Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Genes Chromosomes Cancer ; 62(10): 589-596, 2023 10.
Article in English | MEDLINE | ID: mdl-37222498

ABSTRACT

BACKGROUND: Approximately half of ovarian tumors have defects within the homologous recombination repair pathway. Tumors carrying pathogenic variants (PVs) in BRCA1/BRCA2 are more likely to respond to poly-ADP ribose polymerase (PARP) inhibitor treatment. Large rearrangements (LRs) are a challenging class of variants to identify and characterize in tumor specimens and may therefore be underreported. This study describes the prevalence of pathogenic BRCA1/BRCA2 LRs in ovarian tumors and discusses the importance of their identification using a comprehensive testing strategy. METHODS: Sequencing and LR analyses of BRCA1/BRCA2 were conducted in 20 692 ovarian tumors received between March 18, 2016 and February 14, 2023 for MyChoice CDx testing. MyChoice CDx uses NGS dosage analysis to detect LRs in BRCA1/BRCA2 genes using dense tiling throughout the coding regions and limited flanking regions. RESULTS: Of the 2217 PVs detected, 6.3% (N = 140) were LRs. Overall, 0.67% of tumors analyzed carried a pathogenic LR. The majority of detected LRs were deletions (89.3%), followed by complex LRs (5.7%), duplications (4.3%), and retroelement insertions (0.7%). Notably, 25% of detected LRs encompassed a single or partial single exon. This study identified 84 unique LRs, 2 samples each carried 2 unique LRs in the same gene. We identified 17 LRs that occurred in multiple samples, some of which were specific to certain ancestries. Several cases presented here illustrate the intricacies involved in characterizing LRs, particularly when multiple events occur within the same gene. CONCLUSIONS: Over 6% of PVs detected in the ovarian tumors analyzed were LRs. It is imperative for laboratories to utilize testing methodologies that will accurately detect LRs at a single exon resolution to optimize the identification of patients who may benefit from PARP inhibitor treatment.


Subject(s)
Breast Neoplasms , Ovarian Neoplasms , Female , Humans , BRCA1 Protein/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , BRCA2 Protein/genetics , Genes, BRCA2 , Gene Rearrangement , DNA Repair , High-Throughput Nucleotide Sequencing , Breast Neoplasms/genetics , Germ-Line Mutation
2.
Gynecol Oncol ; 163(3): 563-568, 2021 12.
Article in English | MEDLINE | ID: mdl-34742578

ABSTRACT

OBJECTIVE: Maintenance olaparib provided a progression-free survival benefit in the phase III SOLO2 trial (NCT01874353) in patients with platinum-sensitive relapsed ovarian cancer and a BRCA mutation (BRCAm). However, questions remain regarding tumor versus germline BRCA testing and the impact of heterozygous versus bi-allelic loss of BRCA1 or BRCA2 in the tumor. METHODS: Blood and tumor samples were analyzed. A concordance analysis of germline BRCAm status (BRACAnalysis® CLIA test) and tumor BRCAm status (myChoice® CDx test) was conducted (Myriad Genetic Laboratories, Inc.). Bi-allelic loss of BRCA1 and BRCA2 and a genomic instability score (GIS) (myChoice® CDx test) were also determined. RESULTS: 289 of 295 enrolled patients had a germline BRCAm confirmed centrally and tumor BRCAm status was evaluable in 241 patients. There was 98% and 100% concordance between tumor and germline testing for BRCA1m and BRCA2m, respectively, with discordance found in four cases. Of 210 tumor samples evaluable for BRCA zygosity, 100% of germline BRCA1-mutated tumors (n = 144) and 98% of germline BRCA2-mutated tumors (n = 66) had bi-allelic loss of BRCA. One patient with a heterozygous BRCA2m had a GIS of 53, was progression free for 911 days and remained on olaparib at data cut-off. CONCLUSIONS: Very high concordance was demonstrated between tumor and germline BRCA testing, supporting wider implementation of tumor BRCA testing in ovarian cancer. Near 100% rates of bi-allelic loss of BRCA in platinum-sensitive relapsed ovarian tumors suggest routine testing for BRCA zygosity is not required in this population and reflects BRCA loss being a driver of tumorigenesis.


Subject(s)
BRCA2 Protein/genetics , Germ-Line Mutation , Loss of Heterozygosity , Ovarian Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Antineoplastic Agents/therapeutic use , BRCA2 Protein/blood , Carcinoma, Endometrioid/blood , Carcinoma, Endometrioid/genetics , Clinical Trials, Phase III as Topic , Fallopian Tube Neoplasms/blood , Fallopian Tube Neoplasms/genetics , Female , Humans , Ovarian Neoplasms/blood , Peritoneal Neoplasms/blood , Peritoneal Neoplasms/genetics , Phthalazines/therapeutic use , Piperazines/therapeutic use , Polymorphism, Single Nucleotide , Randomized Controlled Trials as Topic , Ubiquitin-Protein Ligases/blood
3.
Sci Adv ; 5(9): eaaw3095, 2019 09.
Article in English | MEDLINE | ID: mdl-31840077

ABSTRACT

Early childhood growth patterns are associated with adult health, yet the genetic factors and the developmental stages involved are not fully understood. Here, we combine genome-wide association studies with modeling of longitudinal growth traits to study the genetics of infant and child growth, followed by functional, pathway, genetic correlation, risk score, and colocalization analyses to determine how developmental timings, molecular pathways, and genetic determinants of these traits overlap with those of adult health. We found a robust overlap between the genetics of child and adult body mass index (BMI), with variants associated with adult BMI acting as early as 4 to 6 years old. However, we demonstrated a completely distinct genetic makeup for peak BMI during infancy, influenced by variation at the LEPR/LEPROT locus. These findings suggest that different genetic factors control infant and child BMI. In light of the obesity epidemic, these findings are important to inform the timing and targets of prevention strategies.


Subject(s)
Body Mass Index , Genetic Association Studies , Genome-Wide Association Study , Quantitative Trait Loci , Quantitative Trait, Heritable , Adaptor Proteins, Signal Transducing/genetics , Adult , Child , Female , Genetic Predisposition to Disease , Genomics , Growth Charts , Humans , Infant , Intracellular Signaling Peptides and Proteins , Longitudinal Studies , Male , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Receptors, Leptin/genetics
4.
PLoS Med ; 14(9): e1002383, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28898252

ABSTRACT

BACKGROUND: Glycated hemoglobin (HbA1c) is used to diagnose type 2 diabetes (T2D) and assess glycemic control in patients with diabetes. Previous genome-wide association studies (GWAS) have identified 18 HbA1c-associated genetic variants. These variants proved to be classifiable by their likely biological action as erythrocytic (also associated with erythrocyte traits) or glycemic (associated with other glucose-related traits). In this study, we tested the hypotheses that, in a very large scale GWAS, we would identify more genetic variants associated with HbA1c and that HbA1c variants implicated in erythrocytic biology would affect the diagnostic accuracy of HbA1c. We therefore expanded the number of HbA1c-associated loci and tested the effect of genetic risk-scores comprised of erythrocytic or glycemic variants on incident diabetes prediction and on prevalent diabetes screening performance. Throughout this multiancestry study, we kept a focus on interancestry differences in HbA1c genetics performance that might influence race-ancestry differences in health outcomes. METHODS & FINDINGS: Using genome-wide association meta-analyses in up to 159,940 individuals from 82 cohorts of European, African, East Asian, and South Asian ancestry, we identified 60 common genetic variants associated with HbA1c. We classified variants as implicated in glycemic, erythrocytic, or unclassified biology and tested whether additive genetic scores of erythrocytic variants (GS-E) or glycemic variants (GS-G) were associated with higher T2D incidence in multiethnic longitudinal cohorts (N = 33,241). Nineteen glycemic and 22 erythrocytic variants were associated with HbA1c at genome-wide significance. GS-G was associated with higher T2D risk (incidence OR = 1.05, 95% CI 1.04-1.06, per HbA1c-raising allele, p = 3 × 10-29); whereas GS-E was not (OR = 1.00, 95% CI 0.99-1.01, p = 0.60). In Europeans and Asians, erythrocytic variants in aggregate had only modest effects on the diagnostic accuracy of HbA1c. Yet, in African Americans, the X-linked G6PD G202A variant (T-allele frequency 11%) was associated with an absolute decrease in HbA1c of 0.81%-units (95% CI 0.66-0.96) per allele in hemizygous men, and 0.68%-units (95% CI 0.38-0.97) in homozygous women. The G6PD variant may cause approximately 2% (N = 0.65 million, 95% CI 0.55-0.74) of African American adults with T2D to remain undiagnosed when screened with HbA1c. Limitations include the smaller sample sizes for non-European ancestries and the inability to classify approximately one-third of the variants. Further studies in large multiethnic cohorts with HbA1c, glycemic, and erythrocytic traits are required to better determine the biological action of the unclassified variants. CONCLUSIONS: As G6PD deficiency can be clinically silent until illness strikes, we recommend investigation of the possible benefits of screening for the G6PD genotype along with using HbA1c to diagnose T2D in populations of African ancestry or groups where G6PD deficiency is common. Screening with direct glucose measurements, or genetically-informed HbA1c diagnostic thresholds in people with G6PD deficiency, may be required to avoid missed or delayed diagnoses.


Subject(s)
Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/genetics , Genetic Variation , Genome-Wide Association Study , Glycated Hemoglobin/genetics , Diabetes Mellitus, Type 2/epidemiology , Glycated Hemoglobin/metabolism , Humans , Phenotype , Risk
5.
Eur J Clin Pharmacol ; 73(6): 717-726, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28283692

ABSTRACT

PURPOSE: Emerging data on selumetinib, a MEK1/2 inhibitor in clinical development, suggest a possible difference in pharmacokinetics (PK) between Japanese and Western patients. This pooled analysis sought to assess the effect of ethnicity on selumetinib exposure in healthy Western and Asian subjects, and to identify any association between genetic variants in the UGT1A1, CYP2C19 and ABCG2 genes and observed differences in selumetinib PK. METHODS: A pooled analysis of data from ten Phase I studies, one in Asian subjects (encompassing Japanese, non-Japanese Asian and Indian Asian subjects) and nine in Western subjects, was conducted. Key findings were derived from the collective exposure data across doses of 25, 35, 50 and 75 mg selumetinib; primary variables were dose-normalized AUC and Cmax. RESULTS: PK data from 308 subjects (10 studies) were available for the pooled analysis; genetic data from 87 subjects (3 studies) were available for the pharmacogenetic analysis. Dose-normalized AUC and Cmax were 35% (95% CI: 25-47%) and 39% (95% CI: 24-56%) higher in the pooled Asian group, respectively, compared with Western subjects. PK exposure parameters were similar between the Japanese, non-Japanese Asian and Indian groups. There was no evidence that the polymorphisms assessed in the genes UGT1A1, CYP2C19 and ABCG2 account for observed PK differences. CONCLUSIONS: Selumetinib exposure was higher in healthy Asian subjects compared with Western subjects, and these data provide valuable insight for clinicians to consider when treating patients of Asian ethnicity with selumetinib.


Subject(s)
Benzimidazoles/pharmacokinetics , Pharmacogenetics , Protein Kinase Inhibitors/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Area Under Curve , Asian People , Black People , Clinical Trials, Phase I as Topic , Cytochrome P-450 CYP2C19/genetics , Glucuronosyltransferase/genetics , Humans , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , Neoplasm Proteins/genetics , White People
6.
Genome Biol ; 17(1): 255, 2016 12 12.
Article in English | MEDLINE | ID: mdl-27955697

ABSTRACT

BACKGROUND: Chronic low-grade inflammation reflects a subclinical immune response implicated in the pathogenesis of complex diseases. Identifying genetic loci where DNA methylation is associated with chronic low-grade inflammation may reveal novel pathways or therapeutic targets for inflammation. RESULTS: We performed a meta-analysis of epigenome-wide association studies (EWAS) of serum C-reactive protein (CRP), which is a sensitive marker of low-grade inflammation, in a large European population (n = 8863) and trans-ethnic replication in African Americans (n = 4111). We found differential methylation at 218 CpG sites to be associated with CRP (P < 1.15 × 10-7) in the discovery panel of European ancestry and replicated (P < 2.29 × 10-4) 58 CpG sites (45 unique loci) among African Americans. To further characterize the molecular and clinical relevance of the findings, we examined the association with gene expression, genetic sequence variants, and clinical outcomes. DNA methylation at nine (16%) CpG sites was associated with whole blood gene expression in cis (P < 8.47 × 10-5), ten (17%) CpG sites were associated with a nearby genetic variant (P < 2.50 × 10-3), and 51 (88%) were also associated with at least one related cardiometabolic entity (P < 9.58 × 10-5). An additive weighted score of replicated CpG sites accounted for up to 6% inter-individual variation (R2) of age-adjusted and sex-adjusted CRP, independent of known CRP-related genetic variants. CONCLUSION: We have completed an EWAS of chronic low-grade inflammation and identified many novel genetic loci underlying inflammation that may serve as targets for the development of novel therapeutic interventions for inflammation.


Subject(s)
C-Reactive Protein/genetics , Epigenesis, Genetic , Inflammation/genetics , Quantitative Trait Loci/genetics , Black or African American , CpG Islands/genetics , DNA Methylation/genetics , Female , Gene Expression , Genetic Variation , Genome-Wide Association Study , Humans , Inflammation/blood , Male , Nucleotide Motifs/genetics , White People
7.
Circ Cardiovasc Genet ; 9(5): 436-447, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27651444

ABSTRACT

BACKGROUND: DNA methylation leaves a long-term signature of smoking exposure and is one potential mechanism by which tobacco exposure predisposes to adverse health outcomes, such as cancers, osteoporosis, lung, and cardiovascular disorders. METHODS AND RESULTS: To comprehensively determine the association between cigarette smoking and DNA methylation, we conducted a meta-analysis of genome-wide DNA methylation assessed using the Illumina BeadChip 450K array on 15 907 blood-derived DNA samples from participants in 16 cohorts (including 2433 current, 6518 former, and 6956 never smokers). Comparing current versus never smokers, 2623 cytosine-phosphate-guanine sites (CpGs), annotated to 1405 genes, were statistically significantly differentially methylated at Bonferroni threshold of P<1×10-7 (18 760 CpGs at false discovery rate <0.05). Genes annotated to these CpGs were enriched for associations with several smoking-related traits in genome-wide studies including pulmonary function, cancers, inflammatory diseases, and heart disease. Comparing former versus never smokers, 185 of the CpGs that differed between current and never smokers were significant P<1×10-7 (2623 CpGs at false discovery rate <0.05), indicating a pattern of persistent altered methylation, with attenuation, after smoking cessation. Transcriptomic integration identified effects on gene expression at many differentially methylated CpGs. CONCLUSIONS: Cigarette smoking has a broad impact on genome-wide methylation that, at many loci, persists many years after smoking cessation. Many of the differentially methylated genes were novel genes with respect to biological effects of smoking and might represent therapeutic targets for prevention or treatment of tobacco-related diseases. Methylation at these sites could also serve as sensitive and stable biomarkers of lifetime exposure to tobacco smoke.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Smoking/adverse effects , Smoking/genetics , Transcriptome , Aged , Case-Control Studies , CpG Islands , Female , Gene Expression Profiling/methods , Genetic Markers , Genome-Wide Association Study , Genotype , Humans , Leukocytes/chemistry , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Phenotype , Smoking/ethnology , Smoking Cessation , Smoking Prevention , Time Factors
9.
Nat Genet ; 47(11): 1294-1303, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26414677

ABSTRACT

Menopause timing has a substantial impact on infertility and risk of disease, including breast cancer, but the underlying mechanisms are poorly understood. We report a dual strategy in ∼70,000 women to identify common and low-frequency protein-coding variation associated with age at natural menopause (ANM). We identified 44 regions with common variants, including two regions harboring additional rare missense alleles of large effect. We found enrichment of signals in or near genes involved in delayed puberty, highlighting the first molecular links between the onset and end of reproductive lifespan. Pathway analyses identified major association with DNA damage response (DDR) genes, including the first common coding variant in BRCA1 associated with any complex trait. Mendelian randomization analyses supported a causal effect of later ANM on breast cancer risk (∼6% increase in risk per year; P = 3 × 10(-14)), likely mediated by prolonged sex hormone exposure rather than DDR mechanisms.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , DNA Repair , Genetic Predisposition to Disease/genetics , Hypothalamus/metabolism , Signal Transduction/genetics , Adult , Age Factors , Aging/genetics , Female , Gene Regulatory Networks/genetics , Genetic Variation , Genome-Wide Association Study/methods , Genomics/methods , Genotype , Humans , Menopause/genetics , Middle Aged , Models, Genetic , Phenotype , Reproduction/genetics
10.
Nat Commun ; 6: 7756, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26239645

ABSTRACT

More than 100 loci have been identified for age at menarche by genome-wide association studies; however, collectively these explain only ∼3% of the trait variance. Here we test two overlooked sources of variation in 192,974 European ancestry women: low-frequency protein-coding variants and X-chromosome variants. Five missense/nonsense variants (in ALMS1/LAMB2/TNRC6A/TACR3/PRKAG1) are associated with age at menarche (minor allele frequencies 0.08-4.6%; effect sizes 0.08-1.25 years per allele; P<5 × 10(-8)). In addition, we identify common X-chromosome loci at IGSF1 (rs762080, P=9.4 × 10(-13)) and FAAH2 (rs5914101, P=4.9 × 10(-10)). Highlighted genes implicate cellular energy homeostasis, post-transcriptional gene silencing and fatty-acid amide signalling. A frequently reported mutation in TACR3 for idiopathic hypogonatrophic hypogonadism (p.W275X) is associated with 1.25-year-later menarche (P=2.8 × 10(-11)), illustrating the utility of population studies to estimate the penetrance of reportedly pathogenic mutations. Collectively, these novel variants explain ∼0.5% variance, indicating that these overlooked sources of variation do not substantially explain the 'missing heritability' of this complex trait.


Subject(s)
AMP-Activated Protein Kinases/genetics , Autoantigens/genetics , Immunoglobulins/genetics , Laminin/genetics , Membrane Proteins/genetics , Menarche/genetics , Proteins/genetics , RNA-Binding Proteins/genetics , Receptors, Neurokinin-3/genetics , Adolescent , Adult , Age Factors , Aged , Amides , Cell Cycle Proteins , Chromosomes, Human, X/genetics , Codon, Nonsense , Energy Metabolism/genetics , Fatty Acids , Female , Gene Frequency , Genes, X-Linked/genetics , Genetic Variation , Genotype , Humans , Hypogonadism/genetics , Middle Aged , Mutation, Missense , Penetrance , Phenotype , RNA Interference , Signal Transduction/genetics , White People/genetics , Young Adult
11.
Sci Rep ; 5: 11208, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26084728

ABSTRACT

Early puberty timing is associated with higher risks for type 2 diabetes (T2D) and cardiovascular disease in women and therefore represents a potential target for early preventive interventions. We characterised the range of diseases and other adverse health outcomes associated with early or late puberty timing in men and women in the very large UK Biobank study. Recalled puberty timing and past/current diseases were self-reported by questionnaire. We limited analyses to individuals of White ethnicity (250,037 women; 197,714 men) and to disease outcomes with at least 500 cases (~ 0.2% prevalence) and we applied stringent correction for multiple testing (corrected threshold P < 7.48 × 10(-5)). In models adjusted for socioeconomic position and adiposity/body composition variables, both in women and men separately, earlier puberty timing was associated with higher risks for angina, hypertension and T2D. Furthermore, compared to the median/average group, earlier or later puberty timing in women or men was associated with higher risks for 48 adverse outcomes, across a range of cancers, cardio-metabolic, gynaecological/obstetric, gastrointestinal, musculoskeletal, and neuro-cognitive categories. Notably, both early and late menarche were associated with higher risks for early natural menopause in women. Puberty timing in both men and women appears to have a profound impact on later health.


Subject(s)
Cardiovascular Diseases/epidemiology , Diabetes Mellitus, Type 2/epidemiology , Puberty , Adolescent , Adult , Age Factors , Biological Specimen Banks , Cardiovascular Diseases/etiology , Child , Diabetes Mellitus, Type 2/etiology , Disease Susceptibility , Female , Humans , Male , Menarche , Middle Aged , Patient Outcome Assessment , Phenotype , Risk Factors , Self Report , United Kingdom/epidemiology , Young Adult
12.
J Med Genet ; 52(6): 375-80, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25921383

ABSTRACT

BACKGROUND: In populations of European ancestry, the genetic contribution to body mass index (BMI) increases with age during childhood but then declines during adulthood, possibly due to the cumulative effects of environmental factors. How the effects of genetic factors on BMI change with age in other populations is unknown. SUBJECTS AND METHODS: In a rural Gambian population (N=2535), we used a combined allele risk score, comprising genotypes at 28 'Caucasian adult BMI-associated' single nucleotide polymorphisms (SNPs), as a marker of the genetic influence on body composition, and related this to internally-standardised z-scores for birthweight (zBW), weight-for-height (zWT-HT), weight-for-age (zWT), height-for-age (zHT), and zBMI cross-sectionally and longitudinally. RESULTS: Cross-sectionally, the genetic score was positively associated with adult zWT (0.018±0.009 per allele, p=0.034, N=1426) and zWT-HT (0.025±0.009, p=0.006), but not with size at birth or childhood zWT-HT (0.008±0.005, p=0.11, N=2211). The effect of the genetic score on zWT-HT strengthened linearly with age from birth through to late adulthood (age interaction term: 0.0083 z-scores/allele/year; 95% CI 0.0048 to 0.0118, p=0.0000032). CONCLUSIONS: Genetic variants for obesity in populations of European ancestry have direct relevance to bodyweight in nutritionally deprived African settings. In such settings, genetic obesity susceptibility appears to regulate change in weight status throughout the life course, which provides insight into its potential physiological role.


Subject(s)
Black People/genetics , Body Mass Index , Genetic Association Studies , Genetic Markers , Population Surveillance , Rural Population , Adolescent , Adult , Alleles , Child , Child, Preschool , Cross-Sectional Studies , Female , Gambia/epidemiology , Genetic Predisposition to Disease , Humans , Infant , Infant, Newborn , Male , Middle Aged , Obesity/epidemiology , Obesity/genetics , Polymorphism, Single Nucleotide , White People , Young Adult
13.
JAMA Pediatr ; 168(12): 1122-30, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25329327

ABSTRACT

IMPORTANCE: Patterns of body size and body composition associated with genetic obesity susceptibility inform the mechanisms that increase obesity risk. OBJECTIVE: To test associations between genetic obesity susceptibility, represented by a combined obesity risk-allele score, and body size or body composition at birth to age 5 years. DESIGN, SETTING, AND PARTICIPANTS: A total of 3031 children from 4 birth cohort studies in England, France, and Spain were included in a meta-analysis. EXPOSURES: A combined obesity risk-allele score was calculated from genotypes at 16 variants identified by genome-wide association studies of adult body mass index (BMI). MAIN OUTCOMES AND MEASURES: Outcomes were age- and sex-adjusted SD scores (SDS) for weight, length/height, BMI, fat mass, lean mass, and percentage of body fat at birth as well as at ages 1, 2 to 3, and 4 to 5 years. RESULTS: The obesity risk-allele score was not associated with infant size at birth; at age 1 year it was positively associated with weight (ß [SE], 0.020 [0.008] SDS per allele; P = .009) and length (ß [SE], 0.020 [0.008] SDS per allele; P = .01), but not with BMI (ß [SE], 0.013 [0.008] SDS per allele; P = .11). At age 2 to 3 years these associations were stronger (weight: ß [SE], 0.033 [0.008] SDS per allele; P < .001; height: ß [SE], 0.025 [0.008] SDS per allele; P < .001) and were also seen for BMI (ß [SE], 0.024 [0.008] SDS per allele; P = .003). The obesity risk-allele score was positively associated with both postnatal fat mass (1 year: ß [SE], 0.032 [0.017] SDS per allele; P = .05; 2-3 years: ß [SE], 0.049 [0.018] SDS per allele; P = .006; and 4-5 years: ß [SE], 0.028 [0.011] SDS per allele; P = .009) and postnatal lean mass (1 year: ß [SE], 0.038 [0.014] SDS per allele; P = .008; 2-3 years: ß [SE], 0.064 [0.017] SDS per allele; P < .001; and 4-5 years: ß [SE], 0.047 [0.011] SDS per allele; P < .001), but not with the percentage of body fat (P > .15 at all ages). CONCLUSIONS AND RELEVANCE: Genetic obesity susceptibility appears to promote a normally partitioned increase in early postnatal, but not prenatal, growth. These findings suggest that symmetrical rapid growth may identify infants with high life-long susceptibility for obesity.


Subject(s)
Adipose Tissue , Body Weight/genetics , Genetic Predisposition to Disease , Obesity/genetics , Child, Preschool , England , Female , France , Humans , Infant , Male , Risk Factors , Spain
14.
Nature ; 514(7520): 92-97, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25231870

ABSTRACT

Age at menarche is a marker of timing of puberty in females. It varies widely between individuals, is a heritable trait and is associated with risks for obesity, type 2 diabetes, cardiovascular disease, breast cancer and all-cause mortality. Studies of rare human disorders of puberty and animal models point to a complex hypothalamic-pituitary-hormonal regulation, but the mechanisms that determine pubertal timing and underlie its links to disease risk remain unclear. Here, using genome-wide and custom-genotyping arrays in up to 182,416 women of European descent from 57 studies, we found robust evidence (P < 5 × 10(-8)) for 123 signals at 106 genomic loci associated with age at menarche. Many loci were associated with other pubertal traits in both sexes, and there was substantial overlap with genes implicated in body mass index and various diseases, including rare disorders of puberty. Menarche signals were enriched in imprinted regions, with three loci (DLK1-WDR25, MKRN3-MAGEL2 and KCNK9) demonstrating parent-of-origin-specific associations concordant with known parental expression patterns. Pathway analyses implicated nuclear hormone receptors, particularly retinoic acid and γ-aminobutyric acid-B2 receptor signalling, among novel mechanisms that regulate pubertal timing in humans. Our findings suggest a genetic architecture involving at least hundreds of common variants in the coordinated timing of the pubertal transition.


Subject(s)
Alleles , Genetic Loci/genetics , Menarche/genetics , Parents , Adolescent , Age Factors , Body Mass Index , Breast Neoplasms/genetics , Calcium-Binding Proteins , Cardiovascular Diseases/genetics , Child , Diabetes Mellitus, Type 2/genetics , Europe/ethnology , Female , Genome-Wide Association Study , Genomic Imprinting/genetics , Humans , Hypothalamo-Hypophyseal System/physiology , Intercellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Obesity/genetics , Ovary/physiology , Polymorphism, Single Nucleotide/genetics , Potassium Channels, Tandem Pore Domain/genetics , Proteins/genetics , Quantitative Trait Loci/genetics , Receptors, GABA-B/metabolism , Receptors, Retinoic Acid/metabolism , Ribonucleoproteins/genetics , Ubiquitin-Protein Ligases
15.
Am J Epidemiol ; 180(1): 29-40, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24920784

ABSTRACT

We conducted a systematic review and meta-analysis to investigate the associations between menarcheal age and all-cause and cardiovascular death. Medline, Embase, Scopus, and Web of Knowledge were searched for articles published prior to March 2013 reporting on the associations between menarcheal age and death from all causes or from cardiovascular disease (total cardiovascular disease, ischemic heart disease (IHD), and stroke) in adult women. Nine articles were eligible for inclusion; these reported 5 estimates each for death from all causes and total cardiovascular death, 6 estimates for IHD, and 7 estimates for death from stroke. Our meta-analysis showed that each 1-year increase in age at menarche was associated with a 3% lower relative risk of death from all causes (pooled hazard ratio = 0.97, 95% confidence interval: 0.96, 0.98) with low heterogeneity (I(2) = 32.2%). Meta-analysis of 2 cohorts showed a higher risk of death from all causes for women who experienced early menarche (at <12 years of age) versus "not early" menarche (at ≥ 12 years of age) (pooled hazard ratio = 1.23, 95% confidence interval: 1.10, 1.38; I(2) = 0%). An inverse association between age at menarche and death from IHD was observed only among nonsmoking populations or populations with low prevalence of smoking. We found no evidence of association between age at menarche and death from all cardiovascular diseases or stroke. Early menarche was consistently associated with higher risk of death from all causes. Further studies are needed to clarify the role of menarcheal age on cardiovascular outcomes and to investigate the potential modifying role of smoking.


Subject(s)
Cardiovascular Diseases/mortality , Menarche , Mortality , Adolescent , Adult , Age Factors , Aged , Child , Female , Humans , Middle Aged , Proportional Hazards Models , Risk Factors
16.
Hum Mol Genet ; 23(9): 2490-7, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24357391

ABSTRACT

The length of female reproductive lifespan is associated with multiple adverse outcomes, including breast cancer, cardiovascular disease and infertility. The biological processes that govern the timing of the beginning and end of reproductive life are not well understood. Genetic variants are known to contribute to ∼50% of the variation in both age at menarche and menopause, but to date the known genes explain <15% of the genetic component. We have used genome-wide association in a bivariate meta-analysis of both traits to identify genes involved in determining reproductive lifespan. We observed significant genetic correlation between the two traits using genome-wide complex trait analysis. However, we found no robust statistical evidence for individual variants with an effect on both traits. A novel association with age at menopause was detected for a variant rs1800932 in the mismatch repair gene MSH6 (P = 1.9 × 10(-9)), which was also associated with altered expression levels of MSH6 mRNA in multiple tissues. This study contributes to the growing evidence that DNA repair processes play a key role in ovarian ageing and could be an important therapeutic target for infertility.


Subject(s)
DNA-Binding Proteins/metabolism , Menopause/genetics , Age Factors , DNA-Binding Proteins/genetics , Female , Genome-Wide Association Study , Humans , Menopause/physiology , Polymorphism, Single Nucleotide/genetics
18.
Diabetes Care ; 36(11): 3526-34, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24159179

ABSTRACT

OBJECTIVE: Younger age at menarche, a marker of pubertal timing in girls, is associated with higher risk of later type 2 diabetes. We aimed to confirm this association and to examine whether it is explained by adiposity. RESEARCH DESIGN AND METHODS: The prospective European Prospective Investigation into Cancer and Nutrition (EPIC)-InterAct case-cohort study consists of 12,403 incident type 2 diabetes cases and a stratified subcohort of 16,154 individuals from 26 research centers across eight European countries. We tested the association between age at menarche and incident type 2 diabetes using Prentice-weighted Cox regression in 15,168 women (n = 5,995 cases). Models were adjusted in a sequential manner for potential confounding and mediating factors, including adult BMI. RESULTS: Mean menarcheal age ranged from 12.6 to 13.6 years across InterAct countries. Each year later menarche was associated with 0.32 kg/m2 lower adult BMI. Women in the earliest menarche quintile (8-11 years, n = 2,418) had 70% higher incidence of type 2 diabetes compared with those in the middle quintile (13 years, n = 3,634), adjusting for age at recruitment, research center, and a range of lifestyle and reproductive factors (hazard ratio [HR], 1.70; 95% CI, 1.49-1.94; P < 0.001). Adjustment for BMI partially attenuated this association (HR, 1.42; 95% CI, 1.18-1.71; P < 0.001). Later menarche beyond the median age was not protective against type 2 diabetes. CONCLUSIONS: Women with history of early menarche have higher risk of type 2 diabetes in adulthood. Less than half of this association appears to be mediated by higher adult BMI, suggesting that early pubertal development also may directly increase type 2 diabetes risk.


Subject(s)
Diabetes Mellitus, Type 2/epidemiology , Menarche/physiology , Adiposity , Adult , Age Factors , Europe/epidemiology , Female , Humans , Incidence , Life Style , Longitudinal Studies , Middle Aged , Neoplasms/complications , Obesity/epidemiology , Prospective Studies , Risk , Risk Factors
19.
PLoS Genet ; 9(7): e1003607, 2013.
Article in English | MEDLINE | ID: mdl-23935507

ABSTRACT

Numerous obesity loci have been identified using genome-wide association studies. A UK study indicated that physical activity may attenuate the cumulative effect of 12 of these loci, but replication studies are lacking. Therefore, we tested whether the aggregate effect of these loci is diminished in adults of European ancestry reporting high levels of physical activity. Twelve obesity-susceptibility loci were genotyped or imputed in 111,421 participants. A genetic risk score (GRS) was calculated by summing the BMI-associated alleles of each genetic variant. Physical activity was assessed using self-administered questionnaires. Multiplicative interactions between the GRS and physical activity on BMI were tested in linear and logistic regression models in each cohort, with adjustment for age, age(2), sex, study center (for multicenter studies), and the marginal terms for physical activity and the GRS. These results were combined using meta-analysis weighted by cohort sample size. The meta-analysis yielded a statistically significant GRS × physical activity interaction effect estimate (Pinteraction  = 0.015). However, a statistically significant interaction effect was only apparent in North American cohorts (n = 39,810, Pinteraction  = 0.014 vs. n = 71,611, Pinteraction  = 0.275 for Europeans). In secondary analyses, both the FTO rs1121980 (Pinteraction  = 0.003) and the SEC16B rs10913469 (Pinteraction  = 0.025) variants showed evidence of SNP × physical activity interactions. This meta-analysis of 111,421 individuals provides further support for an interaction between physical activity and a GRS in obesity disposition, although these findings hinge on the inclusion of cohorts from North America, indicating that these results are either population-specific or non-causal.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Motor Activity/genetics , Obesity/genetics , Adult , Alleles , Body Mass Index , Female , Humans , Logistic Models , Male , Obesity/epidemiology , Polymorphism, Single Nucleotide , Risk Factors , Surveys and Questionnaires , White People/genetics
20.
Am J Epidemiol ; 178(3): 451-60, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23558354

ABSTRACT

Obesity is of global health concern. There are well-described inverse relationships between female pubertal timing and obesity. Recent genome-wide association studies of age at menarche identified several obesity-related variants. Using data from the ReproGen Consortium, we employed meta-analytical techniques to estimate the associations of 95 a priori and recently identified obesity-related (body mass index (weight (kg)/height (m)(2)), waist circumference, and waist:hip ratio) single-nucleotide polymorphisms (SNPs) with age at menarche in 92,116 women of European descent from 38 studies (1970-2010), in order to estimate associations between genetic variants associated with central or overall adiposity and pubertal timing in girls. Investigators in each study performed a separate analysis of associations between the selected SNPs and age at menarche (ages 9-17 years) using linear regression models and adjusting for birth year, site (as appropriate), and population stratification. Heterogeneity of effect-measure estimates was investigated using meta-regression. Six novel associations of body mass index loci with age at menarche were identified, and 11 adiposity loci previously reported to be associated with age at menarche were confirmed, but none of the central adiposity variants individually showed significant associations. These findings suggest complex genetic relationships between menarche and overall obesity, and to a lesser extent central obesity, in normal processes of growth and development.


Subject(s)
Adiposity/genetics , Menarche/genetics , Obesity/epidemiology , Obesity/genetics , Polymorphism, Single Nucleotide , White People/genetics , Adolescent , Age Factors , Body Mass Index , Child , Female , Genetic Association Studies , Humans , Linkage Disequilibrium , Waist Circumference , Waist-Hip Ratio , Women's Health/statistics & numerical data
SELECTION OF CITATIONS
SEARCH DETAIL
...