Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Publication year range
1.
Am J Hum Genet ; 107(4): 670-682, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32910913

ABSTRACT

Exome sequencing in diabetes presents a diagnostic challenge because depending on frequency, functional impact, and genomic and environmental contexts, HNF1A variants can cause maturity-onset diabetes of the young (MODY), increase type 2 diabetes risk, or be benign. A correct diagnosis matters as it informs on treatment, progression, and family risk. We describe a multi-dimensional functional dataset of 73 HNF1A missense variants identified in exomes of 12,940 individuals. Our aim was to develop an analytical framework for stratifying variants along the HNF1A phenotypic continuum to facilitate diagnostic interpretation. HNF1A variant function was determined by four different molecular assays. Structure of the multi-dimensional dataset was explored using principal component analysis, k-means, and hierarchical clustering. Weights for tissue-specific isoform expression and functional domain were integrated. Functionally annotated variant subgroups were used to re-evaluate genetic diagnoses in national MODY diagnostic registries. HNF1A variants demonstrated a range of behaviors across the assays. The structure of the multi-parametric data was shaped primarily by transactivation. Using unsupervised learning methods, we obtained high-resolution functional clusters of the variants that separated known causal MODY variants from benign and type 2 diabetes risk variants and led to reclassification of 4% and 9% of HNF1A variants identified in the UK and Norway MODY diagnostic registries, respectively. Our proof-of-principle analyses facilitated informative stratification of HNF1A variants along the continuum, allowing improved evaluation of clinical significance, management, and precision medicine in diabetes clinics. Transcriptional activity appears a superior readout supporting pursuit of transactivation-centric experimental designs for high-throughput functional screens.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Hepatocyte Nuclear Factor 1-alpha/genetics , Mutation, Missense , Registries , Unsupervised Machine Learning , Adolescent , Adult , Alleles , Child , Cluster Analysis , Datasets as Topic , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/pathology , Female , Gene Expression , Humans , Male , Norway/epidemiology , Phenotype , Principal Component Analysis , United Kingdom/epidemiology , Exome Sequencing , Young Adult
2.
Nature ; 538(7624): 248-252, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27680694

ABSTRACT

Birth weight (BW) has been shown to be influenced by both fetal and maternal factors and in observational studies is reproducibly associated with future risk of adult metabolic diseases including type 2 diabetes (T2D) and cardiovascular disease. These life-course associations have often been attributed to the impact of an adverse early life environment. Here, we performed a multi-ancestry genome-wide association study (GWAS) meta-analysis of BW in 153,781 individuals, identifying 60 loci where fetal genotype was associated with BW (P < 5 × 10-8). Overall, approximately 15% of variance in BW was captured by assays of fetal genetic variation. Using genetic association alone, we found strong inverse genetic correlations between BW and systolic blood pressure (Rg = -0.22, P = 5.5 × 10-13), T2D (Rg = -0.27, P = 1.1 × 10-6) and coronary artery disease (Rg = -0.30, P = 6.5 × 10-9). In addition, using large -cohort datasets, we demonstrated that genetic factors were the major contributor to the negative covariance between BW and future cardiometabolic risk. Pathway analyses indicated that the protein products of genes within BW-associated regions were enriched for diverse processes including insulin signalling, glucose homeostasis, glycogen biosynthesis and chromatin remodelling. There was also enrichment of associations with BW in known imprinted regions (P = 1.9 × 10-4). We demonstrate that life-course associations between early growth phenotypes and adult cardiometabolic disease are in part the result of shared genetic effects and identify some of the pathways through which these causal genetic effects are mediated.


Subject(s)
Aging/genetics , Birth Weight/genetics , Coronary Artery Disease/genetics , Diabetes Mellitus, Type 2/genetics , Fetus/metabolism , Genetic Predisposition to Disease , Genome-Wide Association Study , Adult , Anthropometry , Blood Pressure/genetics , Chromatin Assembly and Disassembly , Cohort Studies , Datasets as Topic , Female , Genetic Loci/genetics , Genetic Variation/genetics , Genomic Imprinting/genetics , Genotype , Glucose/metabolism , Glycogen/biosynthesis , Humans , Insulin/metabolism , Male , Phenotype , Signal Transduction
3.
PLoS Genet ; 11(12): e1005694, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26624892

ABSTRACT

The intersection of genome-wide association analyses with physiological and functional data indicates that variants regulating islet gene transcription influence type 2 diabetes (T2D) predisposition and glucose homeostasis. However, the specific genes through which these regulatory variants act remain poorly characterized. We generated expression quantitative trait locus (eQTL) data in 118 human islet samples using RNA-sequencing and high-density genotyping. We identified fourteen loci at which cis-exon-eQTL signals overlapped active islet chromatin signatures and were coincident with established T2D and/or glycemic trait associations. At some, these data provide an experimental link between GWAS signals and biological candidates, such as DGKB and ADCY5. At others, the cis-signals implicate genes with no prior connection to islet biology, including WARS and ZMIZ1. At the ZMIZ1 locus, we show that perturbation of ZMIZ1 expression in human islets and beta-cells influences exocytosis and insulin secretion, highlighting a novel role for ZMIZ1 in the maintenance of glucose homeostasis. Together, these findings provide a significant advance in the mechanistic insights of T2D and glycemic trait association loci.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Insulin/genetics , Transcription Factors/genetics , Diabetes Mellitus, Type 2/pathology , Exons , Gene Expression Regulation , Genome-Wide Association Study , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Quantitative Trait Loci/genetics , Signal Transduction , Transcription Factors/biosynthesis
4.
PLoS Genet ; 11(7): e1005230, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26132169

ABSTRACT

Reference panels from the 1000 Genomes (1000G) Project Consortium provide near complete coverage of common and low-frequency genetic variation with minor allele frequency ≥0.5% across European ancestry populations. Within the European Network for Genetic and Genomic Epidemiology (ENGAGE) Consortium, we have undertaken the first large-scale meta-analysis of genome-wide association studies (GWAS), supplemented by 1000G imputation, for four quantitative glycaemic and obesity-related traits, in up to 87,048 individuals of European ancestry. We identified two loci for body mass index (BMI) at genome-wide significance, and two for fasting glucose (FG), none of which has been previously reported in larger meta-analysis efforts to combine GWAS of European ancestry. Through conditional analysis, we also detected multiple distinct signals of association mapping to established loci for waist-hip ratio adjusted for BMI (RSPO3) and FG (GCK and G6PC2). The index variant for one association signal at the G6PC2 locus is a low-frequency coding allele, H177Y, which has recently been demonstrated to have a functional role in glucose regulation. Fine-mapping analyses revealed that the non-coding variants most likely to drive association signals at established and novel loci were enriched for overlap with enhancer elements, which for FG mapped to promoter and transcription factor binding sites in pancreatic islets, in particular. Our study demonstrates that 1000G imputation and genetic fine-mapping of common and low-frequency variant association signals at GWAS loci, integrated with genomic annotation in relevant tissues, can provide insight into the functional and regulatory mechanisms through which their effects on glycaemic and obesity-related traits are mediated.


Subject(s)
Chromosome Mapping , Genetic Predisposition to Disease , Glycemic Index/genetics , Obesity/genetics , Quantitative Trait Loci/genetics , Body Mass Index , Gene Frequency/genetics , Genome-Wide Association Study , Germinal Center Kinases , Glucose-6-Phosphatase/genetics , Humans , Polymorphism, Single Nucleotide/genetics , Protein Serine-Threonine Kinases/genetics , Thrombospondins/genetics
5.
Nature ; 464(7289): 713-20, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20360734

ABSTRACT

Copy number variants (CNVs) account for a major proportion of human genetic polymorphism and have been predicted to have an important role in genetic susceptibility to common disease. To address this we undertook a large, direct genome-wide study of association between CNVs and eight common human diseases. Using a purpose-designed array we typed approximately 19,000 individuals into distinct copy-number classes at 3,432 polymorphic CNVs, including an estimated approximately 50% of all common CNVs larger than 500 base pairs. We identified several biological artefacts that lead to false-positive associations, including systematic CNV differences between DNAs derived from blood and cell lines. Association testing and follow-up replication analyses confirmed three loci where CNVs were associated with disease-IRGM for Crohn's disease, HLA for Crohn's disease, rheumatoid arthritis and type 1 diabetes, and TSPAN8 for type 2 diabetes-although in each case the locus had previously been identified in single nucleotide polymorphism (SNP)-based studies, reflecting our observation that most common CNVs that are well-typed on our array are well tagged by SNPs and so have been indirectly explored through SNP studies. We conclude that common CNVs that can be typed on existing platforms are unlikely to contribute greatly to the genetic basis of common human diseases.


Subject(s)
DNA Copy Number Variations/genetics , Disease , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Arthritis, Rheumatoid/genetics , Case-Control Studies , Crohn Disease/genetics , Diabetes Mellitus/genetics , Gene Frequency/genetics , Humans , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Pilot Projects , Polymorphism, Single Nucleotide/genetics , Quality Control
6.
PLoS Genet ; 7(2): e1001307, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21379325

ABSTRACT

An age-dependent association between variation at the FTO locus and BMI in children has been suggested. We meta-analyzed associations between the FTO locus (rs9939609) and BMI in samples, aged from early infancy to 13 years, from 8 cohorts of European ancestry. We found a positive association between additional minor (A) alleles and BMI from 5.5 years onwards, but an inverse association below age 2.5 years. Modelling median BMI curves for each genotype using the LMS method, we found that carriers of minor alleles showed lower BMI in infancy, earlier adiposity rebound (AR), and higher BMI later in childhood. Differences by allele were consistent with two independent processes: earlier AR equivalent to accelerating developmental age by 2.37% (95% CI 1.87, 2.87, p = 10(-20)) per A allele and a positive age by genotype interaction such that BMI increased faster with age (p = 10(-23)). We also fitted a linear mixed effects model to relate genotype to the BMI curve inflection points adiposity peak (AP) in infancy and AR. Carriage of two minor alleles at rs9939609 was associated with lower BMI at AP (-0.40% (95% CI: -0.74, -0.06), p = 0.02), higher BMI at AR (0.93% (95% CI: 0.22, 1.64), p = 0.01), and earlier AR (-4.72% (-5.81, -3.63), p = 10(-17)), supporting cross-sectional results. Overall, we confirm the expected association between variation at rs9939609 and BMI in childhood, but only after an inverse association between the same variant and BMI in infancy. Patterns are consistent with a shift on the developmental scale, which is reflected in association with the timing of AR rather than just a global increase in BMI. Results provide important information about longitudinal gene effects and about the role of FTO in adiposity. The associated shifts in developmental timing have clinical importance with respect to known relationships between AR and both later-life BMI and metabolic disease risk.


Subject(s)
Body Mass Index , Genetic Association Studies , Genetic Loci/genetics , Genetic Variation , Growth and Development/genetics , Proteins/genetics , Adiposity/genetics , Adolescent , Alleles , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Body Height/genetics , Body Weight/genetics , Child , Child, Preschool , Cross-Sectional Studies , Female , Genotype , Humans , Infant , Infant, Newborn , Longitudinal Studies , Male , Meta-Analysis as Topic , Polymorphism, Single Nucleotide/genetics
7.
PLoS Genet ; 5(3): e1000409, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19266077

ABSTRACT

Recent genome-wide association (GWA) studies have identified dozens of common variants associated with adult height. However, it is unknown how these variants influence height growth during childhood. We derived peak height velocity in infancy (PHV1) and puberty (PHV2) and timing of pubertal height growth spurt from parametric growth curves fitted to longitudinal height growth data to test their association with known height variants. The study consisted of N = 3,538 singletons from the prospective Northern Finland Birth Cohort 1966 with genotype data and frequent height measurements (on average 20 measurements per person) from 0-20 years. Twenty-six of the 48 variants tested associated with adult height (p<0.05, adjusted for sex and principal components) in this sample, all in the same direction as in previous GWA scans. Seven SNPs in or near the genes HHIP, DLEU7, UQCC, SF3B4/SV2A, LCORL, and HIST1H1D associated with PHV1 and five SNPs in or near SOCS2, SF3B4/SV2A, C17orf67, CABLES1, and DOT1L with PHV2 (p<0.05). We formally tested variants for interaction with age (infancy versus puberty) and found biologically meaningful evidence for an age-dependent effect for the SNP in SOCS2 (p = 0.0030) and for the SNP in HHIP (p = 0.045). We did not have similar prior evidence for the association between height variants and timing of pubertal height growth spurt as we had for PHVs, and none of the associations were statistically significant after correction for multiple testing. The fact that in this sample, less than half of the variants associated with adult height had a measurable effect on PHV1 or PHV2 is likely to reflect limited power to detect these associations in this dataset. Our study is the first genetic association analysis on longitudinal height growth in a prospective cohort from birth to adulthood and gives grounding for future research on the genetic regulation of human height during different periods of growth.


Subject(s)
Body Height , Child Development , White People/genetics , Adolescent , Adult , Child , Female , Finland , Genome-Wide Association Study , Genotype , Humans , Infant, Newborn , Male , Polymorphism, Single Nucleotide , Prospective Studies
8.
PLoS Genet ; 5(6): e1000508, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19557161

ABSTRACT

To identify genetic loci influencing central obesity and fat distribution, we performed a meta-analysis of 16 genome-wide association studies (GWAS, N = 38,580) informative for adult waist circumference (WC) and waist-hip ratio (WHR). We selected 26 SNPs for follow-up, for which the evidence of association with measures of central adiposity (WC and/or WHR) was strong and disproportionate to that for overall adiposity or height. Follow-up studies in a maximum of 70,689 individuals identified two loci strongly associated with measures of central adiposity; these map near TFAP2B (WC, P = 1.9x10(-11)) and MSRA (WC, P = 8.9x10(-9)). A third locus, near LYPLAL1, was associated with WHR in women only (P = 2.6x10(-8)). The variants near TFAP2B appear to influence central adiposity through an effect on overall obesity/fat-mass, whereas LYPLAL1 displays a strong female-only association with fat distribution. By focusing on anthropometric measures of central obesity and fat distribution, we have identified three loci implicated in the regulation of human adiposity.


Subject(s)
Adiposity , Body Fat Distribution , Genome-Wide Association Study , Lysophospholipase/genetics , Obesity/genetics , Oxidoreductases/genetics , Transcription Factor AP-2/genetics , Adult , Cohort Studies , Female , Humans , Male , Methionine Sulfoxide Reductases , Obesity/metabolism , Polymorphism, Single Nucleotide , Waist Circumference , Waist-Hip Ratio
9.
Am J Epidemiol ; 172(6): 653-65, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20702506

ABSTRACT

The association between variation in the fat mass and obesity-associated (FTO) gene and adulthood body mass index (BMI; weight (kg)/height (m)(2)) is well-replicated. More thorough analyses utilizing phenotypic data over the life course may deepen our understanding of the development of BMI and thus help in the prevention of obesity. The authors used a structural equation modeling approach to explore the network of variables associated with BMI from the prenatal period to age 31 years (1965-1997) in 4,435 subjects from the Northern Finland Birth Cohort 1966. The use of structural equation modeling permitted the easy inclusion of variables with missing values in the analyses without separate imputation steps, as well as differentiation between direct and indirect effects. There was an association between the FTO single nucleotide polymorphism rs9939609 and BMI at age 31 years that persisted after controlling for several relevant factors during the life course. The total effect of the FTO variant on adult BMI was mostly composed of the direct effect, but a notable part was also arising indirectly via its effects on earlier BMI development. In addition to well-established genetic determinants, many life-course factors such as physical activity, in spite of not showing mediation or interaction, had a strong independent effect on BMI.


Subject(s)
Body Mass Index , Polymorphism, Single Nucleotide , Proteins/genetics , Adolescent , Adult , Alcohol Drinking , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Arctic Regions , Diet , Exercise , Female , Finland/epidemiology , Humans , Male , Models, Statistical , Phenotype , Prospective Studies , Smoking , Socioeconomic Factors
10.
Diabetes Care ; 43(4): 909-912, 2020 04.
Article in English | MEDLINE | ID: mdl-32001615

ABSTRACT

OBJECTIVE: Heterozygous loss-of-function mutations in HNF1A cause maturity-onset diabetes of the young (MODY). Affected individuals can be treated with low-dose sulfonylureas. Individuals with homozygous HNF1A mutations causing MODY have not been reported. RESEARCH DESIGN AND METHODS: We phenotyped a kindred with young-onset diabetes and performed molecular genetic testing, a mixed meal tolerance test, a sulfonylurea challenge, and in vitro assays to assess variant protein function. RESULTS: A homozygous HNF1A variant (p.A251T) was identified in three insulin-treated family members diagnosed with diabetes before 20 years of age. Those with the homozygous variant had low hs-CRP levels (0.2-0.8 mg/L), and those tested demonstrated sensitivity to sulfonylurea given at a low dose, completely transitioning off insulin. In silico modeling predicted a variant of unknown significance; however, in vitro studies supported a modest reduction in transactivation potential (79% of that for the wild type; P < 0.05) in the absence of endogenous HNF1A. CONCLUSIONS: Homozygous hypomorphic HNF1A variants are a cause of HNF1A-MODY. We thus expand the allelic spectrum of variants in dominant genes causing diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Sulfonylurea Compounds/therapeutic use , Adult , Age of Onset , Alleles , Diabetes Mellitus/drug therapy , Diabetes Mellitus/epidemiology , Diabetes Mellitus/genetics , Diabetes Mellitus, Type 2/epidemiology , Drug Resistance/drug effects , Drug Resistance/genetics , Female , Homozygote , Humans , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Mutation , Mutation, Missense , Pregnancy
11.
Diabetes Care ; 42(1): 17-26, 2019 01.
Article in English | MEDLINE | ID: mdl-30455330

ABSTRACT

OBJECTIVE: Maturity-onset diabetes of the young (MODY) due to variants in HNF1A is the most common type of monogenic diabetes. Frequent misdiagnosis results in missed opportunity to use sulfonylureas as first-line treatment. A nongenetic biomarker could improve selection of subjects for genetic testing and increase diagnosis rates. We previously reported that plasma levels of antennary fucosylated N-glycans and high-sensitivity C-reactive protein (hs-CRP) are reduced in individuals with HNF1A-MODY. In this study, we examined the potential use of N-glycans and hs-CRP in discriminating individuals with damaging HNF1A alleles from those without HNF1A variants in an unselected population of young adults with nonautoimmune diabetes. RESEARCH DESIGN AND METHODS: We analyzed the plasma N-glycan profile, measured hs-CRP, and sequenced HNF1A in 989 individuals with diabetes diagnosed when younger than age 45, persistent endogenous insulin production, and absence of pancreatic autoimmunity. Systematic assessment of rare HNF1A variants was performed. RESULTS: We identified 29 individuals harboring 25 rare HNF1A alleles, of which 3 were novel, and 12 (in 16 probands) were considered pathogenic. Antennary fucosylated N-glycans and hs-CRP were able to differentiate subjects with damaging HNF1A alleles from those without rare HNF1A alleles. Glycan GP30 had a receiver operating characteristic curve area under the curve (AUC) of 0.90 (88% sensitivity, 80% specificity, cutoff 0.70%), whereas hs-CRP had an AUC of 0.83 (88% sensitivity, 69% specificity, cutoff 0.81 mg/L). CONCLUSIONS: Half of rare HNF1A sequence variants do not cause MODY. N-glycan profile and hs-CRP could both be used as tools, alone or as adjuncts to existing pathways, for identifying individuals at high risk of carrying a damaging HNF1A allele.


Subject(s)
C-Reactive Protein/metabolism , Diabetes Mellitus, Type 2/blood , Hepatocyte Nuclear Factor 1-alpha/blood , Polysaccharides/blood , Adolescent , Adult , Alleles , Biomarkers/blood , Cholesterol/blood , Diabetes Mellitus, Type 2/drug therapy , Female , Glycated Hemoglobin/metabolism , Humans , Insulin/blood , Insulin/therapeutic use , Male , Middle Aged , Sensitivity and Specificity , Sequence Analysis, DNA , Triglycerides/blood , Young Adult
12.
Biochem Med (Zagreb) ; 28(2): 020703, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29666556

ABSTRACT

INTRODUCTION: Maturity onset diabetes of the young due to HNF1A mutations (HNF1A-MODY) is the most frequent form of monogenic diabetes in adults. It is often misdiagnosed as type 1 or type 2 diabetes, but establishing genetic diagnosis is important, as treatment differs from the common types of diabetes. HNF1A-MODY has not been investigated in Croatia before due to limited access to genetic testing. In this study we aimed to describe the characteristics of young adults diagnosed with diabetes before the age of 45 years, who have rare HNF1A allele variants, and estimate the prevalence of HNF1A-MODY in Croatia. MATERIALS AND METHODS: We recruited 477 C-peptide positive and beta cell antibody negative subjects through the Croatian Diabetes Registry. HNF1A was sequenced for all participants and systematic assessment of the variants found was performed. The prevalence of HNF1A-MODY was calculated in the study group and results extrapolated to estimate the proportion of diabetic individuals with HNF1A-MODY in Croatia and the population prevalence. RESULTS: Our study identified 13 individuals harbouring rare HNF1A allelic variants. After systematic assessment, 8 were assigned a diagnosis of HNF1A-MODY. Two individuals were able to discontinue insulin treatment following the diagnosis. We estimated that HNF1A-MODY in Croatia has a prevalence of 66 (95% CI 61 - 72) cases per million. CONCLUSIONS: The estimated prevalence of HNF1A-MODY in Croatia is similar to that reported in other European countries. Finding cases lead to important treatment changes for patients. This strongly supports the introduction of diagnostic genetic testing for monogenic diabetes in Croatia.


Subject(s)
C-Peptide/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Hepatocyte Nuclear Factor 1-alpha/genetics , Mutation , Registries , Adolescent , Adult , Aged , Alleles , Autoantibodies/blood , Biomarkers/blood , Croatia/epidemiology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Female , Gene Expression , Gene Frequency , Genetic Testing , Hepatocyte Nuclear Factor 1-alpha/immunology , Humans , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/pathology , Male , Middle Aged , Prevalence , Sequence Analysis, DNA
13.
Elife ; 72018 02 07.
Article in English | MEDLINE | ID: mdl-29412141

ABSTRACT

Human genetic studies have emphasised the dominant contribution of pancreatic islet dysfunction to development of Type 2 Diabetes (T2D). However, limited annotation of the islet epigenome has constrained efforts to define the molecular mechanisms mediating the, largely regulatory, signals revealed by Genome-Wide Association Studies (GWAS). We characterised patterns of chromatin accessibility (ATAC-seq, n = 17) and DNA methylation (whole-genome bisulphite sequencing, n = 10) in human islets, generating high-resolution chromatin state maps through integration with established ChIP-seq marks. We found enrichment of GWAS signals for T2D and fasting glucose was concentrated in subsets of islet enhancers characterised by open chromatin and hypomethylation, with the former annotation predominant. At several loci (including CDC123, ADCY5, KLHDC5) the combination of fine-mapping genetic data and chromatin state enrichment maps, supplemented by allelic imbalance in chromatin accessibility pinpointed likely causal variants. The combination of increasingly-precise genetic and islet epigenomic information accelerates definition of causal mechanisms implicated in T2D pathogenesis.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic , Genome-Wide Association Study , Islets of Langerhans/physiopathology , Chromatin/metabolism , DNA Methylation , Humans , White People
14.
Nat Genet ; 50(11): 1505-1513, 2018 11.
Article in English | MEDLINE | ID: mdl-30297969

ABSTRACT

We expanded GWAS discovery for type 2 diabetes (T2D) by combining data from 898,130 European-descent individuals (9% cases), after imputation to high-density reference panels. With these data, we (i) extend the inventory of T2D-risk variants (243 loci, 135 newly implicated in T2D predisposition, comprising 403 distinct association signals); (ii) enrich discovery of lower-frequency risk alleles (80 index variants with minor allele frequency <5%, 14 with estimated allelic odds ratio >2); (iii) substantially improve fine-mapping of causal variants (at 51 signals, one variant accounted for >80% posterior probability of association (PPA)); (iv) extend fine-mapping through integration of tissue-specific epigenomic information (islet regulatory annotations extend the number of variants with PPA >80% to 73); (v) highlight validated therapeutic targets (18 genes with associations attributable to coding variants); and (vi) demonstrate enhanced potential for clinical translation (genome-wide chip heritability explains 18% of T2D risk; individuals in the extremes of a T2D polygenic risk score differ more than ninefold in prevalence).


Subject(s)
Chromosome Mapping/methods , Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic , Genome, Human/genetics , Islets of Langerhans/metabolism , Polymorphism, Single Nucleotide , Body Mass Index , Case-Control Studies , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/pathology , Female , Gene Frequency , Genetic Loci/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , High-Throughput Screening Assays/methods , Humans , Islets of Langerhans/pathology , Linkage Disequilibrium , Male , Meta-Analysis as Topic , Sex Factors , White People/genetics
15.
J Clin Endocrinol Metab ; 92(8): 3219-23, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17535991

ABSTRACT

CONTEXT: Mitochondrial dysfunction is increasingly implicated in pathogenesis of adult metabolic disease. Rare mitochondrial (mt) DNA mutations impair glucose homeostasis, but the contribution of common variants is unclear. In small studies, variation within the OriB origin of replication (at mt16189 in particular) has been associated with both early growth and adult metabolic phenotypes and may contribute to life-course relationships between the two. OBJECTIVE: The aim was to study a large well-characterized cohort to determine whether previously reported small-scale associations between OriB sequence variation and early growth and adult metabolic phenotypes are robust. DESIGN/SETTING/PARTICIPANTS: This was a genetic association study of 5470 individuals from the population-based Northern Finland Birth Cohort of 1966, followed prospectively from pregnancy to age 31 yr. MAIN OUTCOME MEASURES: We measured indices of early growth (including birth weight, placental weight, and ponderal index) and adult metabolic homeostasis (including body mass index, fasting glucose and insulin, indices of insulin action and secretion) and their relationship to variation in the OriB region. RESULTS: Previously reported associations could not be confirmed. There were no significant (P < 0.01, uncorrected) associations between OriB sequence variation and measures of early growth including birth weight (P = 0.52, comparing individuals with mt16189T to those with a homopolymeric C-tract) and placental weight (P = 0.49). There were no significant associations with adult metabolic phenotypes including fasting glucose (P = 0.07), fasting insulin (P = 0.42), and homeostatic model assessment-derived measures of insulin sensitivity or secretion (P = 0.45 and P = 0.56, respectively). CONCLUSION: Despite substantial power to detect previously reported effects, mtDNA variations around OriB are not major contributors to variation in early growth and metabolic phenotypes during early adulthood.


Subject(s)
DNA, Mitochondrial/genetics , Growth/genetics , Metabolism/genetics , Mutation/physiology , Adult , Blood Glucose/metabolism , Blood Pressure/physiology , Body Mass Index , Cohort Studies , DNA Replication/genetics , DNA Replication/physiology , Female , Finland/epidemiology , Gene Frequency , Genetic Variation , Growth/physiology , Humans , Lipids/blood , Male , Metabolism/physiology , Phenotype , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Waist-Hip Ratio
16.
Eur J Hum Genet ; 15(6): 679-84, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17342155

ABSTRACT

Polycystic ovary syndrome (PCOS) is strongly associated with hyperinsulinaemia and type II diabetes (T2D). Sequence variation within KCNJ11 (encoding Kir6.2, the beta-cell inwardly rectifying potassium channel) is implicated in the pathogenesis of neonatal diabetes, hyperinsulinaemia of infancy and multifactorial T2D. Comprehensive tagging studies have demonstrated that the KCNJ11 E23K variant (or ABCC8 A1369S in LD>0.9) is responsible for the known association between KCNJ11 and T2D. Given the phenotypic overlap between PCOS and T2D, we investigated whether E23K is involved in susceptibility to PCOS and related traits. Case-control analyses for the KCNJ11 E23K variant were performed in (a) 374 PCOS cases and 2574 controls of UK British/Irish origin, and (b) 550 women with PCOS symptoms and 1114 controls from a Finnish birth cohort. The relationship between E23K genotype and androgen levels (a key intermediate phenotype relevant to PCOS) in 1380 samples was studied. The UK case-control analysis revealed no association between E23K genotypes and PCOS status (P=0.49; Cochran-Armitage test), and no significant relationship between E23K genotype and androgen measures in the samples for which these phenotypes were available (P=0.19). Similarly, the Finnish case-control analysis showed no association between E23K genotypes and PCOS status (P=0.75; Cochran-Armitage test), and no significant relationship between E23K genotype and androgen measures in the samples for which these phenotypes were available (Finnish controls, P=0.25; Finnish cases, P=0.08). In conclusion, these data (involving >4600 subjects) provide no evidence that common variants of the KCNJ11 E23K polymorphism have a major influence on PCOS susceptibility, though modest effect sizes (OR<1.25) cannot be excluded.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Polycystic Ovary Syndrome/genetics , Potassium Channels, Inwardly Rectifying/genetics , Sex Hormone-Binding Globulin/analysis , Testosterone/blood , Adult , Case-Control Studies , Female , Gene Frequency , Humans
17.
Stem Cell Reports ; 9(5): 1395-1405, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29107594

ABSTRACT

Current in vitro islet differentiation protocols suffer from heterogeneity and low efficiency. Induced pluripotent stem cells (iPSCs) derived from pancreatic beta cells (BiPSCs) preferentially differentiate toward endocrine pancreas-like cells versus those from fibroblasts (FiPSCs). We interrogated genome-wide open chromatin in BiPSCs and FiPSCs via ATAC-seq and identified ∼8.3k significant, differential open chromatin sites (DOCS) between the two iPSC subtypes (false discovery rate [FDR] < 0.05). DOCS where chromatin was more accessible in BiPSCs (Bi-DOCS) were significantly enriched for known regulators of endodermal development, including bivalent and weak enhancers, and FOXA2 binding sites (FDR < 0.05). Bi-DOCS were associated with genes related to pancreas development and beta-cell function, including transcription factors mutated in monogenic diabetes (PDX1, NKX2-2, HNF1A; FDR < 0.05). Moreover, Bi-DOCS correlated with enhanced gene expression in BiPSC-derived definitive endoderm and pancreatic progenitor cells. Bi-DOCS therefore highlight genes and pathways governing islet-lineage commitment, which can be exploited for differentiation protocol optimization, diabetes disease modeling, and therapeutic purposes.


Subject(s)
Cellular Reprogramming , Chromatin/genetics , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 3-beta/genetics , Induced Pluripotent Stem Cells/cytology , Insulin-Secreting Cells/cytology , Cells, Cultured , Chromatin/metabolism , Enhancer Elements, Genetic , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Insulin-Secreting Cells/metabolism , Nuclear Proteins , Protein Binding , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish Proteins
18.
Nat Genet ; 48(9): 1055-1059, 2016 09.
Article in English | MEDLINE | ID: mdl-27500523

ABSTRACT

Metformin is the first-line antidiabetic drug with over 100 million users worldwide, yet its mechanism of action remains unclear. Here the Metformin Genetics (MetGen) Consortium reports a three-stage genome-wide association study (GWAS), consisting of 13,123 participants of different ancestries. The C allele of rs8192675 in the intron of SLC2A2, which encodes the facilitated glucose transporter GLUT2, was associated with a 0.17% (P = 6.6 × 10(-14)) greater metformin-induced reduction in hemoglobin A1c (HbA1c) in 10,577 participants of European ancestry. rs8192675 was the top cis expression quantitative trait locus (cis-eQTL) for SLC2A2 in 1,226 human liver samples, suggesting a key role for hepatic GLUT2 in regulation of metformin action. Among obese individuals, C-allele homozygotes at rs8192675 had a 0.33% (3.6 mmol/mol) greater absolute HbA1c reduction than T-allele homozygotes. This was about half the effect seen with the addition of a DPP-4 inhibitor, and equated to a dose difference of 550 mg of metformin, suggesting rs8192675 as a potential biomarker for stratified medicine.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Glucose Transporter Type 2/genetics , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Polymorphism, Single Nucleotide/genetics , Quantitative Trait, Heritable , Blood Glucose/analysis , Body Mass Index , Diabetes Mellitus, Type 2/drug therapy , Genome-Wide Association Study , Glycated Hemoglobin/analysis , Humans , White People
19.
Diabetes ; 53(8): 2126-31, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277396

ABSTRACT

Variation at the insulin gene (INS-)VNTR (variable number of tandem repeats) minisatellite polymorphism has been reported to be associated with both early growth and adult metabolic phenotypes. However, the samples studied have been small and the relationship between INS-VNTR variation and parameters of early growth inconsistent, with four previous studies producing conflicting results. We have studied the relationship between INS-VNTR class (measured by genotyping the nearby -23HphI variant with which it is in tight linkage disequilibrium) and early growth in 5,646 members of the Northern Finnish Birth Cohort of 1966. Comparing class III homozygotes with other genotypes using multivariate linear regression analysis, we found no significant associations with any early growth measure (birth weight, birth length, ponderal index, and head circumference at 1 year), even after stratifying subjects by growth trajectory during infancy and/or birth order. For example, among infants with limited postnatal growth realignment (n = 2,470), class III/III infants were no heavier at birth (difference [+/-SE] in the means [fully adjusted], 58 +/- 51 g; P = 0.26) than class I/- infants. No significant associations were detected following reanalysis with an additive model (for example, for birth weight, beta = 20 g [95% CI -3 to 44], P = 0.09). Studies of this large population-based cohort have failed to generate convincing evidence that INS-VNTR variation influences early growth.


Subject(s)
Growth/genetics , Insulin/genetics , Minisatellite Repeats/genetics , Polymorphism, Genetic/genetics , Birth Order , Birth Weight , Body Constitution , Cohort Studies , Female , Finland , Genetic Variation , Gestational Age , Humans , Infant, Newborn , Parity , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL