Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Biol ; 21(9): e3002311, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37695771

RESUMEN

Noncommunicable diseases (NCDs) are on the rise worldwide. Obesity, cardiovascular disease, and type 2 diabetes are among a long list of "lifestyle" diseases that were rare throughout human history but are now common. The evolutionary mismatch hypothesis posits that humans evolved in environments that radically differ from those we currently experience; consequently, traits that were once advantageous may now be "mismatched" and disease causing. At the genetic level, this hypothesis predicts that loci with a history of selection will exhibit "genotype by environment" (GxE) interactions, with different health effects in "ancestral" versus "modern" environments. To identify such loci, we advocate for combining genomic tools in partnership with subsistence-level groups experiencing rapid lifestyle change. In these populations, comparisons of individuals falling on opposite extremes of the "matched" to "mismatched" spectrum are uniquely possible. More broadly, the work we propose will inform our understanding of environmental and genetic risk factors for NCDs across diverse ancestries and cultures.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Humanos , Susceptibilidad a Enfermedades , Diabetes Mellitus Tipo 2/genética , Evolución Biológica , Genómica
2.
Am J Hum Genet ; 109(1): 24-32, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34861179

RESUMEN

Genetic correlation is an important parameter in efforts to understand the relationships among complex traits. Current methods that analyze individual genotype data for estimating genetic correlation are challenging to scale to large datasets. Methods that analyze summary data, while being computationally efficient, tend to yield estimates of genetic correlation with reduced precision. We propose SCORE (scalable genetic correlation estimator), a randomized method of moments estimator of genetic correlation that is both scalable and accurate. SCORE obtains more precise estimates of genetic correlations relative to summary-statistic methods that can be applied at scale; it achieves a 44% reduction in standard error relative to LD-score regression (LDSC) and a 20% reduction relative to high-definition likelihood (HDL) (averaged over all simulations). The efficiency of SCORE enables computation of genetic correlations on the UK Biobank dataset, consisting of ≈300 K individuals and ≈500 K SNPs, in a few h (orders of magnitude faster than methods that analyze individual data, such as GCTA). Across 780 pairs of traits in 291,273 unrelated white British individuals in the UK Biobank, SCORE identifies significant genetic correlation between 200 additional pairs of traits over LDSC (beyond the 245 pairs identified by both).


Asunto(s)
Bancos de Muestras Biológicas , Estudios de Asociación Genética , Antecedentes Genéticos , Modelos Genéticos , Fenotipo , Algoritmos , Variación Genética , Humanos , Herencia Multifactorial , Reproducibilidad de los Resultados , Reino Unido
3.
Am J Hum Genet ; 109(1): 66-80, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34995504

RESUMEN

Alternate splicing events can create isoforms that alter gene function, and genetic variants associated with alternate gene isoforms may reveal molecular mechanisms of disease. We used subcutaneous adipose tissue of 426 Finnish men from the METSIM study and identified splice junction quantitative trait loci (sQTLs) for 6,077 splice junctions (FDR < 1%). In the same individuals, we detected expression QTLs (eQTLs) for 59,443 exons and 15,397 genes (FDR < 1%). We identified 595 genes with an sQTL and exon eQTL but no gene eQTL, which could indicate potential isoform differences. Of the significant sQTL signals, 2,114 (39.8%) included at least one proxy variant (linkage disequilibrium r2 > 0.8) located within an intron spanned by the splice junction. We identified 203 sQTLs that colocalized with 141 genome-wide association study (GWAS) signals for cardiometabolic traits, including 25 signals for lipid traits, 24 signals for body mass index (BMI), and 12 signals for waist-hip ratio adjusted for BMI. Among all 141 GWAS signals colocalized with an sQTL, we detected 26 that also colocalized with an exon eQTL for an exon skipped by the sQTL splice junction. At a GWAS signal for high-density lipoprotein cholesterol colocalized with an NR1H3 sQTL splice junction, we show that the alternative splice product encodes an NR1H3 transcription factor that lacks a DNA binding domain and fails to activate transcription. Together, these results detect splicing events and candidate mechanisms that may contribute to gene function at GWAS loci.


Asunto(s)
Empalme Alternativo , Factores de Riesgo Cardiometabólico , Regulación de la Expresión Génica , Sitios de Carácter Cuantitativo , Carácter Cuantitativo Heredable , Grasa Subcutánea/metabolismo , Sitios de Unión , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Biología Computacional/métodos , Exones , Finlandia , Genes Reporteros , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genética de Población , Estudio de Asociación del Genoma Completo/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Receptores X del Hígado/genética , Masculino , Síndrome Metabólico/etiología , Síndrome Metabólico/metabolismo , Anotación de Secuencia Molecular , Fenotipo , Isoformas de Proteínas/genética , Sitios de Empalme de ARN , Proteínas de Unión al ARN
4.
Am J Hum Genet ; 108(3): 411-430, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33626337

RESUMEN

Genetic factors underlying coronary artery disease (CAD) have been widely studied using genome-wide association studies (GWASs). However, the functional understanding of the CAD loci has been limited by the fact that a majority of GWAS variants are located within non-coding regions with no functional role. High cholesterol and dysregulation of the liver metabolism such as non-alcoholic fatty liver disease confer an increased risk of CAD. Here, we studied the function of non-coding single-nucleotide polymorphisms in CAD GWAS loci located within liver-specific enhancer elements by identifying their potential target genes using liver cis-eQTL analysis and promoter Capture Hi-C in HepG2 cells. Altogether, 734 target genes were identified of which 121 exhibited correlations to liver-related traits. To identify potentially causal regulatory SNPs, the allele-specific enhancer activity was analyzed by (1) sequence-based computational predictions, (2) quantification of allele-specific transcription factor binding, and (3) STARR-seq massively parallel reporter assay. Altogether, our analysis identified 1,277 unique SNPs that display allele-specific regulatory activity. Among these, susceptibility enhancers near important cholesterol homeostasis genes (APOB, APOC1, APOE, and LIPA) were identified, suggesting that altered gene regulatory activity could represent another way by which genetic variation regulates serum lipoprotein levels. Using CRISPR-based perturbation, we demonstrate how the deletion/activation of a single enhancer leads to changes in the expression of many target genes located in a shared chromatin interaction domain. Our integrative genomics approach represents a comprehensive effort in identifying putative causal regulatory regions and target genes that could predispose to clinical manifestation of CAD by affecting liver function.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Elementos de Facilitación Genéticos/genética , Predisposición Genética a la Enfermedad , Sitios de Carácter Cuantitativo/genética , Alelos , Cromatina/genética , Enfermedad de la Arteria Coronaria/patología , Femenino , Estudio de Asociación del Genoma Completo/métodos , Genómica , Humanos , Hígado/metabolismo , Masculino , Anotación de Secuencia Molecular , Especificidad de Órganos/genética , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Factores de Riesgo
5.
Arterioscler Thromb Vasc Biol ; 43(10): 1788-1804, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37409528

RESUMEN

BACKGROUND: Adipocytes are crucial regulators of cardiovascular health. However, not much is known about gene expression profiles of adipocytes residing in nonfat cardiovascular tissues, their genetic regulation, and contribution to coronary artery disease. Here, we investigated whether and how the gene expression profiles of adipocytes in the subcutaneous adipose tissue differ from adipocytes residing in the heart. METHODS: We used single-nucleus RNA-sequencing data sets of subcutaneous adipose tissue and heart and performed in-depth analysis of tissue-resident adipocytes and their cell-cell interactions. RESULTS: We first discovered tissue-specific features of tissue-resident adipocytes, identified functional pathways involved in their tissue specificity, and found genes with cell type-specific expression enrichment in tissue-resident adipocytes. By following up these results, we discovered the propanoate metabolism pathway as a novel distinct characteristic of the heart-resident adipocytes and found a significant enrichment of coronary artery disease genome-wide association study risk variants among the right atrium-specific adipocyte marker genes. Our cell-cell communication analysis identified 22 specific heart adipocyte-associated ligand-receptor pairs and signaling pathways, including THBS (thrombospondin) and EPHA (ephrin type-A), further supporting the distinct tissue-resident role of heart adipocytes. Our results also suggest chamber-level coordination of heart adipocyte expression profiles as we observed a consistently larger number of adipocyte-associated ligand-receptor interactions and functional pathways in the atriums than ventricles. CONCLUSIONS: Overall, we introduce a new function and genetic link to coronary artery disease for the previously unexplored heart-resident adipocytes.


Asunto(s)
Enfermedad de la Arteria Coronaria , Propionatos , Humanos , Propionatos/metabolismo , ARN , Enfermedad de la Arteria Coronaria/metabolismo , Estudio de Asociación del Genoma Completo , Ligandos , Adipocitos/metabolismo , Análisis de Secuencia de ARN
6.
PLoS Genet ; 16(9): e1009018, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32925908

RESUMEN

Reverse causality has made it difficult to establish the causal directions between obesity and prediabetes and obesity and insulin resistance. To disentangle whether obesity causally drives prediabetes and insulin resistance already in non-diabetic individuals, we utilized the UK Biobank and METSIM cohort to perform a Mendelian randomization (MR) analyses in the non-diabetic individuals. Our results suggest that both prediabetes and systemic insulin resistance are caused by obesity (p = 1.2×10-3 and p = 3.1×10-24). As obesity reflects the amount of body fat, we next studied how adipose tissue affects insulin resistance. We performed both bulk RNA-sequencing and single nucleus RNA sequencing on frozen human subcutaneous adipose biopsies to assess adipose cell-type heterogeneity and mitochondrial (MT) gene expression in insulin resistance. We discovered that the adipose MT gene expression and body fat percent are both independently associated with insulin resistance (p≤0.05 for each) when adjusting for the decomposed adipose cell-type proportions. Next, we showed that these 3 factors, adipose MT gene expression, body fat percent, and adipose cell types, explain a substantial amount (44.39%) of variance in insulin resistance and can be used to predict it (p≤2.64×10-5 in 3 independent human cohorts). In summary, we demonstrated that obesity is a strong determinant of both prediabetes and insulin resistance, and discovered that individuals' adipose cell-type composition, adipose MT gene expression, and body fat percent predict their insulin resistance, emphasizing the critical role of adipose tissue in systemic insulin resistance.


Asunto(s)
Tejido Adiposo/metabolismo , Resistencia a la Insulina/fisiología , Obesidad/genética , Adipocitos/metabolismo , Adiposidad , Adulto , Índice de Masa Corporal , Estudios de Cohortes , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Humanos , Resistencia a la Insulina/genética , Masculino , Persona de Mediana Edad , Obesidad/fisiopatología , Estado Prediabético/metabolismo , Estado Prediabético/fisiopatología , Grasa Subcutánea/metabolismo
7.
PLoS Genet ; 16(9): e1009019, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32915782

RESUMEN

Loci identified in genome-wide association studies (GWAS) can include multiple distinct association signals. We sought to identify the molecular basis of multiple association signals for adiponectin, a hormone involved in glucose regulation secreted almost exclusively from adipose tissue, identified in the Metabolic Syndrome in Men (METSIM) study. With GWAS data for 9,262 men, four loci were significantly associated with adiponectin: ADIPOQ, CDH13, IRS1, and PBRM1. We performed stepwise conditional analyses to identify distinct association signals, a subset of which are also nearly independent (lead variant pairwise r2<0.01). Two loci exhibited allelic heterogeneity, ADIPOQ and CDH13. Of seven association signals at the ADIPOQ locus, two signals colocalized with adipose tissue expression quantitative trait loci (eQTLs) for three transcripts: trait-increasing alleles at one signal were associated with increased ADIPOQ and LINC02043, while trait-increasing alleles at the other signal were associated with decreased ADIPOQ-AS1. In reporter assays, adiponectin-increasing alleles at two signals showed corresponding directions of effect on transcriptional activity. Putative mechanisms for the seven ADIPOQ signals include a missense variant (ADIPOQ G90S), a splice variant, a promoter variant, and four enhancer variants. Of two association signals at the CDH13 locus, the first signal consisted of promoter variants, including the lead adipose tissue eQTL variant for CDH13, while a second signal included a distal intron 1 enhancer variant that showed ~2-fold allelic differences in transcriptional reporter activity. Fine-mapping and experimental validation demonstrated that multiple, distinct association signals at these loci can influence multiple transcripts through multiple molecular mechanisms.


Asunto(s)
Adiponectina/genética , Adiponectina/metabolismo , Tejido Adiposo/metabolismo , Alelos , Cadherinas/genética , Cadherinas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Frecuencia de los Genes/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo/métodos , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Masculino , Síndrome Metabólico/genética , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo/genética , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Am J Hum Genet ; 105(4): 773-787, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31564431

RESUMEN

Genome-wide association studies (GWASs) have identified thousands of genetic loci associated with cardiometabolic traits including type 2 diabetes (T2D), lipid levels, body fat distribution, and adiposity, although most causal genes remain unknown. We used subcutaneous adipose tissue RNA-seq data from 434 Finnish men from the METSIM study to identify 9,687 primary and 2,785 secondary cis-expression quantitative trait loci (eQTL; <1 Mb from TSS, FDR < 1%). Compared to primary eQTL signals, secondary eQTL signals were located further from transcription start sites, had smaller effect sizes, and were less enriched in adipose tissue regulatory elements compared to primary signals. Among 2,843 cardiometabolic GWAS signals, 262 colocalized by LD and conditional analysis with 318 transcripts as primary and conditionally distinct secondary cis-eQTLs, including some across ancestries. Of cardiometabolic traits examined for adipose tissue eQTL colocalizations, waist-hip ratio (WHR) and circulating lipid traits had the highest percentage of colocalized eQTLs (15% and 14%, respectively). Among alleles associated with increased cardiometabolic GWAS risk, approximately half (53%) were associated with decreased gene expression level. Mediation analyses of colocalized genes and cardiometabolic traits within the 434 individuals provided further evidence that gene expression influences variant-trait associations. These results identify hundreds of candidate genes that may act in adipose tissue to influence cardiometabolic traits.


Asunto(s)
Tejido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/genética , Expresión Génica , Obesidad/genética , Alelos , Índice de Masa Corporal , Finlandia , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Sitios de Carácter Cuantitativo , Relación Cintura-Cadera
9.
Int J Obes (Lond) ; 46(8): 1478-1486, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35589964

RESUMEN

BACKGROUND: COVID-19 severity varies widely. Although some demographic and cardio-metabolic factors, including age and obesity, are associated with increasing risk of severe illness, the underlying mechanism(s) are uncertain. SUBJECTS/METHODS: In a meta-analysis of three independent studies of 1471 participants in total, we investigated phenotypic and genetic factors associated with subcutaneous adipose tissue expression of Angiotensin I Converting Enzyme 2 (ACE2), measured by RNA-Seq, which acts as a receptor for SARS-CoV-2 cellular entry. RESULTS: Lower adipose tissue ACE2 expression was associated with multiple adverse cardio-metabolic health indices, including type 2 diabetes (T2D) (P = 9.14 × 10-6), obesity status (P = 4.81 × 10-5), higher serum fasting insulin (P = 5.32 × 10-4), BMI (P = 3.94 × 10-4), and lower serum HDL levels (P = 1.92 × 10-7). ACE2 expression was also associated with estimated proportions of cell types in adipose tissue: lower expression was associated with a lower proportion of microvascular endothelial cells (P = 4.25 × 10-4) and higher proportion of macrophages (P = 2.74 × 10-5). Despite an estimated heritability of 32%, we did not identify any proximal or distal expression quantitative trait loci (eQTLs) associated with adipose tissue ACE2 expression. CONCLUSIONS: Our results demonstrate that individuals with cardio-metabolic features known to increase risk of severe COVID-19 have lower background ACE2 levels in this highly relevant tissue. Reduced adipose tissue ACE2 expression may contribute to the pathophysiology of cardio-metabolic diseases, as well as the associated increased risk of severe COVID-19.


Asunto(s)
Tejido Adiposo , Enzima Convertidora de Angiotensina 2 , COVID-19 , Tejido Adiposo/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/complicaciones , COVID-19/genética , Factores de Riesgo Cardiometabólico , Diabetes Mellitus Tipo 2/genética , Células Endoteliales/metabolismo , Humanos , Obesidad , SARS-CoV-2
10.
PLoS Genet ; 15(4): e1008009, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30951530

RESUMEN

Recent and classical work has revealed biologically and medically significant subtypes in complex diseases and traits. However, relevant subtypes are often unknown, unmeasured, or actively debated, making automated statistical approaches to subtype definition valuable. We propose reverse GWAS (RGWAS) to identify and validate subtypes using genetics and multiple traits: while GWAS seeks the genetic basis of a given trait, RGWAS seeks to define trait subtypes with distinct genetic bases. Unlike existing approaches relying on off-the-shelf clustering methods, RGWAS uses a novel decomposition, MFMR, to model covariates, binary traits, and population structure. We use extensive simulations to show that modelling these features can be crucial for power and calibration. We validate RGWAS in practice by recovering a recently discovered stress subtype in major depression. We then show the utility of RGWAS by identifying three novel subtypes of metabolic traits. We biologically validate these metabolic subtypes with SNP-level tests and a novel polygenic test: the former recover known metabolic GxE SNPs; the latter suggests subtypes may explain substantial missing heritability. Crucially, statins, which are widely prescribed and theorized to increase diabetes risk, have opposing effects on blood glucose across metabolic subtypes, suggesting the subtypes have potential translational value.


Asunto(s)
Estudio de Asociación del Genoma Completo/métodos , Modelos Genéticos , Herencia Multifactorial , Fenotipo , Algoritmos , Glucemia/efectos de los fármacos , Glucemia/genética , Análisis por Conglomerados , Simulación por Computador , Enfermedad Coronaria/sangre , Enfermedad Coronaria/tratamiento farmacológico , Enfermedad Coronaria/genética , Trastorno Depresivo Mayor/clasificación , Trastorno Depresivo Mayor/genética , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Estudio de Asociación del Genoma Completo/estadística & datos numéricos , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lípidos/sangre , Polimorfismo de Nucleótido Simple , Estado Prediabético/genética , Sitios de Carácter Cuantitativo
11.
Hum Mol Genet ; 28(24): 4161-4172, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31691812

RESUMEN

Integration of genome-wide association study (GWAS) signals with expression quantitative trait loci (eQTL) studies enables identification of candidate genes. However, evaluating whether nearby signals may share causal variants, termed colocalization, is affected by the presence of allelic heterogeneity, different variants at the same locus impacting the same phenotype. We previously identified eQTL in subcutaneous adipose tissue from 770 participants in the Metabolic Syndrome in Men (METSIM) study and detected 15 eQTL signals that colocalized with GWAS signals for waist-hip ratio adjusted for body mass index (WHRadjBMI) from the Genetic Investigation of Anthropometric Traits consortium. Here, we reevaluated evidence of colocalization using two approaches, conditional analysis and the Bayesian test COLOC, and show that providing COLOC with approximate conditional summary statistics at multi-signal GWAS loci can reconcile disagreements in colocalization classification between the two tests. Next, we performed conditional analysis on the METSIM subcutaneous adipose tissue data to identify conditionally distinct or secondary eQTL signals. We used the two approaches to test for colocalization with WHRadjBMI GWAS signals and evaluated the differences in colocalization classification between the two tests. Through these analyses, we identified four GWAS signals colocalized with secondary eQTL signals for FAM13A, SSR3, GRB14 and FMO1. Thus, at loci with multiple eQTL and/or GWAS signals, analyzing each signal independently enabled additional candidate genes to be identified.


Asunto(s)
Tejido Adiposo/fisiología , Distribución de la Grasa Corporal , Estudio de Asociación del Genoma Completo/métodos , Síndrome Metabólico/genética , Sitios de Carácter Cuantitativo , Adulto , Teorema de Bayes , Índice de Masa Corporal , Femenino , Predisposición Genética a la Enfermedad , Humanos , Desequilibrio de Ligamiento , Masculino , Fenotipo , Polimorfismo de Nucleótido Simple , Grasa Subcutánea/metabolismo , Relación Cintura-Cadera/métodos
12.
Am J Hum Genet ; 103(4): 535-552, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30290150

RESUMEN

Although recent studies provide evidence for a common genetic basis between complex traits and Mendelian disorders, a thorough quantification of their overlap in a phenotype-specific manner remains elusive. Here, we have quantified the overlap of genes identified through large-scale genome-wide association studies (GWASs) for 62 complex traits and diseases with genes containing mutations known to cause 20 broad categories of Mendelian disorders. We identified a significant enrichment of genes linked to phenotypically matched Mendelian disorders in GWAS gene sets; of the total 1,240 comparisons, a higher proportion of phenotypically matched or related pairs (n = 50 of 92 [54%]) than phenotypically unmatched pairs (n = 27 of 1,148 [2%]) demonstrated significant overlap, confirming a phenotype-specific enrichment pattern. Further, we observed elevated GWAS effect sizes near genes linked to phenotypically matched Mendelian disorders. Finally, we report examples of GWAS variants localized at the transcription start site or physically interacting with the promoters of genes linked to phenotypically matched Mendelian disorders. Our results are consistent with the hypothesis that genes that are disrupted in Mendelian disorders are dysregulated by non-coding variants in complex traits and demonstrate how leveraging findings from related Mendelian disorders and functional genomic datasets can prioritize genes that are putatively dysregulated by local and distal non-coding GWAS variants.


Asunto(s)
Herencia Multifactorial/genética , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo/métodos , Humanos , Masculino , Fenotipo , Regiones Promotoras Genéticas/genética , Sitio de Iniciación de la Transcripción/fisiología
13.
Liver Int ; 41(4): 754-763, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33219609

RESUMEN

BACKGROUND & AIMS: Non-alcoholic fatty liver disease (NAFLD) has been associated with multiple metabolic abnormalities. By applying a non-targeted metabolomics approach, we aimed at investigating whether serum metabolite profile that associates with NAFLD would differ in its association with NAFLD-related metabolic risk factors. METHODS & RESULTS: A total of 233 subjects (mean ± SD: 48.3 ± 9.3 years old; BMI: 43.1 ± 5.4 kg/m2 ; 64 male) undergoing bariatric surgery were studied. Of these participants, 164 with liver histology could be classified as normal liver (n = 79), simple steatosis (SS, n = 40) or non-alcoholic steatohepatitis (NASH, n = 45). Among the identified fasting serum metabolites with higher levels in those with NASH when compared to those with normal phenotype were the aromatic amino acids (AAAs: tryptophan, tyrosine and phenylalanine), the branched-chain amino acids (BCAAs: leucine and isoleucine), a phosphatidylcholine (PC(16:0/16:1)) and uridine (all FDRp < 0.05). Only tryptophan was significantly higher in those with NASH compared to those with SS (FDRp < 0.05). Only the AAAs tryptophan and tyrosine correlated positively with serum total and LDL cholesterol (FDRp < 0.1), and accordingly, with liver LDLR at mRNA expression level. In addition, tryptophan was the single AA associated with liver DNA methylation of CpG sites known to be differentially methylated in those with NASH. CONCLUSIONS: We found that serum levels of the NASH-related AAAs and BCAAs demonstrate divergent associations with serum lipids. The specific correlation of tryptophan with LDL-c may result from the molecular events affecting LDLR mRNA expression and NASH-associated methylation of genes in the liver.


Asunto(s)
Cirugía Bariátrica , Enfermedad del Hígado Graso no Alcohólico , Adulto , Aminoácidos de Cadena Ramificada , Humanos , Masculino , Persona de Mediana Edad , Fosfatidilcolinas
14.
Am J Hum Genet ; 100(3): 428-443, 2017 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-28257690

RESUMEN

Subcutaneous adipose tissue stores excess lipids and maintains energy balance. We performed expression quantitative trait locus (eQTL) analyses by using abdominal subcutaneous adipose tissue of 770 extensively phenotyped participants of the METSIM study. We identified cis-eQTLs for 12,400 genes at a 1% false-discovery rate. Among an approximately 680 known genome-wide association study (GWAS) loci for cardio-metabolic traits, we identified 140 coincident cis-eQTLs at 109 GWAS loci, including 93 eQTLs not previously described. At 49 of these 140 eQTLs, gene expression was nominally associated (p < 0.05) with levels of the GWAS trait. The size of our dataset enabled identification of five loci associated (p < 5 × 10-8) with at least five genes located >5 Mb away. These trans-eQTL signals confirmed and extended the previously reported KLF14-mediated network to 55 target genes, validated the CIITA regulation of class II MHC genes, and identified ZNF800 as a candidate master regulator. Finally, we observed similar expression-clinical trait correlations of genes associated with GWAS loci in both humans and a panel of genetically diverse mice. These results provide candidate genes for further investigation of their potential roles in adipose biology and in regulating cardio-metabolic traits.


Asunto(s)
Enfermedades Cardiovasculares/genética , Regulación de la Expresión Génica , Síndrome Metabólico/genética , Sitios de Carácter Cuantitativo , Grasa Subcutánea/metabolismo , Anciano , Animales , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Técnicas de Genotipaje , Humanos , Masculino , Ratones , Persona de Mediana Edad , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenotipo , Reproducibilidad de los Resultados , Transactivadores/genética , Transactivadores/metabolismo
15.
Bioinformatics ; 34(8): 1313-1320, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29186329

RESUMEN

Motivation: Mapping bias causes preferential alignment to the reference allele, forming a major obstacle in allele-specific expression (ASE) analysis. The existing methods, such as simulation and SNP-aware alignment, are either inaccurate or relatively slow. To fast and accurately count allelic reads for ASE analysis, we developed a novel approach, ASElux, which utilizes the personal SNP information and counts allelic reads directly from unmapped RNA-sequence (RNA-seq) data. ASElux significantly reduces runtime by disregarding reads outside single nucleotide polymorphisms (SNPs) during the alignment. Results: When compared to other tools on simulated and experimental data, ASElux achieves a higher accuracy on ASE estimation than non-SNP-aware aligners and requires a much shorter time than the benchmark SNP-aware aligner, GSNAP with just a slight loss in performance. ASElux can process 40 million read-pairs from an RNA-sequence (RNA-seq) sample and count allelic reads within 10 min, which is comparable to directly counting the allelic reads from alignments based on other tools. Furthermore, processing an RNA-seq sample using ASElux in conjunction with a general aligner, such as STAR, is more accurate and still ∼4× faster than STAR + WASP, and ∼33× faster than the lead SNP-aware aligner, GSNAP, making ASElux ideal for ASE analysis of large-scale transcriptomic studies. We applied ASElux to 273 lung RNA-seq samples from GTEx and identified a splice-QTL rs11078928 in lung which explains the mechanism underlying an asthma GWAS SNP rs11078927. Thus, our analysis demonstrated ASE as a highly powerful complementary tool to cis-expression quantitative trait locus (eQTL) analysis. Availability and implementation: The software can be downloaded from https://github.com/abl0719/ASElux. Contact: zmiao@ucla.edu or a5ko@ucla.edu. Supplementary information: Supplementary data are available at Bioinformatics online.


Asunto(s)
Alelos , Perfilación de la Expresión Génica/métodos , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Programas Informáticos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Análisis de Secuencia de ARN/métodos
16.
PLoS Genet ; 12(5): e1006078, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27227539

RESUMEN

Familial combined hyperlipidemia (FCH) is a complex and common familial dyslipidemia characterized by elevated total cholesterol and/or triglyceride levels with over five-fold risk of coronary heart disease. The genetic architecture and contribution of rare Mendelian and common variants to FCH susceptibility is unknown. In 53 Finnish FCH families, we genotyped and imputed nine million variants in 715 family members with DNA available. We studied the enrichment of variants previously implicated with monogenic dyslipidemias and/or lipid levels in the general population by comparing allele frequencies between the FCH families and population samples. We also constructed weighted polygenic scores using 212 lipid-associated SNPs and estimated the relative contributions of Mendelian variants and polygenic scores to the risk of FCH in the families. We identified, across the whole allele frequency spectrum, an enrichment of variants known to elevate, and a deficiency of variants known to lower LDL-C and/or TG levels among both probands and affected FCH individuals. The score based on TG associated SNPs was particularly high among affected individuals compared to non-affected family members. Out of 234 affected FCH individuals across the families, seven (3%) carried Mendelian variants and 83 (35%) showed high accumulation of either known LDL-C or TG elevating variants by having either polygenic score over the 90th percentile in the population. The positive predictive value of high score was much higher for affected FCH individuals than for similar sporadic cases in the population. FCH is highly polygenic, supporting the hypothesis that variants across the whole allele frequency spectrum contribute to this complex familial trait. Polygenic SNP panels improve identification of individuals affected with FCH, but their clinical utility remains to be defined.


Asunto(s)
Apolipoproteínas B/genética , Enfermedad de la Arteria Coronaria/genética , Dislipidemias/genética , Hiperlipidemia Familiar Combinada/genética , Adulto , HDL-Colesterol/sangre , HDL-Colesterol/genética , Enfermedad de la Arteria Coronaria/sangre , Dislipidemias/sangre , Dislipidemias/patología , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Hiperlipidemia Familiar Combinada/sangre , Hiperlipidemia Familiar Combinada/patología , Lipoproteínas LDL/sangre , Lipoproteínas LDL/genética , Masculino , Persona de Mediana Edad , Triglicéridos/sangre , Triglicéridos/genética
17.
Rev Invest Clin ; 70(5): 217-223, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30307448

RESUMEN

Cardiovascular disease (CVD) is a broad definition for diseases of the heart and blood vessels with high mortality and morbidity worldwide. Atherosclerosis and hypertension are the most common causes of CVD, and multiple factors confer the susceptibility. Some of the predisposing factors are modifiable such as diet, smoking, and exercise, whereas others, including age, sex, and individual's genetic variations contributing to the CVD composition traits, are non-modifiable. This latter group includes serum lipid traits. High serum lipid levels, specifically high levels of serum low-density lipoprotein cholesterol and triglycerides, are well-established key risk factors of atherosclerosis. This review will discuss genomics and systems biology approaches in the study of common dyslipidemias. The non-Mendelian forms of dyslipidemias are highly complex, and the molecular mechanisms underlying these polygenic lipid disorders are estimated to involve hundreds of genes. Interactions between the different genes and environmental factors also contribute to the clinical outcomes; however, very little is known about these interactions and their molecular mechanisms. To better address the complex genetic architecture and multiple properties leading to high serum lipid levels, networks and systems approach combining information at genomic, transcriptomics, methylomics, proteomics, metabolomics, and phenome level are being developed, with the ultimate goal to elucidate the cascade of dynamic changes leading to CVD in humans. (REV INVEST CLIN. 2018;70:217-23).


Asunto(s)
Dislipidemias/terapia , Genómica/métodos , Biología de Sistemas/métodos , Aterosclerosis/complicaciones , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/prevención & control , Dislipidemias/complicaciones , Humanos , Hipertensión/complicaciones , Lípidos/sangre , Factores de Riesgo
18.
J Lipid Res ; 58(3): 481-493, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28119442

RESUMEN

The Metabolic Syndrome in Men (METSIM) study is a population-based study including 10,197 Finnish men examined in 2005-2010. The aim of the study is to investigate nongenetic and genetic factors associated with the risk of T2D and CVD, and with cardiovascular risk factors. The protocol includes a detailed phenotyping of the participants, an oral glucose tolerance test, fasting laboratory measurements including proton NMR measurements, mass spectometry metabolomics, adipose tissue biopsies from 1,400 participants, and a stool sample. In our ongoing follow-up study, we have, to date, reexamined 6,496 participants. Extensive genotyping and exome sequencing have been performed for essentially all METSIM participants, and >2,000 METSIM participants have been whole-genome sequenced. We have identified several nongenetic markers associated with the development of diabetes and cardiovascular events, and participated in several genetic association studies to identify gene variants associated with diabetes, hyperglycemia, and cardiovascular risk factors. The generation of a phenotype and genotype resource in the METSIM study allows us to proceed toward a "systems genetics" approach, which includes statistical methods to quantitate and integrate intermediate phenotypes, such as transcript, protein, or metabolite levels, to provide a global view of the molecular architecture of complex traits.


Asunto(s)
Enfermedades Cardiovasculares/genética , Diabetes Mellitus Tipo 2/genética , Predisposición Genética a la Enfermedad , Síndrome Metabólico/genética , Metabolómica , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Anciano , Biopsia , Glucemia , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/patología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/patología , Estudios de Asociación Genética , Genoma Humano , Genotipo , Prueba de Tolerancia a la Glucosa , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hiperglucemia/sangre , Hiperglucemia/genética , Hiperglucemia/patología , Masculino , Síndrome Metabólico/sangre , Síndrome Metabólico/epidemiología , Síndrome Metabólico/patología , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Factores de Riesgo
19.
Arterioscler Thromb Vasc Biol ; 36(7): 1350-5, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27199446

RESUMEN

OBJECTIVE: We recently identified a locus on chromosome 18q11.2 for high serum triglycerides in Mexicans. We hypothesize that the lead genome-wide association study single-nucleotide polymorphism rs9949617, or its linkage disequilibrium proxies, regulates 1 of the 5 genes in the triglyceride-associated region. APPROACH AND RESULTS: We performed a linkage disequilibrium analysis and found 9 additional variants in linkage disequilibrium (r(2)>0.7) with the lead single-nucleotide polymorphism. To select the variants for functional analyses, we annotated the 10 variants using DNase I hypersensitive sites, transcription factor and chromatin states and identified rs17259126 as the lead candidate variant for functional in vitro validation. Using luciferase transcriptional reporter assay in liver HepG2 cells, we found that the G allele exhibits a significantly lower effect on transcription (P<0.05). The electrophoretic mobility shift and ChIPqPCR (chromatin immunoprecipitation coupled with quantitative polymerase chain reaction) assays confirmed that the minor G allele of rs17259126 disrupts an hepatocyte nuclear factor 4 α-binding site. To find the regional candidate gene, we performed a local expression quantitative trait locus analysis and found that rs17259126 and its linkage disequilibrium proxies alter expression of the regional transmembrane protein 241 (TMEM241) gene in 795 adipose RNAs from the Metabolic Syndrome In Men (METSIM) cohort (P=6.11×10(-07)-5.80×10(-04)). These results were replicated in expression profiles of TMEM241 from the Multiple Tissue Human Expression Resource (MuTHER; n=856). CONCLUSIONS: The Mexican genome-wide association study signal for high serum triglycerides on chromosome 18q11.2 harbors a regulatory single-nucleotide polymorphism, rs17259126, which disrupts normal hepatocyte nuclear factor 4 α binding and decreases the expression of the regional TMEM241 gene. Our data suggest that decreased transcript levels of TMEM241 contribute to increased triglyceride levels in Mexicans.


Asunto(s)
Cromosomas Humanos Par 18 , Factor Nuclear 4 del Hepatocito/genética , Metabolismo de los Lípidos/genética , Proteínas de la Membrana/genética , Polimorfismo de Nucleótido Simple , Triglicéridos/sangre , Anciano , Sitios de Unión , Finlandia , Genes Reporteros , Marcadores Genéticos , Estudio de Asociación del Genoma Completo , Genotipo , Células Hep G2 , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Desequilibrio de Ligamiento , Masculino , Proteínas de la Membrana/metabolismo , México , Persona de Mediana Edad , Fenotipo , Sitios de Carácter Cuantitativo , Transcripción Genética , Transfección , Estados Unidos , Regulación hacia Arriba
20.
Eur Heart J ; 36(36): 2425-37, 2015 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-26009596

RESUMEN

Familial hypercholesterolaemia (FH) is a common genetic cause of premature coronary heart disease (CHD). Globally, one baby is born with FH every minute. If diagnosed and treated early in childhood, individuals with FH can have normal life expectancy. This consensus paper aims to improve awareness of the need for early detection and management of FH children. Familial hypercholesterolaemia is diagnosed either on phenotypic criteria, i.e. an elevated low-density lipoprotein cholesterol (LDL-C) level plus a family history of elevated LDL-C, premature coronary artery disease and/or genetic diagnosis, or positive genetic testing. Childhood is the optimal period for discrimination between FH and non-FH using LDL-C screening. An LDL-C ≥5 mmol/L (190 mg/dL), or an LDL-C ≥4 mmol/L (160 mg/dL) with family history of premature CHD and/or high baseline cholesterol in one parent, make the phenotypic diagnosis. If a parent has a genetic defect, the LDL-C cut-off for the child is ≥3.5 mmol/L (130 mg/dL). We recommend cascade screening of families using a combined phenotypic and genotypic strategy. In children, testing is recommended from age 5 years, or earlier if homozygous FH is suspected. A healthy lifestyle and statin treatment (from age 8 to 10 years) are the cornerstones of management of heterozygous FH. Target LDL-C is <3.5 mmol/L (130 mg/dL) if >10 years, or ideally 50% reduction from baseline if 8-10 years, especially with very high LDL-C, elevated lipoprotein(a), a family history of premature CHD or other cardiovascular risk factors, balanced against the long-term risk of treatment side effects. Identifying FH early and optimally lowering LDL-C over the lifespan reduces cumulative LDL-C burden and offers health and socioeconomic benefits. To drive policy change for timely detection and management, we call for further studies in the young. Increased awareness, early identification, and optimal treatment from childhood are critical to adding decades of healthy life for children and adolescents with FH.


Asunto(s)
Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Adolescente , Adulto , Aterosclerosis/diagnóstico , Aterosclerosis/tratamiento farmacológico , Grosor Intima-Media Carotídeo , Niño , Técnicas de Laboratorio Clínico/métodos , Costo de Enfermedad , Consejo , Dieta , Suplementos Dietéticos , Diagnóstico Precoz , Economía Médica , Medicina Basada en la Evidencia , Femenino , Pruebas Genéticas , Heterocigoto , Homocigoto , Humanos , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/genética , Esperanza de Vida , Cumplimiento de la Medicación , Persona de Mediana Edad , Embarazo , Complicaciones del Embarazo/etiología , Factores de Riesgo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA