Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.859
Filtrar
Mais filtros

Eixos temáticos
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 185(23): 4409-4427.e18, 2022 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-36368308

RESUMO

Fully understanding autism spectrum disorder (ASD) genetics requires whole-genome sequencing (WGS). We present the latest release of the Autism Speaks MSSNG resource, which includes WGS data from 5,100 individuals with ASD and 6,212 non-ASD parents and siblings (total n = 11,312). Examining a wide variety of genetic variants in MSSNG and the Simons Simplex Collection (SSC; n = 9,205), we identified ASD-associated rare variants in 718/5,100 individuals with ASD from MSSNG (14.1%) and 350/2,419 from SSC (14.5%). Considering genomic architecture, 52% were nuclear sequence-level variants, 46% were nuclear structural variants (including copy-number variants, inversions, large insertions, uniparental isodisomies, and tandem repeat expansions), and 2% were mitochondrial variants. Our study provides a guidebook for exploring genotype-phenotype correlations in families who carry ASD-associated rare variants and serves as an entry point to the expanded studies required to dissect the etiology in the ∼85% of the ASD population that remain idiopathic.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Humanos , Transtorno do Espectro Autista/genética , Predisposição Genética para Doença , Variações do Número de Cópias de DNA/genética , Genômica
2.
Cell ; 182(5): 1214-1231.e11, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32888494

RESUMO

Blood cells play essential roles in human health, underpinning physiological processes such as immunity, oxygen transport, and clotting, which when perturbed cause a significant global health burden. Here we integrate data from UK Biobank and a large-scale international collaborative effort, including data for 563,085 European ancestry participants, and discover 5,106 new genetic variants independently associated with 29 blood cell phenotypes covering a range of variation impacting hematopoiesis. We holistically characterize the genetic architecture of hematopoiesis, assess the relevance of the omnigenic model to blood cell phenotypes, delineate relevant hematopoietic cell states influenced by regulatory genetic variants and gene networks, identify novel splice-altering variants mediating the associations, and assess the polygenic prediction potential for blood traits and clinical disorders at the interface of complex and Mendelian genetics. These results show the power of large-scale blood cell trait GWAS to interrogate clinically meaningful variants across a wide allelic spectrum of human variation.


Assuntos
Predisposição Genética para Doença/genética , Herança Multifatorial/genética , Feminino , Redes Reguladoras de Genes/genética , Estudo de Associação Genômica Ampla/métodos , Hematopoese/genética , Humanos , Masculino , Fenótipo , Polimorfismo de Nucleotídeo Único/genética
3.
Cell ; 173(7): 1705-1715.e16, 2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29906448

RESUMO

Schizophrenia and bipolar disorder are two distinct diagnoses that share symptomology. Understanding the genetic factors contributing to the shared and disorder-specific symptoms will be crucial for improving diagnosis and treatment. In genetic data consisting of 53,555 cases (20,129 bipolar disorder [BD], 33,426 schizophrenia [SCZ]) and 54,065 controls, we identified 114 genome-wide significant loci implicating synaptic and neuronal pathways shared between disorders. Comparing SCZ to BD (23,585 SCZ, 15,270 BD) identified four genomic regions including one with disorder-independent causal variants and potassium ion response genes as contributing to differences in biology between the disorders. Polygenic risk score (PRS) analyses identified several significant correlations within case-only phenotypes including SCZ PRS with psychotic features and age of onset in BD. For the first time, we discover specific loci that distinguish between BD and SCZ and identify polygenic components underlying multiple symptom dimensions. These results point to the utility of genetics to inform symptomology and potential treatment.


Assuntos
Transtorno Bipolar/genética , Estudo de Associação Genômica Ampla , Esquizofrenia/genética , Transtorno Bipolar/patologia , Estudos de Casos e Controles , Loci Gênicos , Humanos , Herança Multifatorial/genética , Razão de Chances , Fenótipo , Risco , Esquizofrenia/patologia , População Branca/genética
4.
Physiol Rev ; 103(3): 2039-2055, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-36634218

RESUMO

Genome-wide association studies (GWAS) aim to identify common genetic variants that are associated with traits and diseases. Since 2005, more than 5,000 GWAS have been published for almost as many traits. These studies have offered insights into the loci and genes underlying phenotypic traits, have highlighted genetic correlations across traits and diseases, and are beginning to demonstrate clinical utility by identifying individuals at increased risk for common diseases. GWAS have been widely utilized across cardiovascular diseases and associated phenotypic traits, with insights facilitated by multicenter registry studies and large biobank data sets. In this review, we describe how GWAS have informed the genetic architecture of cardiovascular diseases and the insights they have provided into disease pathophysiology, using archetypal conditions for both common and rare diseases. We also describe how biobank data sets can complement disease-specific studies, particularly for rarer cardiovascular diseases, and how findings from GWAS have the potential to impact on clinical care. Finally, we discuss the outstanding challenges facing research in this field and how they can be addressed.


Assuntos
Doenças Cardiovasculares , Estudo de Associação Genômica Ampla , Humanos , Doenças Cardiovasculares/genética , Fenótipo , Predisposição Genética para Doença , Estudos Multicêntricos como Assunto
5.
Annu Rev Genet ; 54: 189-211, 2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-32867542

RESUMO

Canalization refers to the evolution of populations such that the number of individuals who deviate from the optimum trait, or experience disease, is minimized. In the presence of rapid cultural, environmental, or genetic change, the reverse process of decanalization may contribute to observed increases in disease prevalence. This review starts by defining relevant concepts, drawing distinctions between the canalization of populations and robustness of individuals. It then considers evidence pertaining to three continuous traits and six domains of disease. In each case, existing genetic evidence for genotype-by-environment interactions is insufficient to support a strong inference of decanalization, but we argue that the advent of genome-wide polygenic risk assessment now makes an empirical evaluation of the role of canalization in preventing disease possible. Finally, the contributions of both rare and common variants to congenital abnormality and adult onset disease are considered in light of a new kerplunk model of genetic effects.


Assuntos
Doenças Genéticas Inatas/genética , Genoma/genética , Genética Humana/métodos , Variação Genética/genética , Genótipo , Humanos , Filogenia
6.
Annu Rev Pharmacol Toxicol ; 64: 33-51, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-37506333

RESUMO

Interindividual variability in genes encoding drug-metabolizing enzymes, transporters, receptors, and human leukocyte antigens has a major impact on a patient's response to drugs with regard to efficacy and safety. Enabled by both technological and conceptual advances, the field of pharmacogenomics is developing rapidly. Major progress in omics profiling methods has enabled novel genotypic and phenotypic characterization of patients and biobanks. These developments are paralleled by advances in machine learning, which have allowed us to parse the immense wealth of data and establish novel genetic markers and polygenic models for drug selection and dosing. Pharmacogenomics has recently become more widespread in clinical practice to personalize treatment and to develop new drugs tailored to specific patient populations. In this review, we provide an overview of the latest developments in the field and discuss the way forward, including how to address the missing heritability, develop novel polygenic models, and further improve the clinical implementation of pharmacogenomics.


Assuntos
Proteínas de Membrana Transportadoras , Farmacogenética , Humanos , Tecnologia
7.
Am J Hum Genet ; 111(2): 242-258, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-38211585

RESUMO

Tumor mutational burden (TMB), the total number of somatic mutations in the tumor, and copy number burden (CNB), the corresponding measure of aneuploidy, are established fundamental somatic features and emerging biomarkers for immunotherapy. However, the genetic and non-genetic influences on TMB/CNB and, critically, the manner by which they influence patient outcomes remain poorly understood. Here, we present a large germline-somatic study of TMB/CNB with >23,000 individuals across 17 cancer types, of which 12,000 also have extensive clinical, treatment, and overall survival (OS) measurements available. We report dozens of clinical associations with TMB/CNB, observing older age and male sex to have a strong effect on TMB and weaker impact on CNB. We additionally identified significant germline influences on TMB/CNB, including fine-scale European ancestry and germline polygenic risk scores (PRSs) for smoking, tanning, white blood cell counts, and educational attainment. We quantify the causal effect of exposures on somatic mutational processes using Mendelian randomization. Many of the identified features associated with TMB/CNB were additionally associated with OS for individuals treated at a single tertiary cancer center. For individuals receiving immunotherapy, we observed a complex relationship between PRSs for educational attainment, self-reported college attainment, TMB, and survival, suggesting that the influence of this biomarker may be substantially modified by socioeconomic status. While the accumulation of somatic alterations is a stochastic process, our work demonstrates that it can be shaped by host characteristics including germline genetics.


Assuntos
Neoplasias , Humanos , Masculino , Mutação/genética , Neoplasias/genética , Neoplasias/patologia , Imunoterapia , Biomarcadores Tumorais/genética , Células Germinativas/patologia
8.
Proc Natl Acad Sci U S A ; 121(33): e2403210121, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39110727

RESUMO

Polygenic risk scores (PRS) enhance population risk stratification and advance personalized medicine, but existing methods face several limitations, encompassing issues related to computational burden, predictive accuracy, and adaptability to a wide range of genetic architectures. To address these issues, we propose Aggregated L0Learn using Summary-level data (ALL-Sum), a fast and scalable ensemble learning method for computing PRS using summary statistics from genome-wide association studies (GWAS). ALL-Sum leverages a L0L2 penalized regression and ensemble learning across tuning parameters to flexibly model traits with diverse genetic architectures. In extensive large-scale simulations across a wide range of polygenicity and GWAS sample sizes, ALL-Sum consistently outperformed popular alternative methods in terms of prediction accuracy, runtime, and memory usage by 10%, 20-fold, and threefold, respectively, and demonstrated robustness to diverse genetic architectures. We validated the performance of ALL-Sum in real data analysis of 11 complex traits using GWAS summary statistics from nine data sources, including the Global Lipids Genetics Consortium, Breast Cancer Association Consortium, and FinnGen Biobank, with validation in the UK Biobank. Our results show that on average, ALL-Sum obtained PRS with 25% higher accuracy on average, with 15 times faster computation and half the memory than the current state-of-the-art methods, and had robust performance across a wide range of traits and diseases. Furthermore, our method demonstrates stable prediction when using linkage disequilibrium computed from different data sources. ALL-Sum is available as a user-friendly R software package with publicly available reference data for streamlined analysis.


Assuntos
Estudo de Associação Genômica Ampla , Herança Multifatorial , Humanos , Herança Multifatorial/genética , Estudo de Associação Genômica Ampla/métodos , Aprendizado de Máquina , Predisposição Genética para Doença , Polimorfismo de Nucleotídeo Único
9.
Hum Mol Genet ; 2024 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-38879759

RESUMO

Venous thromboembolism (VTE) is a significant contributor to morbidity and mortality, with large disparities in incidence rates between Black and White Americans. Polygenic risk scores (PRSs) limited to variants discovered in genome-wide association studies in European-ancestry samples can identify European-ancestry individuals at high risk of VTE. However, there is limited evidence on whether high-dimensional PRS constructed using more sophisticated methods and more diverse training data can enhance the predictive ability and their utility across diverse populations. We developed PRSs for VTE using summary statistics from the International Network against Venous Thrombosis (INVENT) consortium genome-wide association studies meta-analyses of European- (71 771 cases and 1 059 740 controls) and African-ancestry samples (7482 cases and 129 975 controls). We used LDpred2 and PRS-CSx to construct ancestry-specific and multi-ancestry PRSs and evaluated their performance in an independent European- (6781 cases and 103 016 controls) and African-ancestry sample (1385 cases and 12 569 controls). Multi-ancestry PRSs with weights tuned in European-ancestry samples slightly outperformed ancestry-specific PRSs in European-ancestry test samples (e.g. the area under the receiver operating curve [AUC] was 0.609 for PRS-CSx_combinedEUR and 0.608 for PRS-CSxEUR [P = 0.00029]). Multi-ancestry PRSs with weights tuned in African-ancestry samples also outperformed ancestry-specific PRSs in African-ancestry test samples (PRS-CSxAFR: AUC = 0.58, PRS-CSx_combined AFR: AUC = 0.59), although this difference was not statistically significant (P = 0.34). The highest fifth percentile of the best-performing PRS was associated with 1.9-fold and 1.68-fold increased risk for VTE among European- and African-ancestry subjects, respectively, relative to those in the middle stratum. These findings suggest that the multi-ancestry PRS might be used to improve performance across diverse populations to identify individuals at highest risk for VTE.

10.
Hum Mol Genet ; 33(14): 1262-1272, 2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38676403

RESUMO

BACKGROUND: Genetic susceptibility to various chronic diseases has been shown to influence heart failure (HF) risk. However, the underlying biological pathways, particularly the role of leukocyte telomere length (LTL), are largely unknown. We investigated the impact of genetic susceptibility to chronic diseases and various traits on HF risk, and whether LTL mediates or modifies the pathways. METHODS: We conducted prospective cohort analyses on 404 883 European participants from the UK Biobank, including 9989 incident HF cases. Multivariable Cox regression was used to estimate associations between HF risk and 24 polygenic risk scores (PRSs) for various diseases or traits previously generated using a Bayesian approach. We assessed multiplicative interactions between the PRSs and LTL previously measured in the UK Biobank using quantitative PCR. Causal mediation analyses were conducted to estimate the proportion of the total effect of PRSs acting indirectly through LTL, an integrative marker of biological aging. RESULTS: We identified 9 PRSs associated with HF risk, including those for various cardiovascular diseases or traits, rheumatoid arthritis (P = 1.3E-04), and asthma (P = 1.8E-08). Additionally, longer LTL was strongly associated with decreased HF risk (P-trend = 1.7E-08). Notably, LTL strengthened the asthma-HF relationship significantly (P-interaction = 2.8E-03). However, LTL mediated only 1.13% (P < 0.001) of the total effect of the asthma PRS on HF risk. CONCLUSIONS: Our findings shed light onto the shared genetic susceptibility between HF risk, asthma, rheumatoid arthritis, and other traits. Longer LTL strengthened the genetic effect of asthma in the pathway to HF. These results support consideration of LTL and PRSs in HF risk prediction.


Assuntos
Predisposição Genética para Doença , Insuficiência Cardíaca , Leucócitos , Telômero , Humanos , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/epidemiologia , Feminino , Leucócitos/metabolismo , Masculino , Pessoa de Meia-Idade , Telômero/genética , Doença Crônica , Idoso , Estudos Prospectivos , Homeostase do Telômero/genética , Fatores de Risco , Polimorfismo de Nucleotídeo Único , Adulto , Herança Multifatorial/genética , Estudo de Associação Genômica Ampla , População Branca/genética , População Europeia
11.
Trends Genet ; 39(2): 98-108, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36564319

RESUMO

Traditional classification of genetic diseases as monogenic and polygenic has lagged far behind scientific progress. In this opinion article, we propose and define a new terminology, genetically transitional disease (GTD), referring to cases where a large-effect mutation is necessary, but not sufficient, to cause disease. This leads to a working disease nosology based on gradients of four types of genetic architecture: monogenic, polygenic, GTD, and mixed. We present four scenarios under which GTD may occur; namely, subsets of traditionally Mendelian disease, modifiable Tier 1 monogenic conditions, variable penetrance, and situations where a genetic mutational spectrum produces qualitatively divergent pathologies. The implications of the new nosology in precision medicine are discussed, in which therapeutic options may target the molecular cause or the disease phenotype.


Assuntos
Medicina Genômica , Herança Multifatorial , Humanos , Fenótipo , Mutação , Herança Multifatorial/genética , Predisposição Genética para Doença
12.
Am J Hum Genet ; 110(7): 1138-1161, 2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-37339630

RESUMO

Autoimmunity and cancer represent two different aspects of immune dysfunction. Autoimmunity is characterized by breakdowns in immune self-tolerance, while impaired immune surveillance can allow for tumorigenesis. The class I major histocompatibility complex (MHC-I), which displays derivatives of the cellular peptidome for immune surveillance by CD8+ T cells, serves as a common genetic link between these conditions. As melanoma-specific CD8+ T cells have been shown to target melanocyte-specific peptide antigens more often than melanoma-specific antigens, we investigated whether vitiligo- and psoriasis-predisposing MHC-I alleles conferred a melanoma-protective effect. In individuals with cutaneous melanoma from both The Cancer Genome Atlas (n = 451) and an independent validation set (n = 586), MHC-I autoimmune-allele carrier status was significantly associated with a later age of melanoma diagnosis. Furthermore, MHC-I autoimmune-allele carriers were significantly associated with decreased risk of developing melanoma in the Million Veteran Program (OR = 0.962, p = 0.024). Existing melanoma polygenic risk scores (PRSs) did not predict autoimmune-allele carrier status, suggesting these alleles provide orthogonal risk-relevant information. Mechanisms of autoimmune protection were neither associated with improved melanoma-driver mutation association nor improved gene-level conserved antigen presentation relative to common alleles. However, autoimmune alleles showed higher affinity relative to common alleles for particular windows of melanocyte-conserved antigens and loss of heterozygosity of autoimmune alleles caused the greatest reduction in presentation for several conserved antigens across individuals with loss of HLA alleles. Overall, this study presents evidence that MHC-I autoimmune-risk alleles modulate melanoma risk unaccounted for by current PRSs.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Alelos , Melanoma/genética , Melanoma/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Neoplasias Cutâneas/genética , Histocompatibilidade , Antígenos de Histocompatibilidade Classe I/genética
13.
Am J Hum Genet ; 110(7): 1200-1206, 2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-37311464

RESUMO

Genome-wide polygenic risk scores (GW-PRSs) have been reported to have better predictive ability than PRSs based on genome-wide significance thresholds across numerous traits. We compared the predictive ability of several GW-PRS approaches to a recently developed PRS of 269 established prostate cancer-risk variants from multi-ancestry GWASs and fine-mapping studies (PRS269). GW-PRS models were trained with a large and diverse prostate cancer GWAS of 107,247 cases and 127,006 controls that we previously used to develop the multi-ancestry PRS269. Resulting models were independently tested in 1,586 cases and 1,047 controls of African ancestry from the California Uganda Study and 8,046 cases and 191,825 controls of European ancestry from the UK Biobank and further validated in 13,643 cases and 210,214 controls of European ancestry and 6,353 cases and 53,362 controls of African ancestry from the Million Veteran Program. In the testing data, the best performing GW-PRS approach had AUCs of 0.656 (95% CI = 0.635-0.677) in African and 0.844 (95% CI = 0.840-0.848) in European ancestry men and corresponding prostate cancer ORs of 1.83 (95% CI = 1.67-2.00) and 2.19 (95% CI = 2.14-2.25), respectively, for each SD unit increase in the GW-PRS. Compared to the GW-PRS, in African and European ancestry men, the PRS269 had larger or similar AUCs (AUC = 0.679, 95% CI = 0.659-0.700 and AUC = 0.845, 95% CI = 0.841-0.849, respectively) and comparable prostate cancer ORs (OR = 2.05, 95% CI = 1.87-2.26 and OR = 2.21, 95% CI = 2.16-2.26, respectively). Findings were similar in the validation studies. This investigation suggests that current GW-PRS approaches may not improve the ability to predict prostate cancer risk compared to the PRS269 developed from multi-ancestry GWASs and fine-mapping.


Assuntos
Predisposição Genética para Doença , Neoplasias da Próstata , Humanos , Masculino , População Negra/genética , Estudo de Associação Genômica Ampla , Herança Multifatorial/genética , Neoplasias da Próstata/genética , Fatores de Risco , População Branca/genética
14.
Am J Hum Genet ; 110(1): 13-22, 2023 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-36460009

RESUMO

Polygenic risk score (PRS) has demonstrated its great utility in biomedical research through identifying high-risk individuals for different diseases from their genotypes. However, the broader application of PRS to the general population is hindered by the limited transferability of PRS developed in Europeans to non-European populations. To improve PRS prediction accuracy in non-European populations, we develop a statistical method called SDPRX that can effectively integrate genome wide association study summary statistics from different populations. SDPRX automatically adjusts for linkage disequilibrium differences between populations and characterizes the joint distribution of the effect sizes of a variant in two populations to be both null, population specific, or shared with correlation. Through simulations and applications to real traits, we show that SDPRX improves the prediction performance over existing methods in non-European populations.


Assuntos
Estudo de Associação Genômica Ampla , Herança Multifatorial , Humanos , Herança Multifatorial/genética , Estudo de Associação Genômica Ampla/métodos , Predisposição Genética para Doença , Fatores de Risco , Genótipo
15.
Am J Hum Genet ; 110(7): 1207-1215, 2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-37379836

RESUMO

In polygenic score (PGS) analysis, the coefficient of determination (R2) is a key statistic to evaluate efficacy. R2 is the proportion of phenotypic variance explained by the PGS, calculated in a cohort that is independent of the genome-wide association study (GWAS) that provided estimates of allelic effect sizes. The SNP-based heritability (hSNP2, the proportion of total phenotypic variances attributable to all common SNPs) is the theoretical upper limit of the out-of-sample prediction R2. However, in real data analyses R2 has been reported to exceed hSNP2, which occurs in parallel with the observation that hSNP2 estimates tend to decline as the number of cohorts being meta-analyzed increases. Here, we quantify why and when these observations are expected. Using theory and simulation, we show that if heterogeneities in cohort-specific hSNP2 exist, or if genetic correlations between cohorts are less than one, hSNP2 estimates can decrease as the number of cohorts being meta-analyzed increases. We derive conditions when the out-of-sample prediction R2 will be greater than hSNP2 and show the validity of our derivations with real data from a binary trait (major depression) and a continuous trait (educational attainment). Our research calls for a better approach to integrating information from multiple cohorts to address issues of between-cohort heterogeneity.


Assuntos
Estudo de Associação Genômica Ampla , Polimorfismo de Nucleotídeo Único , Humanos , Polimorfismo de Nucleotídeo Único/genética , Herança Multifatorial/genética , Fenótipo , Simulação por Computador
16.
Am J Hum Genet ; 110(5): 741-761, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37030289

RESUMO

The advent of large-scale genome-wide association studies (GWASs) has motivated the development of statistical methods for phenotype prediction with single-nucleotide polymorphism (SNP) array data. These polygenic risk score (PRS) methods use a multiple linear regression framework to infer joint effect sizes of all genetic variants on the trait. Among the subset of PRS methods that operate on GWAS summary statistics, sparse Bayesian methods have shown competitive predictive ability. However, most existing Bayesian approaches employ Markov chain Monte Carlo (MCMC) algorithms, which are computationally inefficient and do not scale favorably to higher dimensions, for posterior inference. Here, we introduce variational inference of polygenic risk scores (VIPRS), a Bayesian summary statistics-based PRS method that utilizes variational inference techniques to approximate the posterior distribution for the effect sizes. Our experiments with 36 simulation configurations and 12 real phenotypes from the UK Biobank dataset demonstrated that VIPRS is consistently competitive with the state-of-the-art in prediction accuracy while being more than twice as fast as popular MCMC-based approaches. This performance advantage is robust across a variety of genetic architectures, SNP heritabilities, and independent GWAS cohorts. In addition to its competitive accuracy on the "White British" samples, VIPRS showed improved transferability when applied to other ethnic groups, with up to 1.7-fold increase in R2 among individuals of Nigerian ancestry for low-density lipoprotein (LDL) cholesterol. To illustrate its scalability, we applied VIPRS to a dataset of 9.6 million genetic markers, which conferred further improvements in prediction accuracy for highly polygenic traits, such as height.


Assuntos
Estudo de Associação Genômica Ampla , Herança Multifatorial , Humanos , Herança Multifatorial/genética , Estudo de Associação Genômica Ampla/métodos , Teorema de Bayes , Polimorfismo de Nucleotídeo Único/genética , Fatores de Risco , Predisposição Genética para Doença
17.
Am J Hum Genet ; 110(11): 1888-1902, 2023 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-37890495

RESUMO

Admixed individuals offer unique opportunities for addressing limited transferability in polygenic scores (PGSs), given the substantial trans-ancestry genetic correlation in many complex traits. However, they are rarely considered in PGS training, given the challenges in representing ancestry-matched linkage-disequilibrium reference panels for admixed individuals. Here we present inclusive PGS (iPGS), which captures ancestry-shared genetic effects by finding the exact solution for penalized regression on individual-level data and is thus naturally applicable to admixed individuals. We validate our approach in a simulation study across 33 configurations with varying heritability, polygenicity, and ancestry composition in the training set. When iPGS is applied to n = 237,055 ancestry-diverse individuals in the UK Biobank, it shows the greatest improvements in Africans by 48.9% on average across 60 quantitative traits and up to 50-fold improvements for some traits (neutrophil count, R2 = 0.058) over the baseline model trained on the same number of European individuals. When we allowed iPGS to use n = 284,661 individuals, we observed an average improvement of 60.8% for African, 11.6% for South Asian, 7.3% for non-British White, 4.8% for White British, and 17.8% for the other individuals. We further developed iPGS+refit to jointly model the ancestry-shared and -dependent genetic effects when heterogeneous genetic associations were present. For neutrophil count, for example, iPGS+refit showed the highest predictive performance in the African group (R2 = 0.115), which exceeds the best predictive performance for the White British group (R2 = 0.090 in the iPGS model), even though only 1.49% of individuals used in the iPGS training are of African ancestry. Our results indicate the power of including diverse individuals for developing more equitable PGS models.


Assuntos
Herança Multifatorial , População Branca , Humanos , Herança Multifatorial/genética , População Branca/genética , Fenótipo , População Negra/genética , Povo Asiático/genética , Estudo de Associação Genômica Ampla/métodos
18.
Am J Hum Genet ; 110(10): 1673-1689, 2023 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-37716346

RESUMO

Accurate polygenic scores (PGSs) facilitate the genetic prediction of complex traits and aid in the development of personalized medicine. Here, we develop a statistical method called multi-trait assisted PGS (mtPGS), which can construct accurate PGSs for a target trait of interest by leveraging multiple traits relevant to the target trait. Specifically, mtPGS borrows SNP effect size similarity information between the target trait and its relevant traits to improve the effect size estimation on the target trait, thus achieving accurate PGSs. In the process, mtPGS flexibly models the shared genetic architecture between the target and the relevant traits to achieve robust performance, while explicitly accounting for the environmental covariance among them to accommodate different study designs with various sample overlap patterns. In addition, mtPGS uses only summary statistics as input and relies on a deterministic algorithm with several algebraic techniques for scalable computation. We evaluate the performance of mtPGS through comprehensive simulations and applications to 25 traits in the UK Biobank, where in the real data mtPGS achieves an average of 0.90%-52.91% accuracy gain compared to the state-of-the-art PGS methods. Overall, mtPGS represents an accurate, fast, and robust solution for PGS construction in biobank-scale datasets.


Assuntos
Estudo de Associação Genômica Ampla , Herança Multifatorial , Humanos , Herança Multifatorial/genética , Estudo de Associação Genômica Ampla/métodos , Fenótipo , Algoritmos , Projetos de Pesquisa
19.
Am J Hum Genet ; 110(5): 722-740, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37060905

RESUMO

Coronary artery disease (CAD) is a pandemic disease where up to half of the risk is explained by genetic factors. Advanced insights into the genetic basis of CAD require deeper understanding of the contributions of different cell types, molecular pathways, and genes to disease heritability. Here, we investigate the biological diversity of atherosclerosis-associated cell states and interrogate their contribution to the genetic risk of CAD by using single-cell and bulk RNA sequencing (RNA-seq) of mouse and human lesions. We identified 12 disease-associated cell states that we characterized further by gene set functional profiling, ligand-receptor prediction, and transcription factor inference. Importantly, Vcam1+ smooth muscle cell state genes contributed most to SNP-based heritability of CAD. In line with this, genetic variants near smooth muscle cell state genes and regulatory elements explained the largest fraction of CAD-risk variance between individuals. Using this information for variant prioritization, we derived a hybrid polygenic risk score (PRS) that demonstrated improved performance over a classical PRS. Our results provide insights into the biological mechanisms associated with CAD risk, which could make a promising contribution to precision medicine and tailored therapeutic interventions in the future.


Assuntos
Aterosclerose , Doença da Artéria Coronariana , Humanos , Aterosclerose/genética , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/patologia , Fatores de Risco , Regulação da Expressão Gênica , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla/métodos , Polimorfismo de Nucleotídeo Único/genética
20.
Brief Bioinform ; 25(3)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38770718

RESUMO

Polygenetic Risk Scores are used to evaluate an individual's vulnerability to developing specific diseases or conditions based on their genetic composition, by taking into account numerous genetic variations. This article provides an overview of the concept of Polygenic Risk Scores (PRS). We elucidate the historical advancements of PRS, their advantages and shortcomings in comparison with other predictive methods, and discuss their conceptual limitations in light of the complexity of biological systems. Furthermore, we provide a survey of published tools for computing PRS and associated resources. The various tools and software packages are categorized based on their technical utility for users or prospective developers. Understanding the array of available tools and their limitations is crucial for accurately assessing and predicting disease risks, facilitating early interventions, and guiding personalized healthcare decisions. Additionally, we also identify potential new avenues for future bioinformatic analyzes and advancements related to PRS.


Assuntos
Predisposição Genética para Doença , Herança Multifatorial , Software , Humanos , Biologia Computacional/métodos , Estudo de Associação Genômica Ampla/métodos , Fatores de Risco , Medição de Risco/métodos , Estratificação de Risco Genético
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa