Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Am Coll Cardiol ; 73(23): 2946-2957, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31196451

RESUMEN

BACKGROUND: Genetic variants currently known to affect coronary artery disease (CAD) risk explain less than one-quarter of disease heritability. The heritability contribution of gene regulatory networks (GRNs) in CAD, which are modulated by both genetic and environmental factors, is unknown. OBJECTIVES: This study sought to determine the heritability contributions of single nucleotide polymorphisms affecting gene expression (eSNPs) in GRNs causally linked to CAD. METHODS: Seven vascular and metabolic tissues collected in 2 independent genetics-of-gene-expression studies of patients with CAD were used to identify eSNPs and to infer coexpression networks. To construct GRNs with causal relations to CAD, the prior information of eSNPs in the coexpression networks was used in a Bayesian algorithm. Narrow-sense CAD heritability conferred by the GRNs was calculated from individual-level genotype data from 9 European genome-wide association studies (GWAS) (13,612 cases, 13,758 control cases). RESULTS: The authors identified and replicated 28 independent GRNs active in CAD. The genetic variation in these networks contributed to 10.0% of CAD heritability beyond the 22% attributable to risk loci identified by GWAS. GRNs in the atherosclerotic arterial wall (n = 7) and subcutaneous or visceral abdominal fat (n = 9) were most strongly implicated, jointly explaining 8.2% of CAD heritability. In all, these 28 GRNs (each contributing to >0.2% of CAD heritability) comprised 24 to 841 genes, whereof 1 to 28 genes had strong regulatory effects (key disease drivers) and harbored many relevant functions previously associated with CAD. The gene activity in these 28 GRNs also displayed strong associations with genetic and phenotypic cardiometabolic disease variations both in humans and mice, indicative of their pivotal roles as mediators of gene-environmental interactions in CAD. CONCLUSIONS: GRNs capture a major portion of genetic variance and contribute to heritability beyond that of genetic loci currently known to affect CAD risk. These networks provide a framework to identify novel risk genes/pathways and study molecular interactions within and across disease-relevant tissues leading to CAD.


Asunto(s)
Enfermedad de la Arteria Coronaria/epidemiología , Enfermedad de la Arteria Coronaria/genética , Redes Reguladoras de Genes/genética , Estudio de Asociación del Genoma Completo/métodos , Polimorfismo de Nucleótido Simple/genética , Tejido Adiposo/patología , Tejido Adiposo/fisiología , Animales , Enfermedad de la Arteria Coronaria/diagnóstico , Endotelio Vascular/patología , Endotelio Vascular/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Suecia/epidemiología
2.
Circ Genom Precis Med ; 12(6): e002390, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31059280

RESUMEN

BACKGROUND: Antiretroviral therapy (ART) for HIV infection increases risk for coronary artery disease (CAD), presumably by causing dyslipidemia and increased atherosclerosis. We applied systems pharmacology to identify and validate specific regulatory gene networks through which ART drugs may promote CAD. METHODS: Transcriptional responses of human cell lines to 15 ART drugs retrieved from the Library of Integrated Cellular Signatures (overall 1127 experiments) were used to establish consensus ART gene/transcriptional signatures. Next, enrichments of differentially expressed genes and gene-gene connectivity within these ART-consensus signatures were sought in 30 regulatory gene networks associated with CAD and CAD-related phenotypes in the Stockholm Atherosclerosis Gene Expression study. RESULTS: Ten of 15 ART signatures were significantly enriched both for differential expression and connectivity in a specific atherosclerotic arterial wall regulatory gene network (AR-RGN) causal for CAD involving RNA processing genes. An atherosclerosis in vitro model of cholestryl ester-loaded foam cells was then used for experimental validation. Treatments of these foam cells with ritonavir, nelfinavir, and saquinavir at least doubled cholestryl ester accumulation ( P=0.02, 0.0009, and 0.02, respectively), whereas RNA silencing of the AR-RGN top key driver, PQBP1 (polyglutamine binding protein 1), significantly curbed cholestryl ester accumulation following treatment with any of these ART drugs by >37% ( P<0.05). CONCLUSIONS: By applying a novel systems pharmacology data analysis framework, 3 commonly used ARTs (ritonavir, nelfinavir, and saquinavir) were found altering the activity of AR-RGN, a regulatory gene network promoting foam cell formation and risk of CAD. Targeting AR-RGN or its top key driver PQBP1 may help reduce CAD side effects of these ART drugs.


Asunto(s)
Antirretrovirales/farmacología , Enfermedad de la Arteria Coronaria/genética , Proteínas de Unión al ADN/metabolismo , Redes Reguladoras de Genes/efectos de los fármacos , Antirretrovirales/efectos adversos , Arterias/metabolismo , Aterosclerosis/genética , Aterosclerosis/metabolismo , Ésteres del Colesterol/sangre , Ésteres del Colesterol/genética , Enfermedad de la Arteria Coronaria/metabolismo , Proteínas de Unión al ADN/genética , Bases de Datos de Ácidos Nucleicos , Células Espumosas/efectos de los fármacos , Células Espumosas/metabolismo , Infecciones por VIH/tratamiento farmacológico , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Nelfinavir/efectos adversos , Nelfinavir/farmacología , Ritonavir/efectos adversos , Ritonavir/farmacología , Saquinavir/efectos adversos , Saquinavir/farmacología , Células THP-1
3.
Sci Rep ; 8(1): 3434, 2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29467471

RESUMEN

Genome-wide association studies (GWAS) have identified over two hundred chromosomal loci that modulate risk of coronary artery disease (CAD). The genes affected by variants at these loci are largely unknown and an untapped resource to improve our understanding of CAD pathophysiology and identify potential therapeutic targets. Here, we prioritized 68 genes as the most likely causal genes at genome-wide significant loci identified by GWAS of CAD and examined their regulatory roles in 286 metabolic and vascular tissue gene-protein sub-networks ("modules"). The modules and genes within were scored for CAD druggability potential. The scoring enriched for targets of cardiometabolic drugs currently in clinical use and in-depth analysis of the top-scoring modules validated established and revealed novel target tissues, biological processes, and druggable targets. This study provides an unprecedented resource of tissue-defined gene-protein interactions directly affected by genetic variance in CAD risk loci.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Redes Reguladoras de Genes , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Descubrimiento de Drogas , Redes Reguladoras de Genes/efectos de los fármacos , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Desequilibrio de Ligamiento , Terapia Molecular Dirigida , Polimorfismo de Nucleótido Simple/efectos de los fármacos , Sitios de Carácter Cuantitativo/efectos de los fármacos
4.
Science ; 353(6301): 827-30, 2016 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-27540175

RESUMEN

Genome-wide association studies (GWAS) have identified hundreds of cardiometabolic disease (CMD) risk loci. However, they contribute little to genetic variance, and most downstream gene-regulatory mechanisms are unknown. We genotyped and RNA-sequenced vascular and metabolic tissues from 600 coronary artery disease patients in the Stockholm-Tartu Atherosclerosis Reverse Networks Engineering Task study (STARNET). Gene expression traits associated with CMD risk single-nucleotide polymorphism (SNPs) identified by GWAS were more extensively found in STARNET than in tissue- and disease-unspecific gene-tissue expression studies, indicating sharing of downstream cis-/trans-gene regulation across tissues and CMDs. In contrast, the regulatory effects of other GWAS risk SNPs were tissue-specific; abdominal fat emerged as an important gene-regulatory site for blood lipids, such as for the low-density lipoprotein cholesterol and coronary artery disease risk gene PCSK9 STARNET provides insights into gene-regulatory mechanisms for CMD risk loci, facilitating their translation into opportunities for diagnosis, therapy, and prevention.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Regulación de la Expresión Génica , Grasa Abdominal/metabolismo , Enfermedad de Alzheimer/genética , LDL-Colesterol/sangre , LDL-Colesterol/genética , Enfermedad de la Arteria Coronaria/epidemiología , Femenino , Redes Reguladoras de Genes , Estudio de Asociación del Genoma Completo , Humanos , Hígado/metabolismo , Masculino , Músculo Esquelético/metabolismo , Especificidad de Órganos/genética , Polimorfismo de Nucleótido Simple , Proproteína Convertasa 9 , Proproteína Convertasas/genética , Sitios de Carácter Cuantitativo , Riesgo , Serina Endopeptidasas/genética
5.
Cell Syst ; 2(3): 196-208, 2016 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-27135365

RESUMEN

Inferring molecular networks can reveal how genetic perturbations interact with environmental factors to cause common complex diseases. We analyzed genetic and gene expression data from seven tissues relevant to coronary artery disease (CAD) and identified regulatory gene networks (RGNs) and their key drivers. By integrating data from genome-wide association studies, we identified 30 CAD-causal RGNs interconnected in vascular and metabolic tissues, and we validated them with corresponding data from the Hybrid Mouse Diversity Panel. As proof of concept, by targeting the key drivers AIP, DRAP1, POLR2I, and PQBP1 in a cross-species-validated, arterial-wall RGN involving RNA-processing genes, we re-identified this RGN in THP-1 foam cells and independent data from CAD macrophages and carotid lesions. This characterization of the molecular landscape in CAD will help better define the regulation of CAD candidate genes identified by genome-wide association studies and is a first step toward achieving the goals of precision medicine.


Asunto(s)
Redes Reguladoras de Genes , Animales , Proteínas Portadoras , Enfermedad de la Arteria Coronaria , Proteínas de Unión al ADN , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Proteínas Nucleares , Proteínas Represoras
6.
Circ Cardiovasc Genet ; 8(2): 305-15, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25578447

RESUMEN

BACKGROUND: Despite recent discoveries of new genetic risk factors, the majority of risk for coronary artery disease (CAD) remains elusive. As the most proximal sensor of DNA variation, RNA abundance can help identify subpopulations of genetic variants active in and across tissues mediating CAD risk through gene expression. METHODS AND RESULTS: By generating new genomic data on DNA and RNA samples from the Stockholm Atherosclerosis Gene Expression (STAGE) study, 8156 cis-acting expression quantitative trait loci (eQTLs) for 6450 genes across 7 CAD-relevant tissues were detected. The inherited risk enrichments of tissue-defined sets of these eQTLs were assessed using 2 independent genome-wide association data sets. eQTLs acting across increasing numbers of tissues were found increasingly enriched for CAD risk and resided at regulatory hot spots. The risk enrichment of 42 eQTLs acting across 5 to 6 tissues was particularly high (≤7.3-fold) and confirmed in the combined genome-wide association data from Coronary Artery Disease Genome Wide Replication And Meta-Analysis Consortium. Sixteen of the 42 eQTLs associated with 19 master regulatory genes and 29 downstream gene sets (n>30) were further risk enriched comparable to that of the 153 genome-wide association risk single-nucleotide polymorphisms established for CAD (8.4-fold versus 10-fold). Three gene sets, governed by the master regulators FLYWCH1, PSORSIC3, and G3BP1, segregated the STAGE patients according to extent of CAD, and small interfering RNA targeting of these master regulators affected cholesterol-ester accumulation in foam cells of the THP1 monocytic cell line. CONCLUSIONS: eQTLs acting across multiple tissues are significant carriers of inherited risk for CAD. FLYWCH1, PSORSIC3, and G3BP1 are novel master regulatory genes in CAD that may be suitable targets.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/metabolismo , Bases de Datos Genéticas , Regulación de la Expresión Génica , Proteínas Musculares , Sitios de Carácter Cuantitativo , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Especificidad de Órganos/genética
7.
Arterioscler Thromb Vasc Biol ; 34(9): 2068-77, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24925974

RESUMEN

OBJECTIVE: Using a multi-tissue, genome-wide gene expression approach, we recently identified a gene module linked to the extent of human atherosclerosis. This atherosclerosis module was enriched with inherited risk for coronary and carotid artery disease (CAD) and overlapped with genes in the transendothelial migration of leukocyte (TEML) pathway. Among the atherosclerosis module genes, the transcription cofactor Lim domain binding 2 (LDB2) was the most connected in a CAD vascular wall regulatory gene network. Here, we used human genomics and atherosclerosis-prone mice to evaluate the possible role of LDB2 in TEML and atherosclerosis. APPROACH AND RESULTS: mRNA profiles generated from blood macrophages in patients with CAD were used to infer transcription factor regulatory gene networks; Ldlr(-/-)Apob(100/100) mice were used to study the effects of Ldb2 deficiency on TEML activity and atherogenesis. LDB2 was the most connected gene in a transcription factor regulatory network inferred from TEML and atherosclerosis module genes in CAD macrophages. In Ldlr(-/-)Apob(100/100) mice, loss of Ldb2 increased atherosclerotic lesion size ≈2-fold and decreased plaque stability. The exacerbated atherosclerosis was caused by increased TEML activity, as demonstrated in air-pouch and retinal vasculature models in vivo, by ex vivo perfusion of primary leukocytes, and by leukocyte migration in vitro. In THP1 cells, migration was increased by overexpression and decreased by small interfering RNA inhibition of LDB2. A functional LDB2 variant (rs10939673) was associated with the risk and extent of CAD across several cohorts. CONCLUSIONS: As a key driver of the TEML pathway in CAD macrophages, LDB2 is a novel candidate to target CAD by inhibiting the overall activity of TEML.


Asunto(s)
Aterosclerosis/fisiopatología , Enfermedades de las Arterias Carótidas/patología , Quimiotaxis de Leucocito/fisiología , Enfermedad de la Arteria Coronaria/patología , Proteínas con Dominio LIM/fisiología , Factores de Transcripción/fisiología , Migración Transendotelial y Transepitelial/fisiología , Animales , Apolipoproteína B-100/genética , Enfermedades de las Arterias Carótidas/genética , Línea Celular Tumoral , Quimiocina CCL2/farmacología , Enfermedad de la Arteria Coronaria/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Proteínas con Dominio LIM/deficiencia , Proteínas con Dominio LIM/genética , Macrófagos/metabolismo , Ratones , Ratones Noqueados , ARN Mensajero/biosíntesis , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Migración Transendotelial y Transepitelial/genética
8.
PLoS Genet ; 10(2): e1004201, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24586211

RESUMEN

Plasma cholesterol lowering (PCL) slows and sometimes prevents progression of atherosclerosis and may even lead to regression. Little is known about how molecular processes in the atherosclerotic arterial wall respond to PCL and modify responses to atherosclerosis regression. We studied atherosclerosis regression and global gene expression responses to PCL (≥80%) and to atherosclerosis regression itself in early, mature, and advanced lesions. In atherosclerotic aortic wall from Ldlr(-/-)Apob (100/100) Mttp (flox/flox)Mx1-Cre mice, atherosclerosis regressed after PCL regardless of lesion stage. However, near-complete regression was observed only in mice with early lesions; mice with mature and advanced lesions were left with regression-resistant, relatively unstable plaque remnants. Atherosclerosis genes responding to PCL before regression, unlike those responding to the regression itself, were enriched in inherited risk for coronary artery disease and myocardial infarction, indicating causality. Inference of transcription factor (TF) regulatory networks of these PCL-responsive gene sets revealed largely different networks in early, mature, and advanced lesions. In early lesions, PPARG was identified as a specific master regulator of the PCL-responsive atherosclerosis TF-regulatory network, whereas in mature and advanced lesions, the specific master regulators were MLL5 and SRSF10/XRN2, respectively. In a THP-1 foam cell model of atherosclerosis regression, siRNA targeting of these master regulators activated the time-point-specific TF-regulatory networks and altered the accumulation of cholesterol esters. We conclude that PCL leads to complete atherosclerosis regression only in mice with early lesions. Identified master regulators and related PCL-responsive TF-regulatory networks will be interesting targets to enhance PCL-mediated regression of mature and advanced atherosclerotic lesions.


Asunto(s)
Aorta/metabolismo , Aterosclerosis/sangre , Colesterol/sangre , Receptores de LDL/genética , Animales , Aorta/efectos de los fármacos , Apolipoproteínas B/genética , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/patología , Regulación de la Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/biosíntesis , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Ratones , Ratones Transgénicos , Proteínas Nucleares/biosíntesis , Ribonucleoproteínas/biosíntesis , Factores de Empalme Serina-Arginina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...