Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Acta Neuropathol ; 147(1): 65, 2024 04 01.
Article in English | MEDLINE | ID: mdl-38557897

ABSTRACT

Human microglia are critically involved in Alzheimer's disease (AD) progression, as shown by genetic and molecular studies. However, their role in tau pathology progression in human brain has not been well described. Here, we characterized 32 human donors along progression of AD pathology, both in time-from early to late pathology-and in space-from entorhinal cortex (EC), inferior temporal gyrus (ITG), prefrontal cortex (PFC) to visual cortex (V2 and V1)-with biochemistry, immunohistochemistry, and single nuclei-RNA-sequencing, profiling a total of 337,512 brain myeloid cells, including microglia. While the majority of microglia are similar across brain regions, we identified a specific subset unique to EC which may contribute to the early tau pathology present in this region. We calculated conversion of microglia subtypes to diseased states and compared conversion patterns to those from AD animal models. Targeting genes implicated in this conversion, or their upstream/downstream pathways, could halt gene programs initiated by early tau progression. We used expression patterns of early tau progression to identify genes whose expression is reversed along spreading of spatial tau pathology (EC > ITG > PFC > V2 > V1) and identified their potential involvement in microglia subtype conversion to a diseased state. This study provides a data resource that builds on our knowledge of myeloid cell contribution to AD by defining the heterogeneity of microglia and brain macrophages during both temporal and regional pathology aspects of AD progression at an unprecedented resolution.


Subject(s)
Alzheimer Disease , Animals , Humans , Alzheimer Disease/pathology , tau Proteins/genetics , tau Proteins/metabolism , Transcriptome , Brain/pathology , Myeloid Cells/pathology , Microglia/pathology , Amyloid beta-Peptides/metabolism
2.
Nature ; 541(7635): 81-86, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28002404

ABSTRACT

Approximately 1.5 billion people worldwide are overweight or affected by obesity, and are at risk of developing type 2 diabetes, cardiovascular disease and related metabolic and inflammatory disturbances. Although the mechanisms linking adiposity to associated clinical conditions are poorly understood, recent studies suggest that adiposity may influence DNA methylation, a key regulator of gene expression and molecular phenotype. Here we use epigenome-wide association to show that body mass index (BMI; a key measure of adiposity) is associated with widespread changes in DNA methylation (187 genetic loci with P < 1 × 10-7, range P = 9.2 × 10-8 to 6.0 × 10-46; n = 10,261 samples). Genetic association analyses demonstrate that the alterations in DNA methylation are predominantly the consequence of adiposity, rather than the cause. We find that methylation loci are enriched for functional genomic features in multiple tissues (P < 0.05), and show that sentinel methylation markers identify gene expression signatures at 38 loci (P < 9.0 × 10-6, range P = 5.5 × 10-6 to 6.1 × 10-35, n = 1,785 samples). The methylation loci identify genes involved in lipid and lipoprotein metabolism, substrate transport and inflammatory pathways. Finally, we show that the disturbances in DNA methylation predict future development of type 2 diabetes (relative risk per 1 standard deviation increase in methylation risk score: 2.3 (2.07-2.56); P = 1.1 × 10-54). Our results provide new insights into the biologic pathways influenced by adiposity, and may enable development of new strategies for prediction and prevention of type 2 diabetes and other adverse clinical consequences of obesity.


Subject(s)
Adiposity/genetics , Body Mass Index , DNA Methylation/genetics , Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic , Epigenomics , Genome-Wide Association Study , Obesity/genetics , Adipose Tissue/metabolism , Asian People/genetics , Blood/metabolism , Cohort Studies , Diabetes Mellitus, Type 2/complications , Europe/ethnology , Female , Genetic Markers , Genetic Predisposition to Disease , Humans , India/ethnology , Male , Obesity/blood , Obesity/complications , Overweight/blood , Overweight/complications , Overweight/genetics , White People/genetics
3.
Nature ; 492(7429): 369-75, 2012 Dec 20.
Article in English | MEDLINE | ID: mdl-23222517

ABSTRACT

Anaemia is a chief determinant of global ill health, contributing to cognitive impairment, growth retardation and impaired physical capacity. To understand further the genetic factors influencing red blood cells, we carried out a genome-wide association study of haemoglobin concentration and related parameters in up to 135,367 individuals. Here we identify 75 independent genetic loci associated with one or more red blood cell phenotypes at P < 10(-8), which together explain 4-9% of the phenotypic variance per trait. Using expression quantitative trait loci and bioinformatic strategies, we identify 121 candidate genes enriched in functions relevant to red blood cell biology. The candidate genes are expressed preferentially in red blood cell precursors, and 43 have haematopoietic phenotypes in Mus musculus or Drosophila melanogaster. Through open-chromatin and coding-variant analyses we identify potential causal genetic variants at 41 loci. Our findings provide extensive new insights into genetic mechanisms and biological pathways controlling red blood cell formation and function.


Subject(s)
Erythrocytes/metabolism , Genetic Loci , Genome-Wide Association Study , Phenotype , Animals , Cell Cycle/genetics , Cytokines/metabolism , Drosophila melanogaster/genetics , Erythrocytes/cytology , Female , Gene Expression Regulation/genetics , Hematopoiesis/genetics , Hemoglobins/genetics , Humans , Male , Mice , Organ Specificity , Polymorphism, Single Nucleotide/genetics , RNA Interference , Signal Transduction/genetics
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.
J Lipid Res ; 58(9): 1834-1844, 2017 09.
Article in English | MEDLINE | ID: mdl-28512139

ABSTRACT

High lipoprotein (a) [Lp(a)] concentrations are an independent risk factor for cardiovascular outcomes. Concentrations are strongly influenced by apo(a) kringle IV repeat isoforms. We aimed to identify genetic loci associated with Lp(a) concentrations using data from five genome-wide association studies (n = 13,781). We identified 48 independent SNPs in the LPA and 1 SNP in the APOE gene region to be significantly associated with Lp(a) concentrations. We also adjusted for apo(a) isoforms to identify loci affecting Lp(a) levels independently from them, which resulted in 31 SNPs (30 in the LPA, 1 in the APOE gene region). Seven SNPs showed a genome-wide significant association with coronary artery disease (CAD) risk. A rare SNP (rs186696265; MAF ∼1%) showed the highest effect on Lp(a) and was also associated with increased risk of CAD (odds ratio = 1.73, P = 3.35 × 10-30). Median Lp(a) values increased from 2.1 to 91.1 mg/dl with increasing number of Lp(a)-increasing alleles. We found the APOE2-determining allele of rs7412 to be significantly associated with Lp(a) concentrations (P = 3.47 × 10-10). Each APOE2 allele decreased Lp(a) by 3.34 mg/dl corresponding to ∼15% of the population's mean values. Performing a gene-based test of association, including suspected Lp(a) receptors and regulators, resulted in one significant association of the TLR2 gene with Lp(a) (P = 3.4 × 10-4). In summary, we identified a large number of independent SNPs in the LPA gene region, as well as the APOE2 allele, to be significantly associated with Lp(a) concentrations.


Subject(s)
Apolipoproteins A/metabolism , Genome-Wide Association Study/methods , Lipoprotein(a)/genetics , Lipoprotein(a)/metabolism , Animals , Apolipoproteins A/genetics , Humans , Polymorphism, Single Nucleotide , Protein Isoforms/metabolism , Sex Characteristics
6.
Nature ; 477(7362): 54-60, 2011 Aug 31.
Article in English | MEDLINE | ID: mdl-21886157

ABSTRACT

Genome-wide association studies (GWAS) have identified many risk loci for complex diseases, but effect sizes are typically small and information on the underlying biological processes is often lacking. Associations with metabolic traits as functional intermediates can overcome these problems and potentially inform individualized therapy. Here we report a comprehensive analysis of genotype-dependent metabolic phenotypes using a GWAS with non-targeted metabolomics. We identified 37 genetic loci associated with blood metabolite concentrations, of which 25 show effect sizes that are unusually high for GWAS and account for 10-60% differences in metabolite levels per allele copy. Our associations provide new functional insights for many disease-related associations that have been reported in previous studies, including those for cardiovascular and kidney disorders, type 2 diabetes, cancer, gout, venous thromboembolism and Crohn's disease. The study advances our knowledge of the genetic basis of metabolic individuality in humans and generates many new hypotheses for biomedical and pharmaceutical research.


Subject(s)
Biomedical Research , Drug Industry , Genetic Variation , Genome-Wide Association Study , Metabolism/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Blood/metabolism , Child , Chronic Disease , Coronary Artery Disease/genetics , Diabetes Mellitus/genetics , Female , Genetic Loci/genetics , Genotype , Humans , Male , Metabolomics , Middle Aged , Pharmacogenetics , Renal Insufficiency/genetics , Risk Factors , Venous Thromboembolism/genetics , Young Adult
7.
Hum Mol Genet ; 23(21): 5847-57, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24927737

ABSTRACT

Availability of standardized metabolite panels and genome-wide single-nucleotide polymorphism data endorse the comprehensive analysis of gene-metabolite association. Currently, many studies use genome-wide association analysis to investigate the genetic effects on single metabolites (mGWAS) separately. Such studies have identified several loci that are associated not only with one but with multiple metabolites, facilitated by the fact that metabolite panels often include metabolites of the same or related pathways. Strategies that analyse several phenotypes in a combined way were shown to be able to detect additional genetic loci. One of those methods is the phenotype set enrichment analysis (PSEA) that tests sets of metabolites for enrichment at genes. Here we applied PSEA on two different panels of serum metabolites together with genome-wide data. All analyses were performed as a two-step identification-validation approach, using data from the population-based KORA cohort and the TwinsUK study. In addition to confirming genes that were already known from mGWAS, we were able to identify and validate 12 new genes. Knowledge about gene function was supported by the enriched metabolite sets. For loci with unknown gene functions, the results suggest a function that is interrelated with the metabolites, and hint at the underlying pathways.


Subject(s)
Biomarkers/blood , Genetic Association Studies , Metabolome , Phenotype , Cohort Studies , Computational Biology , Genetic Loci , Genotype , Humans , Metabolomics/methods , Polymorphism, Single Nucleotide , Reproducibility of Results
8.
PLoS Genet ; 9(8): e1003585, 2013.
Article in English | MEDLINE | ID: mdl-23990791

ABSTRACT

Several infrequent genetic polymorphisms in the SERPINA1 gene are known to substantially reduce concentration of alpha1-antitrypsin (AAT) in the blood. Since low AAT serum levels fail to protect pulmonary tissue from enzymatic degradation, these polymorphisms also increase the risk for early onset chronic obstructive pulmonary disease (COPD). The role of more common SERPINA1 single nucleotide polymorphisms (SNPs) in respiratory health remains poorly understood. We present here an agnostic investigation of genetic determinants of circulating AAT levels in a general population sample by performing a genome-wide association study (GWAS) in 1392 individuals of the SAPALDIA cohort. Five common SNPs, defined by showing minor allele frequencies (MAFs) >5%, reached genome-wide significance, all located in the SERPINA gene cluster at 14q32.13. The top-ranking genotyped SNP rs4905179 was associated with an estimated effect of ß = -0.068 g/L per minor allele (P = 1.20*10(-12)). But denser SERPINA1 locus genotyping in 5569 participants with subsequent stepwise conditional analysis, as well as exon-sequencing in a subsample (N = 410), suggested that AAT serum level is causally determined at this locus by rare (MAF<1%) and low-frequent (MAF 1-5%) variants only, in particular by the well-documented protein inhibitor S and Z (PI S, PI Z) variants. Replication of the association of rs4905179 with AAT serum levels in the Copenhagen City Heart Study (N = 8273) was successful (P<0.0001), as was the replication of its synthetic nature (the effect disappeared after adjusting for PI S and Z, P = 0.57). Extending the analysis to lung function revealed a more complex situation. Only in individuals with severely compromised pulmonary health (N = 397), associations of common SNPs at this locus with lung function were driven by rarer PI S or Z variants. Overall, our meta-analysis of lung function in ever-smokers does not support a functional role of common SNPs in the SERPINA gene cluster in the general population.


Subject(s)
Genome-Wide Association Study , Pulmonary Disease, Chronic Obstructive/genetics , alpha 1-Antitrypsin/blood , Denmark , Gene Frequency , Genetic Predisposition to Disease , Genotype , Humans , Lung/pathology , Multigene Family , Polymorphism, Single Nucleotide/genetics , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/pathology , alpha 1-Antitrypsin/genetics
9.
Genet Epidemiol ; 38(5): 381-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24962562

ABSTRACT

Genome-wide association studies are usually accompanied by imputation techniques to complement genome-wide SNP chip genotypes. Current imputation approaches separate the phasing of study data from imputing, which makes the phasing independent from the reference data. The two-step approach allows for updating the imputation for a new reference panel without repeating the tedious phasing step. This advantage, however, does no longer hold, when the build of the study data differs from the build of the reference data. In this case, the current approach is to harmonize the study data annotation with the reference data (prephasing lift-over), requiring rephasing and re-imputing. As a novel approach, we propose to harmonize study haplotypes with reference haplotypes (postphasing lift-over). This allows for updating imputed study data for new reference panels without requiring rephasing. With continuously updated reference panels, our approach can save considerable computing time of up to 1 month per re-imputation. We evaluated the rephasing and postphasing lift-over approaches by using data from 1,644 unrelated individuals imputed by both approaches and comparing it with directly typed genotypes. On average, both approaches perform equally well with mean concordances of 93% between imputed and typed genotypes for both approaches. Also, imputation qualities are similar (mean difference in RSQ < 0.1%). We demonstrate that our novel postphasing lift-over approach is a practical and time-saving alternative to the prephasing lift-over. This might encourage study partners to accommodate updated reference builds and ultimately improve the information content of study data. Our novel approach is implemented in the software PhaseLift.


Subject(s)
Genome-Wide Association Study/methods , Software , Haplotypes/genetics , Humans , Meta-Analysis as Topic , Polymorphism, Single Nucleotide/genetics , Time Factors
10.
Hum Genet ; 134(2): 131-46, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25367360

ABSTRACT

To identify genetic variants associated with refractive astigmatism in the general population, meta-analyses of genome-wide association studies were performed for: White Europeans aged at least 25 years (20 cohorts, N = 31,968); Asian subjects aged at least 25 years (7 cohorts, N = 9,295); White Europeans aged <25 years (4 cohorts, N = 5,640); and all independent individuals from the above three samples combined with a sample of Chinese subjects aged <25 years (N = 45,931). Participants were classified as cases with refractive astigmatism if the average cylinder power in their two eyes was at least 1.00 diopter and as controls otherwise. Genome-wide association analysis was carried out for each cohort separately using logistic regression. Meta-analysis was conducted using a fixed effects model. In the older European group the most strongly associated marker was downstream of the neurexin-1 (NRXN1) gene (rs1401327, P = 3.92E-8). No other region reached genome-wide significance, and association signals were lower for the younger European group and Asian group. In the meta-analysis of all cohorts, no marker reached genome-wide significance: The most strongly associated regions were, NRXN1 (rs1401327, P = 2.93E-07), TOX (rs7823467, P = 3.47E-07) and LINC00340 (rs12212674, P = 1.49E-06). For 34 markers identified in prior GWAS for spherical equivalent refractive error, the beta coefficients for genotype versus spherical equivalent, and genotype versus refractive astigmatism, were highly correlated (r = -0.59, P = 2.10E-04). This work revealed no consistent or strong genetic signals for refractive astigmatism; however, the TOX gene region previously identified in GWAS for spherical equivalent refractive error was the second most strongly associated region. Analysis of additional markers provided evidence supporting widespread genetic co-susceptibility for spherical and astigmatic refractive errors.


Subject(s)
Astigmatism/genetics , Cell Adhesion Molecules, Neuronal/genetics , Genome-Wide Association Study , High Mobility Group Proteins/genetics , Nerve Tissue Proteins/genetics , Adult , Age Factors , Asian People , Astigmatism/pathology , Calcium-Binding Proteins , Cohort Studies , Female , Genetic Markers , Humans , Male , Middle Aged , Neural Cell Adhesion Molecules , White People
11.
PLoS Genet ; 7(8): e1002215, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21852955

ABSTRACT

Metabolomic profiling and the integration of whole-genome genetic association data has proven to be a powerful tool to comprehensively explore gene regulatory networks and to investigate the effects of genetic variation at the molecular level. Serum metabolite concentrations allow a direct readout of biological processes, and association of specific metabolomic signatures with complex diseases such as Alzheimer's disease and cardiovascular and metabolic disorders has been shown. There are well-known correlations between sex and the incidence, prevalence, age of onset, symptoms, and severity of a disease, as well as the reaction to drugs. However, most of the studies published so far did not consider the role of sexual dimorphism and did not analyse their data stratified by gender. This study investigated sex-specific differences of serum metabolite concentrations and their underlying genetic determination. For discovery and replication we used more than 3,300 independent individuals from KORA F3 and F4 with metabolite measurements of 131 metabolites, including amino acids, phosphatidylcholines, sphingomyelins, acylcarnitines, and C6-sugars. A linear regression approach revealed significant concentration differences between males and females for 102 out of 131 metabolites (p-values<3.8×10(-4); Bonferroni-corrected threshold). Sex-specific genome-wide association studies (GWAS) showed genome-wide significant differences in beta-estimates for SNPs in the CPS1 locus (carbamoyl-phosphate synthase 1, significance level: p<3.8×10(-10); Bonferroni-corrected threshold) for glycine. We showed that the metabolite profiles of males and females are significantly different and, furthermore, that specific genetic variants in metabolism-related genes depict sexual dimorphism. Our study provides new important insights into sex-specific differences of cell regulatory processes and underscores that studies should consider sex-specific effects in design and interpretation.


Subject(s)
Metabolome/genetics , Polymorphism, Single Nucleotide , Sex Characteristics , Adult , Aged , Aged, 80 and over , Amino Acids/blood , Carnitine/analogs & derivatives , Carnitine/blood , Female , Genetic Markers , Genome-Wide Association Study , Glycine/blood , Humans , Lipids/blood , Male , Middle Aged
12.
PLoS Genet ; 7(10): e1002333, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22028671

ABSTRACT

Recent genome-wide association (GWA) studies described 95 loci controlling serum lipid levels. These common variants explain ∼25% of the heritability of the phenotypes. To date, no unbiased screen for gene-environment interactions for circulating lipids has been reported. We screened for variants that modify the relationship between known epidemiological risk factors and circulating lipid levels in a meta-analysis of genome-wide association (GWA) data from 18 population-based cohorts with European ancestry (maximum N = 32,225). We collected 8 further cohorts (N = 17,102) for replication, and rs6448771 on 4p15 demonstrated genome-wide significant interaction with waist-to-hip-ratio (WHR) on total cholesterol (TC) with a combined P-value of 4.79×10(-9). There were two potential candidate genes in the region, PCDH7 and CCKAR, with differential expression levels for rs6448771 genotypes in adipose tissue. The effect of WHR on TC was strongest for individuals carrying two copies of G allele, for whom a one standard deviation (sd) difference in WHR corresponds to 0.19 sd difference in TC concentration, while for A allele homozygous the difference was 0.12 sd. Our findings may open up possibilities for targeted intervention strategies for people characterized by specific genomic profiles. However, more refined measures of both body-fat distribution and metabolic measures are needed to understand how their joint dynamics are modified by the newly found locus.


Subject(s)
Cadherins/genetics , Cholesterol/genetics , Chromosomes, Human, Pair 4/genetics , Lipids/blood , Lipids/genetics , Waist-Hip Ratio , Adipose Tissue/metabolism , Body Fat Distribution , Cholesterol/blood , Chromosome Mapping , Genome-Wide Association Study , Genotype , Humans , Lipoproteins/blood , Lipoproteins/genetics , Phenotype , Polymorphism, Single Nucleotide , Protocadherins , Quantitative Trait Loci/genetics , Risk Factors , Triglycerides/blood , Triglycerides/genetics , White People/genetics
13.
SLAS Discov ; 29(2): 100137, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38128829

ABSTRACT

Aberrant protein aggregation is a pathological cellular hallmark of many neurodegenerative diseases, such as Alzheimer's disease (AD) and frontotemporal dementia (FTD), where the tau protein is aggregating, forming neurofibrillary tangles (NFTs), and propagating from neuron to neuron. These processes have been linked to disease progression and a decline in cognitive function. Various therapeutic approaches aim at the prevention or reduction of tau aggregates in neurons. Human induced pluripotent stem cells (hiPSCs) are a very valuable tool in neuroscience discovery, as they offer access to potentially unlimited amounts of cell types that are affected in disease, including cortical neurons, for in vitro studies. We have generated an in vitro model for tau aggregation that uses hiPSC - derived neurons expressing an aggregation prone, fluorescently tagged version of the human tau protein after lentiviral transduction. Upon addition of tau seeds in the form of recombinant sonicated paired helical filaments (sPHFs), the neurons show robust, disease-like aggregation of the tau protein. The model was developed as a plate-based high content screening assay coupled with an image analysis algorithm to evaluate the impact of small molecules or genetic perturbations on tau. We show that the assay can be used to evaluate small molecules or screen targeted compound libraries. Using siRNA-based gene knockdown, genes of interest can be evaluated, and we could show that a targeted gene library can be screened, by screening nearly 100 deubiquitinating enzymes (DUBs) in that assay. The assay uses an imaging-based readout, a relatively short timeline, quantifies the extent of tau aggregation, and also allows the assessment of cell viability. Furthermore, it can be easily adapted to different hiPSC lines or neuronal subtypes. Taken together, this complex and highly relevant approach can be routinely applied on a weekly basis in the screening funnels of several projects and generates data with a turnaround time of approximately five weeks.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Humans , tau Proteins/genetics , tau Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Alzheimer Disease/metabolism , Neurons/metabolism
14.
Genet Epidemiol ; 36(3): 244-52, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22714936

ABSTRACT

Most genome-wide association studies (GWAS) are restricted to one phenotype, even if multiple related or unrelated phenotypes are available. However, an integrated analysis of multiple phenotypes can provide insight into their shared genetic basis and may improve the power of association studies. We present a new method, called "phenotype set enrichment analysis" (PSEA), which uses ideas of gene set enrichment analysis for the investigation of phenotype sets. PSEA combines statistics of univariate phenotype analyses and tests by permutation. It does not only allow analyzing predefined phenotype sets, but also to identify new phenotype sets. Apart from the application to situations where phenotypes and genotypes are available for each person, the method was adjusted to the analysis of GWAS summary statistics. PSEA was applied to data from the population-based cohort KORA F4 (N = 1,814) using iron-related and blood count traits. By confirming associations previously found in large meta-analyses on these traits, PSEA was shown to be a reliable tool. Many of these associations were not detectable by GWAS on single phenotypes in KORA F4. Therefore, the results suggest that PSEA can be more powerful than a single phenotype GWAS for the identification of association with multiple phenotypes. PSEA is a valuable method for analysis of multiple phenotypes, which can help to understand phenotype networks. Its flexible design enables both the use of prior knowledge and the generation of new knowledge on connection of multiple phenotypes. A software program for PSEA based on GWAS results is available upon request.


Subject(s)
Genome-Wide Association Study/methods , Phenotype , Blood Proteins/genetics , Cohort Studies , Erythrocytes/physiology , Genome-Wide Association Study/statistics & numerical data , Genotype , Humans , Models, Genetic , Polymorphism, Single Nucleotide , Software
15.
PLoS Med ; 10(6): e1001474, 2013.
Article in English | MEDLINE | ID: mdl-23824655

ABSTRACT

BACKGROUND: The association between adiposity and cardiometabolic traits is well known from epidemiological studies. Whilst the causal relationship is clear for some of these traits, for others it is not. We aimed to determine whether adiposity is causally related to various cardiometabolic traits using the Mendelian randomization approach. METHODS AND FINDINGS: We used the adiposity-associated variant rs9939609 at the FTO locus as an instrumental variable (IV) for body mass index (BMI) in a Mendelian randomization design. Thirty-six population-based studies of individuals of European descent contributed to the analyses. Age- and sex-adjusted regression models were fitted to test for association between (i) rs9939609 and BMI (n  =  198,502), (ii) rs9939609 and 24 traits, and (iii) BMI and 24 traits. The causal effect of BMI on the outcome measures was quantified by IV estimators. The estimators were compared to the BMI-trait associations derived from the same individuals. In the IV analysis, we demonstrated novel evidence for a causal relationship between adiposity and incident heart failure (hazard ratio, 1.19 per BMI-unit increase; 95% CI, 1.03-1.39) and replicated earlier reports of a causal association with type 2 diabetes, metabolic syndrome, dyslipidemia, and hypertension (odds ratio for IV estimator, 1.1-1.4; all p < 0.05). For quantitative traits, our results provide novel evidence for a causal effect of adiposity on the liver enzymes alanine aminotransferase and gamma-glutamyl transferase and confirm previous reports of a causal effect of adiposity on systolic and diastolic blood pressure, fasting insulin, 2-h post-load glucose from the oral glucose tolerance test, C-reactive protein, triglycerides, and high-density lipoprotein cholesterol levels (all p < 0.05). The estimated causal effects were in agreement with traditional observational measures in all instances except for type 2 diabetes, where the causal estimate was larger than the observational estimate (p  =  0.001). CONCLUSIONS: We provide novel evidence for a causal relationship between adiposity and heart failure as well as between adiposity and increased liver enzymes.


Subject(s)
Adiposity/genetics , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Mendelian Randomization Analysis , Quantitative Trait, Heritable , Alpha-Ketoglutarate-Dependent Dioxygenase FTO , Body Mass Index , Case-Control Studies , Confounding Factors, Epidemiologic , Genetic Association Studies , Humans , Meta-Analysis as Topic , Polymorphism, Single Nucleotide/genetics , Proteins/genetics
16.
Hum Mol Genet ; 20(5): 1042-7, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21149283

ABSTRACT

The level of body iron storage and the erythropoietic need for iron are indicated by the serum levels of ferritin and soluble transferrin receptor (sTfR), respectively. A meta-analysis of five genome-wide association studies on sTfR and ferritin revealed novel association to the PCSK7 and TMPRSS6 loci for sTfR and the HFE locus for both parameters. The PCSK7 association was the most significant (rs236918, P = 1.1 × 10E-27) suggesting that proprotein convertase 7, the gene product of PCSK7, may be involved in sTfR generation and/or iron homeostasis. Conditioning the sTfR analyses on transferrin saturation abolished the HFE signal and substantially diminished the TMPRSS6 signal while the PCSK7 association was unaffected, suggesting that the former may be mediated by transferrin saturation whereas the PCSK7-associated effect on sTfR generation appears to be more direct.


Subject(s)
Receptors, Transferrin/genetics , Subtilisins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Genome-Wide Association Study , Humans , Iron/metabolism , Male , Middle Aged , Receptors, Transferrin/metabolism , Young Adult
17.
Epilepsia ; 54(2): 265-71, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23350840

ABSTRACT

PURPOSE: Structural variations disrupting the gene encoding the neuron-specific splicing regulator RBFOX1 have been reported in three patients exhibiting epilepsy in comorbidity with other neuropsychiatric disorders. Consistently, the Rbfox1 knockout mouse model showed an increased susceptibility of seizures. The present candidate gene study tested whether exon-disrupting deletions of RBFOX1 increase the risk of idiopathic generalized epilepsies (IGEs), representing the largest group of genetically determined epilepsies. METHODS: Screening of microdeletions (size: >40 kb, coverage >20 markers) affecting the genomic sequence of the RBFOX1 gene was carried out by high-resolution single-nucleotide polymorphism (SNP) arrays in 1,408 European patients with idiopathic generalized epilepsy (IGE) and 2,256 population controls. Validation of RBFOX1 deletions and familial segregation analysis were performed by quantitative polymerase chain reaction (qPCR). KEY FINDINGS: We detected five exon-disrupting RBFOX1 deletions in the IGE patients, whereas none was observed in the controls (p = 0.008, Fisher's exact test). The size of the exonic deletions ranged from 68 to 896 kb and affected the untranslated 5'-terminal RBFOX1 exons. Segregation analysis in four families indicated that the deletions were inherited, display incomplete penetrance, and heterogeneous cosegregation patterns with IGE. SIGNIFICANCE: Rare deletions affecting the untranslated 5'-terminal RBFOX1 exons increase risk of common IGE syndromes. Variable expressivity, incomplete penetrance, and heterogeneous cosegregation patterns suggest that RBFOX1 deletions act as susceptibility factor in a genetically complex etiology, where heterogeneous combinations of genetic factors determine the disease phenotype.


Subject(s)
Epilepsy, Generalized/genetics , Exons/genetics , Gene Deletion , RNA-Binding Proteins/genetics , 5' Untranslated Regions , Age of Onset , Case-Control Studies , Child , Comorbidity , DNA/genetics , Epilepsy, Generalized/epidemiology , Female , Humans , Male , Pedigree , Polymerase Chain Reaction , Polymorphism, Single Nucleotide/genetics , RNA Splicing Factors , White People
18.
PLoS Genet ; 6(11): e1001213, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-21124955

ABSTRACT

Elevated levels of acute-phase serum amyloid A (A-SAA) cause amyloidosis and are a risk factor for atherosclerosis and its clinical complications, type 2 diabetes, as well as various malignancies. To investigate the genetic basis of A-SAA levels, we conducted the first genome-wide association study on baseline A-SAA concentrations in three population-based studies (KORA, TwinsUK, Sorbs) and one prospective case cohort study (LURIC), including a total of 4,212 participants of European descent, and identified two novel genetic susceptibility regions at 11p15.5-p13 and 1p31. The region at 11p15.5-p13 (rs4150642; p = 3.20×10(-111)) contains serum amyloid A1 (SAA1) and the adjacent general transcription factor 2 H1 (GTF2H1), Hermansky-Pudlak Syndrome 5 (HPS5), lactate dehydrogenase A (LDHA), and lactate dehydrogenase C (LDHC). This region explains 10.84% of the total variation of A-SAA levels in our data, which makes up 18.37% of the total estimated heritability. The second region encloses the leptin receptor (LEPR) gene at 1p31 (rs12753193; p = 1.22×10(-11)) and has been found to be associated with CRP and fibrinogen in previous studies. Our findings demonstrate a key role of the 11p15.5-p13 region in the regulation of baseline A-SAA levels and provide confirmative evidence of the importance of the 1p31 region for inflammatory processes and the close interplay between A-SAA, leptin, and other acute-phase proteins.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 1/genetics , Genome-Wide Association Study , Serum Amyloid A Protein/genetics , Female , Genetic Predisposition to Disease , Humans , Male , Meta-Analysis as Topic
19.
PLoS Genet ; 6(8)2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20700443

ABSTRACT

Magnesium, potassium, and sodium, cations commonly measured in serum, are involved in many physiological processes including energy metabolism, nerve and muscle function, signal transduction, and fluid and blood pressure regulation. To evaluate the contribution of common genetic variation to normal physiologic variation in serum concentrations of these cations, we conducted genome-wide association studies of serum magnesium, potassium, and sodium concentrations using approximately 2.5 million genotyped and imputed common single nucleotide polymorphisms (SNPs) in 15,366 participants of European descent from the international CHARGE Consortium. Study-specific results were combined using fixed-effects inverse-variance weighted meta-analysis. SNPs demonstrating genome-wide significant (p<5 x 10(-8)) or suggestive associations (p<4 x 10(-7)) were evaluated for replication in an additional 8,463 subjects of European descent. The association of common variants at six genomic regions (in or near MUC1, ATP2B1, DCDC5, TRPM6, SHROOM3, and MDS1) with serum magnesium levels was genome-wide significant when meta-analyzed with the replication dataset. All initially significant SNPs from the CHARGE Consortium showed nominal association with clinically defined hypomagnesemia, two showed association with kidney function, two with bone mineral density, and one of these also associated with fasting glucose levels. Common variants in CNNM2, a magnesium transporter studied only in model systems to date, as well as in CNNM3 and CNNM4, were also associated with magnesium concentrations in this study. We observed no associations with serum sodium or potassium levels exceeding p<4 x 10(-7). Follow-up studies of newly implicated genomic loci may provide additional insights into the regulation and homeostasis of human serum magnesium levels.


Subject(s)
Genome-Wide Association Study , Magnesium/blood , Potassium/blood , Sodium/blood , White People/genetics , Adult , Aged , Female , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
20.
J Am Soc Nephrol ; 21(7): 1223-32, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20558539

ABSTRACT

Phosphorus is an essential mineral that maintains cellular energy and mineralizes the skeleton. Because complex actions of ion transporters and regulatory hormones regulate serum phosphorus concentrations, genetic variation may determine interindividual variation in phosphorus metabolism. Here, we report a comprehensive genome-wide association study of serum phosphorus concentration. We evaluated 16,264 participants of European ancestry from the Cardiovascular Heath Study, Atherosclerosis Risk in Communities Study, Framingham Offspring Study, and the Rotterdam Study. We excluded participants with an estimated GFR <45 ml/min per 1.73 m(2) to focus on phosphorus metabolism under normal conditions. We imputed genotypes to approximately 2.5 million single-nucleotide polymorphisms in the HapMap and combined study-specific findings using meta-analysis. We tested top polymorphisms from discovery cohorts in a 5444-person replication sample. Polymorphisms in seven loci with minor allele frequencies 0.08 to 0.49 associate with serum phosphorus concentration (P = 3.5 x 10(-16) to 3.6 x 10(-7)). Three loci were near genes encoding the kidney-specific type IIa sodium phosphate co-transporter (SLC34A1), the calcium-sensing receptor (CASR), and fibroblast growth factor 23 (FGF23), proteins that contribute to phosphorus metabolism. We also identified genes encoding phosphatases, kinases, and phosphodiesterases that have yet-undetermined roles in phosphorus homeostasis. In the replication sample, five of seven top polymorphisms associate with serum phosphorous concentrations (P < 0.05 for each). In conclusion, common genetic variants associate with serum phosphorus in the general population. Further study of the loci identified in this study may help elucidate mechanisms of phosphorus regulation.


Subject(s)
Genetic Loci/genetics , Genetic Variation/genetics , Genome-Wide Association Study , Phosphorus/blood , Polymorphism, Single Nucleotide/genetics , Adult , Aged , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Gene Frequency/genetics , Humans , Kidney/physiology , Male , Middle Aged , Receptors, Calcium-Sensing/genetics , Sex Factors , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , White People
SELECTION OF CITATIONS
SEARCH DETAIL