Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 51
Filtrer
1.
Nat Genet ; 56(2): 273-280, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38233595

RÉSUMÉ

Myeloproliferative neoplasms (MPNs) are chronic cancers characterized by overproduction of mature blood cells. Their causative somatic mutations, for example, JAK2V617F, are common in the population, yet only a minority of carriers develop MPN. Here we show that the inherited polygenic loci that underlie common hematological traits influence JAK2V617F clonal expansion. We identify polygenic risk scores (PGSs) for monocyte count and plateletcrit as new risk factors for JAK2V617F positivity. PGSs for several hematological traits influenced the risk of different MPN subtypes, with low PGSs for two platelet traits also showing protective effects in JAK2V617F carriers, making them two to three times less likely to have essential thrombocythemia than carriers with high PGSs. We observed that extreme hematological PGSs may contribute to an MPN diagnosis in the absence of somatic driver mutations. Our study showcases how polygenic backgrounds underlying common hematological traits influence both clonal selection on somatic mutations and the subsequent phenotype of cancer.


Sujet(s)
Syndromes myéloprolifératifs , Tumeurs , Humains , Mutation , Syndromes myéloprolifératifs/génétique , Syndromes myéloprolifératifs/diagnostic , Phénotype , Kinase Janus-2/génétique ,
2.
Nat Commun ; 14(1): 5023, 2023 08 18.
Article de Anglais | MEDLINE | ID: mdl-37596262

RÉSUMÉ

Blood cells contain functionally important intracellular structures, such as granules, critical to immunity and thrombosis. Quantitative variation in these structures has not been subjected previously to large-scale genetic analysis. We perform genome-wide association studies of 63 flow-cytometry derived cellular phenotypes-including cell-type specific measures of granularity, nucleic acid content and reactivity-in 41,515 participants in the INTERVAL study. We identify 2172 distinct variant-trait associations, including associations near genes coding for proteins in organelles implicated in inflammatory and thrombotic diseases. By integrating with epigenetic data we show that many intracellular structures are likely to be determined in immature precursor cells. By integrating with proteomic data we identify the transcription factor FOG2 as an early regulator of platelet formation and α-granularity. Finally, we show that colocalisation of our associations with disease risk signals can suggest aetiological cell-types-variants in IL2RA and ITGA4 respectively mirror the known effects of daclizumab in multiple sclerosis and vedolizumab in inflammatory bowel disease.


Sujet(s)
Étude d'association pangénomique , Protéomique , Microscopie , Facteurs de transcription , Causalité
3.
Hum Mol Genet ; 32(5): 790-797, 2023 02 19.
Article de Anglais | MEDLINE | ID: mdl-36136759

RÉSUMÉ

Few genome-wide association studies (GWAS) analyzing genetic regulation of morphological traits of white blood cells have been reported. We carried out a GWAS of 12 morphological traits in 869 individuals from the general population of Sardinia, Italy. These traits, included measures of cell volume, conductivity and light scatter in four white-cell populations (eosinophils, lymphocytes, monocytes, neutrophils). This analysis yielded seven statistically significant signals, four of which were novel (four novel, PRG2, P2RX3, two of CDK6). Five signals were replicated in the independent INTERVAL cohort of 11 822 individuals. The most interesting signal with large effect size on eosinophil scatter (P-value = 8.33 x 10-32, beta = -1.651, se = 0.1351) falls within the innate immunity cluster on chromosome 11, and is located in the PRG2 gene. Computational analyses revealed that a rare, Sardinian-specific PRG2:p.Ser148Pro mutation modifies PRG2 amino acid contacts and protein dynamics in a manner that could possibly explain the changes observed in eosinophil morphology. Our discoveries shed light on genetics of morphological traits. For the first time, we describe such large effect size on eosinophils morphology that is relatively frequent in Sardinian population.


Sujet(s)
Granulocytes éosinophiles , Étude d'association pangénomique , Humains , Chromosomes humains de la paire 11 , Polymorphisme de nucléotide simple , Immunité innée
4.
PLoS Genet ; 18(11): e1010367, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-36327219

RÉSUMÉ

Host genetics is a key determinant of COVID-19 outcomes. Previously, the COVID-19 Host Genetics Initiative genome-wide association study used common variants to identify multiple loci associated with COVID-19 outcomes. However, variants with the largest impact on COVID-19 outcomes are expected to be rare in the population. Hence, studying rare variants may provide additional insights into disease susceptibility and pathogenesis, thereby informing therapeutics development. Here, we combined whole-exome and whole-genome sequencing from 21 cohorts across 12 countries and performed rare variant exome-wide burden analyses for COVID-19 outcomes. In an analysis of 5,085 severe disease cases and 571,737 controls, we observed that carrying a rare deleterious variant in the SARS-CoV-2 sensor toll-like receptor TLR7 (on chromosome X) was associated with a 5.3-fold increase in severe disease (95% CI: 2.75-10.05, p = 5.41x10-7). This association was consistent across sexes. These results further support TLR7 as a genetic determinant of severe disease and suggest that larger studies on rare variants influencing COVID-19 outcomes could provide additional insights.


Sujet(s)
COVID-19 , Exome , Humains , Exome/génétique , Étude d'association pangénomique , COVID-19/génétique , Prédisposition génétique à une maladie , Récepteur de type Toll-7/génétique , SARS-CoV-2/génétique
5.
Am J Hum Genet ; 109(6): 1038-1054, 2022 06 02.
Article de Anglais | MEDLINE | ID: mdl-35568032

RÉSUMÉ

Metabolite levels measured in the human population are endophenotypes for biological processes. We combined sequencing data for 3,924 (whole-exome sequencing, WES, discovery) and 2,805 (whole-genome sequencing, WGS, replication) donors from a prospective cohort of blood donors in England. We used multiple approaches to select and aggregate rare genetic variants (minor allele frequency [MAF] < 0.1%) in protein-coding regions and tested their associations with 995 metabolites measured in plasma by using ultra-high-performance liquid chromatography-tandem mass spectrometry. We identified 40 novel associations implicating rare coding variants (27 genes and 38 metabolites), of which 28 (15 genes and 28 metabolites) were replicated. We developed algorithms to prioritize putative driver variants at each locus and used mediation and Mendelian randomization analyses to test directionality at associations of metabolite and protein levels at the ACY1 locus. Overall, 66% of reported associations implicate gene targets of approved drugs or bioactive drug-like compounds, contributing to drug targets' validating efforts.


Sujet(s)
Exome , Exome/génétique , Fréquence d'allèle/génétique , Humains , Études prospectives , /méthodes , Séquençage du génome entier
6.
Nat Genet ; 54(3): 251-262, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35288711

RÉSUMÉ

The resolution of causal genetic variants informs understanding of disease biology. We used regulatory quantitative trait loci (QTLs) from the BLUEPRINT, GTEx and eQTLGen projects to fine-map putative causal variants for 12 immune-mediated diseases. We identify 340 unique loci that colocalize with high posterior probability (≥98%) with regulatory QTLs and apply Bayesian frameworks to fine-map associations at each locus. We show that fine-mapping credible sets derived from regulatory QTLs are smaller compared to disease summary statistics. Further, they are enriched for more functionally interpretable candidate causal variants and for putatively causal insertion/deletion (INDEL) polymorphisms. Finally, we use massively parallel reporter assays to evaluate candidate causal variants at the ITGA4 locus associated with inflammatory bowel disease. Overall, our findings suggest that fine-mapping applied to disease-colocalizing regulatory QTLs can enhance the discovery of putative causal disease variants and enhance insights into the underlying causal genes and molecular mechanisms.


Sujet(s)
Étude d'association pangénomique , Locus de caractère quantitatif , Théorème de Bayes , Causalité , Phénotype , Polymorphisme de nucléotide simple/génétique , Locus de caractère quantitatif/génétique
7.
Am J Hum Genet ; 108(10): 1836-1851, 2021 10 07.
Article de Anglais | MEDLINE | ID: mdl-34582791

RÉSUMÉ

Many common and rare variants associated with hematologic traits have been discovered through imputation on large-scale reference panels. However, the majority of genome-wide association studies (GWASs) have been conducted in Europeans, and determining causal variants has proved challenging. We performed a GWAS of total leukocyte, neutrophil, lymphocyte, monocyte, eosinophil, and basophil counts generated from 109,563,748 variants in the autosomes and the X chromosome in the Trans-Omics for Precision Medicine (TOPMed) program, which included data from 61,802 individuals of diverse ancestry. We discovered and replicated 7 leukocyte trait associations, including (1) the association between a chromosome X, pseudo-autosomal region (PAR), noncoding variant located between cytokine receptor genes (CSF2RA and CLRF2) and lower eosinophil count; and (2) associations between single variants found predominantly among African Americans at the S1PR3 (9q22.1) and HBB (11p15.4) loci and monocyte and lymphocyte counts, respectively. We further provide evidence indicating that the newly discovered eosinophil-lowering chromosome X PAR variant might be associated with reduced susceptibility to common allergic diseases such as atopic dermatitis and asthma. Additionally, we found a burden of very rare FLT3 (13q12.2) variants associated with monocyte counts. Together, these results emphasize the utility of whole-genome sequencing in diverse samples in identifying associations missed by European-ancestry-driven GWASs.


Sujet(s)
Asthme/épidémiologie , Marqueurs biologiques/métabolisme , Eczéma atopique/épidémiologie , Leucocytes/anatomopathologie , Polymorphisme de nucléotide simple , Broncho-pneumopathie chronique obstructive/épidémiologie , Locus de caractère quantitatif , Asthme/génétique , Asthme/métabolisme , Asthme/anatomopathologie , Eczéma atopique/génétique , Eczéma atopique/métabolisme , Eczéma atopique/anatomopathologie , Prédisposition génétique à une maladie , Génome humain , Étude d'association pangénomique , Humains , National Heart, Lung, and Blood Institute (USA) , Phénotype , Pronostic , Protéome/analyse , Protéome/métabolisme , Broncho-pneumopathie chronique obstructive/génétique , Broncho-pneumopathie chronique obstructive/métabolisme , Broncho-pneumopathie chronique obstructive/anatomopathologie , Royaume-Uni/épidémiologie , États-Unis/épidémiologie , Séquençage du génome entier
8.
Nat Med ; 27(9): 1564-1575, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34426706

RÉSUMÉ

Mitochondrial DNA (mtDNA) variants influence the risk of late-onset human diseases, but the reasons for this are poorly understood. Undertaking a hypothesis-free analysis of 5,689 blood-derived biomarkers with mtDNA variants in 16,220 healthy donors, here we show that variants defining mtDNA haplogroups Uk and H4 modulate the level of circulating N-formylmethionine (fMet), which initiates mitochondrial protein translation. In human cytoplasmic hybrid (cybrid) lines, fMet modulated both mitochondrial and cytosolic proteins on multiple levels, through transcription, post-translational modification and proteolysis by an N-degron pathway, abolishing known differences between mtDNA haplogroups. In a further 11,966 individuals, fMet levels contributed to all-cause mortality and the disease risk of several common cardiovascular disorders. Together, these findings indicate that fMet plays a key role in common age-related disease through pleiotropic effects on cell proteostasis.


Sujet(s)
Marqueurs biologiques/sang , Maladies cardiovasculaires/génétique , ADN mitochondrial/génétique , Mitochondries/génétique , Âge de début , Donneurs de sang , Maladies cardiovasculaires/sang , Maladies cardiovasculaires/épidémiologie , ADN mitochondrial/sang , Femelle , Études de suivi , Haplotypes/génétique , Humains , Mâle , Adulte d'âge moyen , Mitochondries/anatomopathologie , N-Formyl-méthionine/métabolisme , Homéostasie protéique , Facteurs de risque , Royaume-Uni/épidémiologie
9.
Int J Obes (Lond) ; 45(10): 2221-2229, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34226637

RÉSUMÉ

BACKGROUND: Variation in adiposity is associated with cardiometabolic disease outcomes, but mechanisms leading from this exposure to disease are unclear. This study aimed to estimate effects of body mass index (BMI) on an extensive set of circulating proteins. METHODS: We used SomaLogic proteomic data from up to 2737 healthy participants from the INTERVAL study. Associations between self-reported BMI and 3622 unique plasma proteins were explored using linear regression. These were complemented by Mendelian randomisation (MR) analyses using a genetic risk score (GRS) comprised of 654 BMI-associated polymorphisms from a recent genome-wide association study (GWAS) of adult BMI. A disease enrichment analysis was performed using DAVID Bioinformatics 6.8 for proteins which were altered by BMI. RESULTS: Observationally, BMI was associated with 1576 proteins (P < 1.4 × 10-5), with particularly strong evidence for a positive association with leptin and fatty acid-binding protein-4 (FABP4), and a negative association with sex hormone-binding globulin (SHBG). Observational estimates were likely confounded, but the GRS for BMI did not associate with measured confounders. MR analyses provided evidence for a causal relationship between BMI and eight proteins including leptin (0.63 standard deviation (SD) per SD BMI, 95% CI 0.48-0.79, P = 1.6 × 10-15), FABP4 (0.64 SD per SD BMI, 95% CI 0.46-0.83, P = 6.7 × 10-12) and SHBG (-0.45 SD per SD BMI, 95% CI -0.65 to -0.25, P = 1.4 × 10-5). There was agreement in the magnitude of observational and MR estimates (R2 = 0.33) and evidence that proteins most strongly altered by BMI were enriched for genes involved in cardiovascular disease. CONCLUSIONS: This study provides evidence for a broad impact of adiposity on the human proteome. Proteins strongly altered by BMI include those involved in regulating appetite, sex hormones and inflammation; such proteins are also enriched for cardiovascular disease-related genes. Altogether, results help focus attention onto new proteomic signatures of obesity-related disease.


Sujet(s)
Adiposité/physiologie , Protéome/analyse , Adulte , Indice de masse corporelle , Études de cohortes , Femelle , Humains , Mâle , Analyse de randomisation mendélienne , Adulte d'âge moyen , Études prospectives , Protéome/métabolisme , Enquêtes et questionnaires
10.
Nat Commun ; 12(1): 2298, 2021 04 16.
Article de Anglais | MEDLINE | ID: mdl-33863903

RÉSUMÉ

Neutrophils play fundamental roles in innate immune response, shape adaptive immunity, and are a potentially causal cell type underpinning genetic associations with immune system traits and diseases. Here, we profile the binding of myeloid master regulator PU.1 in primary neutrophils across nearly a hundred volunteers. We show that variants associated with differential PU.1 binding underlie genetically-driven differences in cell count and susceptibility to autoimmune and inflammatory diseases. We integrate these results with other multi-individual genomic readouts, revealing coordinated effects of PU.1 binding variants on the local chromatin state, enhancer-promoter contacts and downstream gene expression, and providing a functional interpretation for 27 genes underlying immune traits. Collectively, these results demonstrate the functional role of PU.1 and its target enhancers in neutrophil transcriptional control and immune disease susceptibility.


Sujet(s)
Maladies auto-immunes/génétique , Éléments activateurs (génétique)/génétique , Régulation de l'expression des gènes/immunologie , Granulocytes neutrophiles/immunologie , Protéines proto-oncogènes/métabolisme , Transactivateurs/métabolisme , Adulte , Sujet âgé , Maladies auto-immunes/immunologie , Chromatine/métabolisme , Séquençage après immunoprécipitation de la chromatine , Femelle , Humains , Mâle , Adulte d'âge moyen , Granulocytes neutrophiles/métabolisme , Régions promotrices (génétique)/génétique , Locus de caractère quantitatif/génétique , Locus de caractère quantitatif/immunologie , Jeune adulte
11.
PLoS Genet ; 16(3): e1008605, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-32150548

RÉSUMÉ

Circulating metabolite levels are biomarkers for cardiovascular disease (CVD). Here we studied, association of rare variants and 226 serum lipoproteins, lipids and amino acids in 7,142 (discovery plus follow-up) healthy participants. We leveraged the information from multiple metabolite measurements on the same participants to improve discovery in rare variant association analyses for gene-based and gene-set tests by incorporating correlated metabolites as covariates in the validation stage. Gene-based analysis corrected for the effective number of tests performed, confirmed established associations at APOB, APOC3, PAH, HAL and PCSK (p<1.32x10-7) and identified novel gene-trait associations at a lower stringency threshold with ACSL1, MYCN, FBXO36 and B4GALNT3 (p<2.5x10-6). Regulation of the pyruvate dehydrogenase (PDH) complex was associated for the first time, in gene-set analyses also corrected for effective number of tests, with IDL and LDL parameters, as well as circulating cholesterol (pMETASKAT<2.41x10-6). In conclusion, using an approach that leverages metabolite measurements obtained in the same participants, we identified novel loci and pathways involved in the regulation of these important metabolic biomarkers. As large-scale biobanks continue to amass sequencing and phenotypic information, analytical approaches such as ours will be useful to fully exploit the copious amounts of biological data generated in these efforts.


Sujet(s)
Marqueurs biologiques/sang , Maladies cardiovasculaires/sang , Maladies cardiovasculaires/génétique , Variation génétique/génétique , Cholestérol/sang , Cholestérol LDL/sang , Femelle , Étude d'association pangénomique/méthodes , Humains , Lipoprotéines/sang , Mâle , Phénotype , Triglycéride/sang
12.
Genet Epidemiol ; 44(1): 79-89, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31520489

RÉSUMÉ

Copy number variants (CNVs) play an important role in a number of human diseases, but the accurate calling of CNVs remains challenging. Most current approaches to CNV detection use raw read alignments, which are computationally intensive to process. We use a regression tree-based approach to call germline CNVs from whole-genome sequencing (WGS, >18x) variant call sets in 6,898 samples across four European cohorts, and describe a rich large variation landscape comprising 1,320 CNVs. Eighty-one percent of detected events have been previously reported in the Database of Genomic Variants. Twenty-three percent of high-quality deletions affect entire genes, and we recapitulate known events such as the GSTM1 and RHD gene deletions. We test for association between the detected deletions and 275 protein levels in 1,457 individuals to assess the potential clinical impact of the detected CNVs. We describe complex CNV patterns underlying an association with levels of the CCL3 protein (MAF = 0.15, p = 3.6x10-12 ) at the CCL3L3 locus, and a novel cis-association between a low-frequency NOMO1 deletion and NOMO1 protein levels (MAF = 0.02, p = 2.2x10-7 ). This study demonstrates that existing population-wide WGS call sets can be mined for germline CNVs with minimal computational overhead, delivering insight into a less well-studied, yet potentially impactful class of genetic variant.


Sujet(s)
Variations de nombre de copies de segment d'ADN/génétique , Génétique des populations/méthodes , Génome humain/génétique , Chimiokine CCL3/génétique , Délétion de gène , Étude d'association pangénomique , Génomique , Glutathione transferase/génétique , Humains , Protéine Nodal/génétique , Protéines de fusion recombinantes/génétique , Séquençage du génome entier
13.
Nat Genet ; 51(2): 343-353, 2019 02.
Article de Anglais | MEDLINE | ID: mdl-30692680

RÉSUMÉ

Loci discovered by genome-wide association studies predominantly map outside protein-coding genes. The interpretation of the functional consequences of non-coding variants can be greatly enhanced by catalogs of regulatory genomic regions in cell lines and primary tissues. However, robust and readily applicable methods are still lacking by which to systematically evaluate the contribution of these regions to genetic variation implicated in diseases or quantitative traits. Here we propose a novel approach that leverages genome-wide association studies' findings with regulatory or functional annotations to classify features relevant to a phenotype of interest. Within our framework, we account for major sources of confounding not offered by current methods. We further assess enrichment of genome-wide association studies for 19 traits within Encyclopedia of DNA Elements- and Roadmap-derived regulatory regions. We characterize unique enrichment patterns for traits and annotations driving novel biological insights. The method is implemented in standalone software and an R package, to facilitate its application by the research community.


Sujet(s)
Maladie/génétique , Génome/génétique , Étude d'association pangénomique/méthodes , Génomique/méthodes , Humains , Annotation de séquence moléculaire/méthodes , Phénotype , Polymorphisme de nucléotide simple/génétique , Locus de caractère quantitatif/génétique , Séquences d'acides nucléiques régulatrices/génétique , Logiciel
14.
Nat Commun ; 10(1): 357, 2019 01 21.
Article de Anglais | MEDLINE | ID: mdl-30664637

RÉSUMÉ

Cranial growth and development is a complex process which affects the closely related traits of head circumference (HC) and intracranial volume (ICV). The underlying genetic influences shaping these traits during the transition from childhood to adulthood are little understood, but might include both age-specific genetic factors and low-frequency genetic variation. Here, we model the developmental genetic architecture of HC, showing this is genetically stable and correlated with genetic determinants of ICV. Investigating up to 46,000 children and adults of European descent, we identify association with final HC and/or final ICV + HC at 9 novel common and low-frequency loci, illustrating that genetic variation from a wide allele frequency spectrum contributes to cranial growth. The largest effects are reported for low-frequency variants within TP53, with 0.5 cm wider heads in increaser-allele carriers versus non-carriers during mid-childhood, suggesting a previously unrecognized role of TP53 transcripts in human cranial development.


Sujet(s)
Allèles , Locus génétiques , Variation génétique , ARN messager/génétique , Crâne/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Céphalométrie , Enfant , Femelle , Régulation de l'expression des gènes au cours du développement , Fréquence d'allèle , Génome humain , Humains , Mâle , Adulte d'âge moyen , Crâne/anatomie et histologie ,
15.
Nat Commun ; 9(1): 5460, 2018 12 19.
Article de Anglais | MEDLINE | ID: mdl-30568165

RÉSUMÉ

The original version of this Article contained an error in Fig. 2. In panel a, the two legend items "rare" and "common" were inadvertently swapped. This has been corrected in both the PDF and HTML versions of the Article.

16.
Nat Commun ; 9(1): 4674, 2018 11 07.
Article de Anglais | MEDLINE | ID: mdl-30405126

RÉSUMÉ

The role of rare variants in complex traits remains uncharted. Here, we conduct deep whole genome sequencing of 1457 individuals from an isolated population, and test for rare variant burdens across six cardiometabolic traits. We identify a role for rare regulatory variation, which has hitherto been missed. We find evidence of rare variant burdens that are independent of established common variant signals (ADIPOQ and adiponectin, P = 4.2 × 10-8; APOC3 and triglyceride levels, P = 1.5 × 10-26), and identify replicating evidence for a burden associated with triglyceride levels in FAM189B (P = 2.2 × 10-8), indicating a role for this gene in lipid metabolism.


Sujet(s)
Allèles , Caractère quantitatif héréditaire , Séquençage du génome entier , Études de cohortes , Fréquence d'allèle/génétique , Variation génétique , Humains
18.
Methods Mol Biol ; 1793: 25-36, 2018.
Article de Anglais | MEDLINE | ID: mdl-29876889

RÉSUMÉ

Thorough data quality control (QC) is a key step to the success of high-throughput genotyping approaches. Following extensive research several criteria and thresholds have been established for data QC at the sample and variant level. Sample QC is aimed at the identification and removal (when appropriate) of individuals with (1) low call rate, (2) discrepant sex or other identity-related information, (3) excess genome-wide heterozygosity and homozygosity, (4) relations to other samples, (5) ethnicity differences, (6) batch effects, and (7) contamination. Variant QC is aimed at identification and removal or refinement of variants with (1) low call rate, (2) call rate differences by phenotypic status, (3) gross deviation from Hardy-Weinberg Equilibrium (HWE), (4) bad genotype intensity plots, (5) batch effects, (6) differences in allele frequencies with published data sets, (7) very low minor allele counts (MAC), (8) low imputation quality score, (9) low variant quality score log-odds, and (10) few or low quality reads.


Sujet(s)
Variation génétique , Étude d'association pangénomique , Contrôle de qualité , Animaux , Génome , Étude d'association pangénomique/méthodes , Étude d'association pangénomique/normes , Génomique/méthodes , Génotype , Humains , Phénotype
19.
Lancet Haematol ; 5(6): e241-e251, 2018 Jun.
Article de Anglais | MEDLINE | ID: mdl-29780001

RÉSUMÉ

BACKGROUND: There are more than 300 known red blood cell (RBC) antigens and 33 platelet antigens that differ between individuals. Sensitisation to antigens is a serious complication that can occur in prenatal medicine and after blood transfusion, particularly for patients who require multiple transfusions. Although pre-transfusion compatibility testing largely relies on serological methods, reagents are not available for many antigens. Methods based on single-nucleotide polymorphism (SNP) arrays have been used, but typing for ABO and Rh-the most important blood groups-cannot be done with SNP typing alone. We aimed to develop a novel method based on whole-genome sequencing to identify RBC and platelet antigens. METHODS: This whole-genome sequencing study is a subanalysis of data from patients in the whole-genome sequencing arm of the MedSeq Project randomised controlled trial (NCT01736566) with no measured patient outcomes. We created a database of molecular changes in RBC and platelet antigens and developed an automated antigen-typing algorithm based on whole-genome sequencing (bloodTyper). This algorithm was iteratively improved to address cis-trans haplotype ambiguities and homologous gene alignments. Whole-genome sequencing data from 110 MedSeq participants (30 × depth) were used to initially validate bloodTyper through comparison with conventional serology and SNP methods for typing of 38 RBC antigens in 12 blood-group systems and 22 human platelet antigens. bloodTyper was further validated with whole-genome sequencing data from 200 INTERVAL trial participants (15 × depth) with serological comparisons. FINDINGS: We iteratively improved bloodTyper by comparing its typing results with conventional serological and SNP typing in three rounds of testing. The initial whole-genome sequencing typing algorithm was 99·5% concordant across the first 20 MedSeq genomes. Addressing discordances led to development of an improved algorithm that was 99·8% concordant for the remaining 90 MedSeq genomes. Additional modifications led to the final algorithm, which was 99·2% concordant across 200 INTERVAL genomes (or 99·9% after adjustment for the lower depth of coverage). INTERPRETATION: By enabling more precise antigen-matching of patients with blood donors, antigen typing based on whole-genome sequencing provides a novel approach to improve transfusion outcomes with the potential to transform the practice of transfusion medicine. FUNDING: National Human Genome Research Institute, Doris Duke Charitable Foundation, National Health Service Blood and Transplant, National Institute for Health Research, and Wellcome Trust.


Sujet(s)
Système ABO de groupes sanguins/génétique , Antigènes plaquettaires humains/génétique , Groupage sanguin et épreuve de compatibilité croisée/méthodes , Système Rhésus/génétique , Séquençage du génome entier , Système ABO de groupes sanguins/classification , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Algorithmes , Antigènes plaquettaires humains/classification , Plaquettes/immunologie , Bases de données génétiques , Érythrocytes/immunologie , Génome humain , Humains , Adulte d'âge moyen , Essais contrôlés randomisés comme sujet , Système Rhésus/classification , Jeune adulte
20.
Am J Hum Genet ; 100(6): 865-884, 2017 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-28552196

RÉSUMÉ

Deep sequence-based imputation can enhance the discovery power of genome-wide association studies by assessing previously unexplored variation across the common- and low-frequency spectra. We applied a hybrid whole-genome sequencing (WGS) and deep imputation approach to examine the broader allelic architecture of 12 anthropometric traits associated with height, body mass, and fat distribution in up to 267,616 individuals. We report 106 genome-wide significant signals that have not been previously identified, including 9 low-frequency variants pointing to functional candidates. Of the 106 signals, 6 are in genomic regions that have not been implicated with related traits before, 28 are independent signals at previously reported regions, and 72 represent previously reported signals for a different anthropometric trait. 71% of signals reside within genes and fine mapping resolves 23 signals to one or two likely causal variants. We confirm genetic overlap between human monogenic and polygenic anthropometric traits and find signal enrichment in cis expression QTLs in relevant tissues. Our results highlight the potential of WGS strategies to enhance biologically relevant discoveries across the frequency spectrum.


Sujet(s)
Anthropométrie , Génome humain , Étude d'association pangénomique , Locus de caractère quantitatif/génétique , Analyse de séquence d'ADN/méthodes , Taille/génétique , Études de cohortes , Méthylation de l'ADN/génétique , Bases de données génétiques , Femelle , Variation génétique , Humains , Lipodystrophie/génétique , Mâle , Méta-analyse comme sujet , Obésité/génétique , Cartographie physique de chromosome , Caractères sexuels , Syndrome , Royaume-Uni
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...