterine Ligament) were obtained and RNA was isolated from these tissues. After purifying and quantifying RNA samples, qPCR was used to determine the expression levels of anti-apoptotic and pro-apoptotic genes. RESULTS: BCL-2 gene expression levels were significantly lower in all the tissues of menopausal patients compared to those of premenopausal patients. In comparison to premenopausal patients, the sacrouterine ligament tissue BAD expression level was significantly high (p = 0.035), and the BCL-2/BAD ratio was significantly lower in menopausal patients (p = 0.006). CONCLUSION: Apoptosis-related protein levels change during menopause; pro-apoptotic gene expressions decrease and anti-apoptotic gene expressions increase. The significant alteration of BCL-2 and BAD expression in sacrouterine ligament with respect to menopausal status was observed and this suggested that when compared to other pelvic tissues, the sacrouterine ligament, which plays a crucial role for genital organs in restoring normal pelvic anatomy and providing support, could be affected more by menopause.


Subject(s)
Menopause , Proto-Oncogene Proteins c-bcl-2 , Female , Humans , Proto-Oncogene Proteins c-bcl-2/genetics , Menopause/genetics , Premenopause/metabolism , Apoptosis/genetics , RNA
20.
BMC Med Genomics ; 16(1): 231, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37784116

ABSTRACT

BACKGROUND: Vasomotor symptoms (VMS) can often significantly impact women's quality of life at menopause. In vivo studies have shown that increased neurokinin B (NKB) / neurokinin 3 receptor (NK3R) signalling contributes to VMS, with previous genetic studies implicating the TACR3 gene locus that encodes NK3R. Large-scale genomic analyses offer the possibility of biological insights but few such studies have collected data on VMS, while proxy phenotypes such as hormone replacement therapy (HRT) use are likely to be affected by changes in clinical practice. We investigated the genetic basis of VMS by analysing routinely-collected health records. METHODS: We performed a GWAS of VMS derived from linked primary-care records and cross-sectional self-reported HRT use in up to 153,152 women from UK Biobank, a population-based cohort. In a subset of this cohort (n = 39,356), we analysed exome-sequencing data to test the association with VMS of rare deleterious genetic variants. Finally, we used Mendelian randomisation analysis to investigate the reasons for HRT use over time. RESULTS: Our GWAS of health-records derived VMS identified a genetic signal near TACR3 associated with a lower risk of VMS (OR=0.76 (95% CI 0.72,0.80) per A allele, P=3.7x10-27), which was consistent with previous studies, validating this approach. Conditional analyses demonstrated independence of genetic signals for puberty timing and VMS at the TACR3 locus, including a rare variant predicted to reduce functional NK3R levels that was associated with later menarche (P = 5 × 10-9) but showed no association with VMS (P = 0.6). Younger menopause age was causally-associated with greater HRT use before 2002 but not after. CONCLUSIONS: We provide support for TACR3 in the genetic basis of VMS but unexpectedly find that rare genomic variants predicted to lower NK3R levels did not modify VMS, despite the proven efficacy of NK3R antagonists. Using genomics we demonstrate changes in genetic associations with HRT use over time, arising from a change in clinical practice since the early 2000s, which is likely to reflect a switch from preventing post-menopausal complications in women with earlier menopause to primarily treating VMS. Our study demonstrates that integrating routinely-collected primary care health records and genomic data offers great potential for exploring the genetic basis of symptoms.


Subject(s)
Genome-Wide Association Study , Hot Flashes , Female , Humans , Hot Flashes/genetics , Quality of Life , Cross-Sectional Studies , Menopause/genetics , Primary Health Care
SELECTION OF CITATIONS
SEARCH DETAIL