Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 26(13): 2577-2588, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28449029

RESUMEN

Periodontitis is one of the most common inflammatory diseases, with a prevalence of 11% worldwide for the severe forms and an estimated heritability of 50%. The disease is characterized by destruction of the alveolar bone due to an aberrant host inflammatory response to a dysbiotic oral microbiome. Previous genome-wide association studies (GWAS) have reported several suggestive susceptibility loci. Here, we conducted a GWAS using a German and Dutch case-control sample of aggressive periodontitis (AgP, 896 cases, 7,104 controls), a rare but highly severe and early-onset form of periodontitis, validated the associations in a German sample of severe forms of the more moderate phenotype chronic periodontitis (CP) (993 cases, 1,419 controls). Positive findings were replicated in a Turkish sample of AgP (223 cases, 564 controls). A locus at SIGLEC5 (sialic acid binding Ig-like lectin 5) and a chromosomal region downstream of the DEFA1A3 locus (defensin alpha 1-3) showed association with both disease phenotypes and were associated with periodontitis at a genome-wide significance level in the pooled samples, with P = 1.09E-08 (rs4284742,-G; OR = 1.34, 95% CI = 1.21-1.48) and P = 5.48E-10 (rs2738058,-T; OR = 1.28, 95% CI = 1.18-1.38), respectively. SIGLEC5 is expressed in various myeloid immune cells and classified as an inhibitory receptor with the potential to mediate tyrosine phosphatases SHP-1/-2 dependent signaling. Alpha defensins are antimicrobial peptides with expression in neutrophils and mucosal surfaces and a role in phagocyte-mediated host defense. This study identifies the first shared genetic risk loci of AgP and CP with genome-wide significance and highlights the role of innate and adaptive immunity in the etiology of periodontitis.


Asunto(s)
Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Periodontitis Crónica/genética , Lectinas/genética , Péptidos Cíclicos/genética , alfa-Defensinas/genética , Adulto , Periodontitis Agresiva/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Estudios de Casos y Controles , Femenino , Sitios Genéticos , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Lectinas/metabolismo , Masculino , Persona de Mediana Edad , Nucleótidos , Péptidos Cíclicos/metabolismo , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo , Turquía , alfa-Defensinas/metabolismo
2.
Nature ; 504(7480): 432-6, 2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24213632

RESUMEN

Myocardial infarction, a leading cause of death in the Western world, usually occurs when the fibrous cap overlying an atherosclerotic plaque in a coronary artery ruptures. The resulting exposure of blood to the atherosclerotic material then triggers thrombus formation, which occludes the artery. The importance of genetic predisposition to coronary artery disease and myocardial infarction is best documented by the predictive value of a positive family history. Next-generation sequencing in families with several affected individuals has revolutionized mutation identification. Here we report the segregation of two private, heterozygous mutations in two functionally related genes, GUCY1A3 (p.Leu163Phefs*24) and CCT7 (p.Ser525Leu), in an extended myocardial infarction family. GUCY1A3 encodes the α1 subunit of soluble guanylyl cyclase (α1-sGC), and CCT7 encodes CCTη, a member of the tailless complex polypeptide 1 ring complex, which, among other functions, stabilizes soluble guanylyl cyclase. After stimulation with nitric oxide, soluble guanylyl cyclase generates cGMP, which induces vasodilation and inhibits platelet activation. We demonstrate in vitro that mutations in both GUCY1A3 and CCT7 severely reduce α1-sGC as well as ß1-sGC protein content, and impair soluble guanylyl cyclase activity. Moreover, platelets from digenic mutation carriers contained less soluble guanylyl cyclase protein and consequently displayed reduced nitric-oxide-induced cGMP formation. Mice deficient in α1-sGC protein displayed accelerated thrombus formation in the microcirculation after local trauma. Starting with a severely affected family, we have identified a link between impaired soluble-guanylyl-cyclase-dependent nitric oxide signalling and myocardial infarction risk, possibly through accelerated thrombus formation. Reversing this defect may provide a new therapeutic target for reducing the risk of myocardial infarction.


Asunto(s)
Susceptibilidad a Enfermedades/metabolismo , Infarto del Miocardio/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal , Animales , Chaperonina con TCP-1/genética , Chaperonina con TCP-1/metabolismo , GMP Cíclico/metabolismo , Exoma/genética , Femenino , Predisposición Genética a la Enfermedad , Guanilato Ciclasa/deficiencia , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Mutación/genética , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Linaje , Activación Plaquetaria , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Reproducibilidad de los Resultados , Solubilidad , Guanilil Ciclasa Soluble , Trombosis/metabolismo , Vasodilatación
3.
Circulation ; 135(24): 2336-2353, 2017 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-28461624

RESUMEN

BACKGROUND: Common diseases such as coronary heart disease (CHD) are complex in etiology. The interaction of genetic susceptibility with lifestyle factors may play a prominent role. However, gene-lifestyle interactions for CHD have been difficult to identify. Here, we investigate interaction of smoking behavior, a potent lifestyle factor, with genotypes that have been shown to associate with CHD risk. METHODS: We analyzed data on 60 919 CHD cases and 80 243 controls from 29 studies for gene-smoking interactions for genetic variants at 45 loci previously reported to be associated with CHD risk. We also studied 5 loci associated with smoking behavior. Study-specific gene-smoking interaction effects were calculated and pooled using fixed-effects meta-analyses. Interaction analyses were declared to be significant at a P value of <1.0×10-3 (Bonferroni correction for 50 tests). RESULTS: We identified novel gene-smoking interaction for a variant upstream of the ADAMTS7 gene. Every T allele of rs7178051 was associated with lower CHD risk by 12% in never-smokers (P=1.3×10-16) in comparison with 5% in ever-smokers (P=2.5×10-4), translating to a 60% loss of CHD protection conferred by this allelic variation in people who smoked tobacco (interaction P value=8.7×10-5). The protective T allele at rs7178051 was also associated with reduced ADAMTS7 expression in human aortic endothelial cells and lymphoblastoid cell lines. Exposure of human coronary artery smooth muscle cells to cigarette smoke extract led to induction of ADAMTS7. CONCLUSIONS: Allelic variation at rs7178051 that associates with reduced ADAMTS7 expression confers stronger CHD protection in never-smokers than in ever-smokers. Increased vascular ADAMTS7 expression may contribute to the loss of CHD protection in smokers.


Asunto(s)
Enfermedad Coronaria/genética , Enfermedad Coronaria/prevención & control , Sitios Genéticos/genética , Predisposición Genética a la Enfermedad/genética , Fumar/genética , Proteína ADAMTS7/genética , Adulto , Anciano , Anciano de 80 o más Años , Células Cultivadas , Enfermedad Coronaria/epidemiología , Vasos Coronarios/patología , Vasos Coronarios/fisiología , Femenino , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Fumar/efectos adversos , Fumar/epidemiología
4.
N Engl J Med ; 372(17): 1608-18, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25853659

RESUMEN

BACKGROUND: The nature and underlying mechanisms of an inverse association between adult height and the risk of coronary artery disease (CAD) are unclear. METHODS: We used a genetic approach to investigate the association between height and CAD, using 180 height-associated genetic variants. We tested the association between a change in genetically determined height of 1 SD (6.5 cm) with the risk of CAD in 65,066 cases and 128,383 controls. Using individual-level genotype data from 18,249 persons, we also examined the risk of CAD associated with the presence of various numbers of height-associated alleles. To identify putative mechanisms, we analyzed whether genetically determined height was associated with known cardiovascular risk factors and performed a pathway analysis of the height-associated genes. RESULTS: We observed a relative increase of 13.5% (95% confidence interval [CI], 5.4 to 22.1; P<0.001) in the risk of CAD per 1-SD decrease in genetically determined height. There was a graded relationship between the presence of an increased number of height-raising variants and a reduced risk of CAD (odds ratio for height quartile 4 versus quartile 1, 0.74; 95% CI, 0.68 to 0.84; P<0.001). Of the 12 risk factors that we studied, we observed significant associations only with levels of low-density lipoprotein cholesterol and triglycerides (accounting for approximately 30% of the association). We identified several overlapping pathways involving genes associated with both development and atherosclerosis. CONCLUSIONS: There is a primary association between a genetically determined shorter height and an increased risk of CAD, a link that is partly explained by the association between shorter height and an adverse lipid profile. Shared biologic processes that determine achieved height and the development of atherosclerosis may explain some of the association. (Funded by the British Heart Foundation and others.).


Asunto(s)
Estatura/genética , Enfermedad de la Arteria Coronaria/genética , Variación Genética , Adulto , LDL-Colesterol/sangre , Enfermedad de la Arteria Coronaria/etiología , Humanos , Hiperlipidemias/complicaciones , Oportunidad Relativa , Factores de Riesgo , Triglicéridos/sangre
5.
PLoS Genet ; 11(7): e1005230, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26132169

RESUMEN

Reference panels from the 1000 Genomes (1000G) Project Consortium provide near complete coverage of common and low-frequency genetic variation with minor allele frequency ≥0.5% across European ancestry populations. Within the European Network for Genetic and Genomic Epidemiology (ENGAGE) Consortium, we have undertaken the first large-scale meta-analysis of genome-wide association studies (GWAS), supplemented by 1000G imputation, for four quantitative glycaemic and obesity-related traits, in up to 87,048 individuals of European ancestry. We identified two loci for body mass index (BMI) at genome-wide significance, and two for fasting glucose (FG), none of which has been previously reported in larger meta-analysis efforts to combine GWAS of European ancestry. Through conditional analysis, we also detected multiple distinct signals of association mapping to established loci for waist-hip ratio adjusted for BMI (RSPO3) and FG (GCK and G6PC2). The index variant for one association signal at the G6PC2 locus is a low-frequency coding allele, H177Y, which has recently been demonstrated to have a functional role in glucose regulation. Fine-mapping analyses revealed that the non-coding variants most likely to drive association signals at established and novel loci were enriched for overlap with enhancer elements, which for FG mapped to promoter and transcription factor binding sites in pancreatic islets, in particular. Our study demonstrates that 1000G imputation and genetic fine-mapping of common and low-frequency variant association signals at GWAS loci, integrated with genomic annotation in relevant tissues, can provide insight into the functional and regulatory mechanisms through which their effects on glycaemic and obesity-related traits are mediated.


Asunto(s)
Mapeo Cromosómico , Predisposición Genética a la Enfermedad , Índice Glucémico/genética , Obesidad/genética , Sitios de Carácter Cuantitativo/genética , Índice de Masa Corporal , Frecuencia de los Genes/genética , Estudio de Asociación del Genoma Completo , Quinasas del Centro Germinal , Glucosa-6-Fosfatasa/genética , Humanos , Polimorfismo de Nucleótido Simple/genética , Proteínas Serina-Treonina Quinasas/genética , Trombospondinas/genética
6.
PLoS Genet ; 10(7): e1004502, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25033284

RESUMEN

The majority of the heritability of coronary artery disease (CAD) remains unexplained, despite recent successes of genome-wide association studies (GWAS) in identifying novel susceptibility loci. Integrating functional genomic data from a variety of sources with a large-scale meta-analysis of CAD GWAS may facilitate the identification of novel biological processes and genes involved in CAD, as well as clarify the causal relationships of established processes. Towards this end, we integrated 14 GWAS from the CARDIoGRAM Consortium and two additional GWAS from the Ottawa Heart Institute (25,491 cases and 66,819 controls) with 1) genetics of gene expression studies of CAD-relevant tissues in humans, 2) metabolic and signaling pathways from public databases, and 3) data-driven, tissue-specific gene networks from a multitude of human and mouse experiments. We not only detected CAD-associated gene networks of lipid metabolism, coagulation, immunity, and additional networks with no clear functional annotation, but also revealed key driver genes for each CAD network based on the topology of the gene regulatory networks. In particular, we found a gene network involved in antigen processing to be strongly associated with CAD. The key driver genes of this network included glyoxalase I (GLO1) and peptidylprolyl isomerase I (PPIL1), which we verified as regulatory by siRNA experiments in human aortic endothelial cells. Our results suggest genetic influences on a diverse set of both known and novel biological processes that contribute to CAD risk. The key driver genes for these networks highlight potential novel targets for further mechanistic studies and therapeutic interventions.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Redes Reguladoras de Genes , Predisposición Genética a la Enfermedad , Transducción de Señal/genética , Animales , Enfermedad de la Arteria Coronaria/patología , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Genómica , Humanos , Ratones
7.
Arterioscler Thromb Vasc Biol ; 35(7): 1712-22, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25977570

RESUMEN

OBJECTIVE: Genome-wide association studies have identified multiple genetic variants affecting the risk of coronary artery disease (CAD). However, individually these explain only a small fraction of the heritability of CAD and for most, the causal biological mechanisms remain unclear. We sought to obtain further insights into potential causal processes of CAD by integrating large-scale GWA data with expertly curated databases of core human pathways and functional networks. APPROACHES AND RESULTS: Using pathways (gene sets) from Reactome, we carried out a 2-stage gene set enrichment analysis strategy. From a meta-analyzed discovery cohort of 7 CAD genome-wide association study data sets (9889 cases/11 089 controls), nominally significant gene sets were tested for replication in a meta-analysis of 9 additional studies (15 502 cases/55 730 controls) from the Coronary ARtery DIsease Genome wide Replication and Meta-analysis (CARDIoGRAM) Consortium. A total of 32 of 639 Reactome pathways tested showed convincing association with CAD (replication P<0.05). These pathways resided in 9 of 21 core biological processes represented in Reactome, and included pathways relevant to extracellular matrix (ECM) integrity, innate immunity, axon guidance, and signaling by PDRF (platelet-derived growth factor), NOTCH, and the transforming growth factor-ß/SMAD receptor complex. Many of these pathways had strengths of association comparable to those observed in lipid transport pathways. Network analysis of unique genes within the replicated pathways further revealed several interconnected functional and topologically interacting modules representing novel associations (eg, semaphoring-regulated axonal guidance pathway) besides confirming known processes (lipid metabolism). The connectivity in the observed networks was statistically significant compared with random networks (P<0.001). Network centrality analysis (degree and betweenness) further identified genes (eg, NCAM1, FYN, FURIN, etc) likely to play critical roles in the maintenance and functioning of several of the replicated pathways. CONCLUSIONS: These findings provide novel insights into how genetic variation, interpreted in the context of biological processes and functional interactions among genes, may help define the genetic architecture of CAD.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Estudio de Asociación del Genoma Completo , Enfermedad de la Arteria Coronaria/metabolismo , Humanos
9.
Mol Genet Genomics ; 290(5): 1911-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25893417

RESUMEN

Epidemiological evidence suggests that patients with celiac disease are at increased risk for coronary artery disease (CAD). Genetic-epidemiological analyses identified many single nucleotide polymorphisms (SNPs) associated with celiac disease. If there is a causal relation between celiac disease and CAD, one might expect that risk alleles primarily associated with celiac disease also increase the risk of CAD. In this study we identified from literature 41 SNPs that have been previously described to be genome-wide associated with celiac disease (p < 5 × 10(-08)). These SNPs were evaluated for their association with CAD in the Coronary ARtery DIsease Genome-wide Replication and Meta-analysis (CARDIoGRAM) dataset, a meta-analysis comprising genome-wide SNP association data from 22,233 CAD cases and 64,762 controls. 24 out of 41 (58.5 %) risk alleles for celiac disease displayed a positive association with CAD (CAD-OR range 1.001-1.081). The remaining risk alleles for celiac disease (n = 16) revealed CAD-ORs of ≤1.0 (range 0.951-1.0). The proportion of CAD associated alleles was greater but did not differ significantly from the proportion of 50 % expected by chance (p = 0.069). One SNP (rs653178 at the SH2B3/ATXN2 locus) displayed study-wise statistically significant association with CAD with directionality consistent effects on celiac disease and CAD. However, the effect of this locus is most likely driven by pleiotropic effects on multiple other diseases. In conclusion, this genetically based approach provided no convincing evidence that SNPs associated with celiac disease contribute to the risk of CAD. Hence, common non-genetic factors may play a more important role explaining the coincidence of these two complex disease conditions.


Asunto(s)
Enfermedad Celíaca/genética , Enfermedad de la Arteria Coronaria/genética , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , Humanos
10.
PLoS Genet ; 5(12): e1000768, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20011104

RESUMEN

The adipocyte-derived protein adiponectin is highly heritable and inversely associated with risk of type 2 diabetes mellitus (T2D) and coronary heart disease (CHD). We meta-analyzed 3 genome-wide association studies for circulating adiponectin levels (n = 8,531) and sought validation of the lead single nucleotide polymorphisms (SNPs) in 5 additional cohorts (n = 6,202). Five SNPs were genome-wide significant in their relationship with adiponectin (P< or =5x10(-8)). We then tested whether these 5 SNPs were associated with risk of T2D and CHD using a Bonferroni-corrected threshold of P< or =0.011 to declare statistical significance for these disease associations. SNPs at the adiponectin-encoding ADIPOQ locus demonstrated the strongest associations with adiponectin levels (P-combined = 9.2x10(-19) for lead SNP, rs266717, n = 14,733). A novel variant in the ARL15 (ADP-ribosylation factor-like 15) gene was associated with lower circulating levels of adiponectin (rs4311394-G, P-combined = 2.9x10(-8), n = 14,733). This same risk allele at ARL15 was also associated with a higher risk of CHD (odds ratio [OR] = 1.12, P = 8.5x10(-6), n = 22,421) more nominally, an increased risk of T2D (OR = 1.11, P = 3.2x10(-3), n = 10,128), and several metabolic traits. Expression studies in humans indicated that ARL15 is well-expressed in skeletal muscle. These findings identify a novel protein, ARL15, which influences circulating adiponectin levels and may impact upon CHD risk.


Asunto(s)
Adiponectina/sangre , Estudio de Asociación del Genoma Completo , Adiponectina/genética , Adiponectina/fisiología , Enfermedad Coronaria/genética , Diabetes Mellitus Tipo 2/genética , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Polimorfismo de Nucleótido Simple
11.
Eur Heart J ; 32(2): 158-68, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21088011

RESUMEN

AIMS: Recent genome-wide association (GWA) studies identified 10 chromosomal loci for coronary artery disease (CAD) or myocardial infarction (MI). However, these loci explain only a small proportion of the genetic variability of these pertinent diseases. We sought to identify additional CAD/MI loci by applying a three-stage approach. METHODS AND RESULTS: We genotyped n = 1157 MI cases and n = 1748 controls from a population-based study population [German MI Family Study (GerMIFS) III (KORA)] with genome-wide SNP arrays. At this first stage, n = 462 SNPs showed association with MI at P<1 × 10(-3) in two-sided logistic regression. In a second stage, 415 of these SNPs were evaluated in silico in two independent GWA samples, the GerMIFS I (875 cases/1644 controls) and GerMIFS II (1222 cases/1298 controls). Nine SNPs, representing three regions, displayed consistent replication in this in silico analysis (P<0.05 for each GWA sample): five SNPs at 9p21.3, a well-known CAD/MI locus, two SNPs at 10p11.21, and two SNPs at 2p24.3. Wet-lab replication, i.e. the third stage, of SNP rs3739998 (representing the novel locus at 10p11.21, p.S1002T in the KIAA1462 gene) in additional 5790 cases and 5302 controls confirmed the association (P=9.54 × 10(-4)), but not for the 2p24.3 locus. The combined P-value across all stages for SNP rs3739998 is P=1.27 × 10(-11) [odds ratio (OR) = 1.15 (1.11-1.20)]. CONCLUSION: Analysis of a GWA study followed by in silico and wet-lab replication steps identified the KIAA1462 gene, encoding a yet uncharacterized protein, on chromosome 10p11.23 with genome-wide significant association for CAD/MI. Further studies are needed to characterize the functional role of this locus in the aetiology of these diseases.


Asunto(s)
Cromosomas Humanos Par 10/genética , Enfermedad de la Arteria Coronaria/genética , Infarto del Miocardio/genética , Polimorfismo de Nucleótido Simple/genética , Adulto , Moléculas de Adhesión Celular/genética , Cromosomas Humanos Par 9/genética , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Técnicas de Genotipaje/métodos , Humanos , Masculino , Persona de Mediana Edad , Linaje , Factores de Riesgo , Adulto Joven
12.
Cardiovasc Res ; 118(4): 1088-1102, 2022 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33878186

RESUMEN

AIMS: Coronary artery disease (CAD) has a strong genetic predisposition. However, despite substantial discoveries made by genome-wide association studies (GWAS), a large proportion of heritability awaits identification. Non-additive genetic effects might be responsible for part of the unaccounted genetic variance. Here, we attempted a proof-of-concept study to identify non-additive genetic effects, namely epistatic interactions, associated with CAD. METHODS AND RESULTS: We tested for epistatic interactions in 10 CAD case-control studies and UK Biobank with focus on 8068 SNPs at 56 loci with known associations with CAD risk. We identified a SNP pair located in cis at the LPA locus, rs1800769 and rs9458001, to be jointly associated with risk for CAD [odds ratio (OR) = 1.37, P = 1.07 × 10-11], peripheral arterial disease (OR = 1.22, P = 2.32 × 10-4), aortic stenosis (OR = 1.47, P = 6.95 × 10-7), hepatic lipoprotein(a) (Lp(a)) transcript levels (beta = 0.39, P = 1.41 × 10-8), and Lp(a) serum levels (beta = 0.58, P = 8.7 × 10-32), while individual SNPs displayed no association. Further exploration of the LPA locus revealed a strong dependency of these associations on a rare variant, rs140570886, that was previously associated with Lp(a) levels. We confirmed increased CAD risk for heterozygous (relative OR = 1.46, P = 9.97 × 10-32) and individuals homozygous for the minor allele (relative OR = 1.77, P = 0.09) of rs140570886. Using forward model selection, we also show that epistatic interactions between rs140570886, rs9458001, and rs1800769 modulate the effects of the rs140570886 risk allele. CONCLUSIONS: These results demonstrate the feasibility of a large-scale knowledge-based epistasis scan and provide rare evidence of an epistatic interaction in a complex human disease. We were directed to a variant (rs140570886) influencing risk through additive genetic as well as epistatic effects. In summary, this study provides deeper insights into the genetic architecture of a locus important for cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares , Enfermedad de la Arteria Coronaria , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/genética , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Enfermedad de la Arteria Coronaria/genética , Epistasis Genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Lipoproteína(a)/genética , Polimorfismo de Nucleótido Simple
13.
J Clin Endocrinol Metab ; 107(7): e2952-e2961, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35306566

RESUMEN

BACKGROUND: Accelerated reproductive aging, in women indicated by early natural menopause, is associated with increased coronary heart disease (CHD) risk in observational studies. Conversely, an adverse CHD risk profile has been suggested to accelerate menopause. OBJECTIVES: To study the direction and evidence for causality of the relationship between reproductive aging and (non-)fatal CHD and CHD risk factors in a bidirectional Mendelian randomization (MR) approach, using age at natural menopause (ANM) genetic variants as a measure for genetically determined reproductive aging in women. We also studied the association of these variants with CHD risk (factors) in men. DESIGN: Two-sample MR, using both cohort data as well as summary statistics, with 4 methods: simple and weighted median-based, standard inverse-variance weighted (IVW) regression, and MR-Egger regression. PARTICIPANTS: Data from EPIC-CVD and summary statistics from UK Biobank and publicly available genome-wide association studies were pooled for the different analyses. MAIN OUTCOME MEASURES: CHD, CHD risk factors, and ANM. RESULTS: Across different methods of MR, no association was found between genetically determined reproductive aging and CHD risk in women (relative risk estimateIVW = 0.99; 95% confidence interval (CI), 0.97-1.01), or any of the CHD risk factors. Similarly, no associations were found in men. Neither did the reversed analyses show evidence for an association between CHD (risk factors) and reproductive aging. CONCLUSION: Genetically determined reproductive aging is not causally associated with CHD risk (factors) in women, nor were the genetic variants associated in men. We found no evidence for a reverse association in a combined sample of women and men.


Asunto(s)
Enfermedad Coronaria , Estudio de Asociación del Genoma Completo , Envejecimiento/genética , Enfermedad Coronaria/epidemiología , Enfermedad Coronaria/genética , Femenino , Estudio de Asociación del Genoma Completo/métodos , Humanos , Masculino , Análisis de la Aleatorización Mendeliana , Polimorfismo de Nucleótido Simple
14.
Front Psychiatry ; 9: 338, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30100883

RESUMEN

Background: Psychological problems are common in patients with coronary artery disease (CAD) and are associated with poor outcome. However, data on the prevalence of distinct mental disorders and their relevance to patients' functioning in daily life are scarce. Method: In this retrospective study, a total of 514 German patients with CAD as diagnosed by cardiac catheterization were assessed using the Mini International Neuropsychiatric Interview 5.0.0 (M.I.N.I.) and psychosocial functioning was evaluated using the Global Assessment of Functioning (GAF) scale. Results: Twenty-nine percent of the participants suffered from at least one mental disorder after the onset of their CAD (mean time since onset = 10.86 years, SD = 8.15). In comparison to the period before onset of CAD, elevated prevalence rates were found for severe depressive episodes, agoraphobia, dysthymia, panic disorder, and hypochondria. Predictors of mental disorders after the onset of CAD were female gender, younger age at onset of CAD as well as mental disorders and low GAF scores before onset. GAF scores decreased after the onset of CAD, recovered only partially, and were influenced by mental disorders before onset in women but not in men. Conclusions: Mental disorders-especially depression and agoraphobia-are frequent in patients with CAD, with women, patients with a younger age at onset of CAD and patients with any history of mental disorders especially at risk. Regardless of whether patients meet any specific diagnostic criteria, psychosocial functioning is markedly impaired after the onset of CAD, underscoring the need for specific mental health programs for this patient population. Future research, ideally using a prospective design, is necessary to confirm these findings and to further the knowledge of prevalence rates of mental disorders and of modifiable risk factors for the development of mental disorders in patients with CAD.

15.
Front Psychiatry ; 9: 75, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29593584

RESUMEN

BACKGROUND: Comorbid mental disorders in patients with coronary artery disease (CAD) are common and associated with adverse somatic outcomes. However, data on utilization rates of mental health care and treatment efficiency are scarce and inconsistent, which we tried to remedy with the present preliminary study on Northern German CAD patients. METHOD: A total of 514 German CAD patients, as diagnosed by cardiac catheterization, were assessed using the Mini International Neuropsychiatric Interview and the Global Assessment of Functioning (GAF) scale. RESULTS: Global utilization of mental health care since onset of CAD was 21.0%. Depressive disorders, younger age, and lower GAF at onset of CAD were associated with higher utilization rates, while anxiety disorders and gender were not. Lower GAF at onset of CAD, female gender, and psychotherapy was positively associated with higher gains in GAF, while younger age and anxiety disorders were negatively associated. CONCLUSION: The majority of CAD patients with comorbid depression reported to have received mental health treatment and seemed to have benefited from it. However, we found preliminary evidence of insufficiencies in the diagnosis and treatment of anxiety disorders in CAD patients. Further studies, preferably prospective and with representative samples, are needed to corroborate or falsify these findings and explore possible further mediators of health-care utilization by CAD patients such as race, ethnicity, and socioeconomic status.

16.
Biomolecules ; 8(4)2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30563176

RESUMEN

Despite its substantial clinical importance, specific genetic variants associated with depression have not yet been identified. We sought to identify genetic variants associated with depression by (a) focusing on a more homogenous subsample (vascular depression) and (b) applying a three-stage approach. First, we contacted 730 participants with a confirmed atherosclerotic disease (coronary artery disease) from a population-based study population (German Myocardial Infarction Family Study IV) for psychiatric assessment with the Mini International Neuropsychiatric Interview. Second, we genotyped these patients using genome-wide single nucleotide polymorphism (SNP) arrays. Third, we characterized the SNP via in-silico analysis. The final sample consisted of 342 patients (78.3% male, age = 63.2 ± 9.9 years), 22.8% with a severe depressive disorder. Variant rs528732638 on chromosome 18q11.2 was a genome-wide significant variant and was associated with 3.6-fold increase in the odds of lifetime depression. The locus belongs to a linkage disequilibrium block showing expression quantitative trait loci effects on three putative cis-regulated genes, including the aquaporin 4 (AQP4) locus. AQP4 is already known to mediate the formation of ischemic edema in the brain and heart, increasing the size and extent of resulting lesions. Our findings indicate that AQP4 may also play a role in the etiopathology of vascular depression.


Asunto(s)
Acuaporina 4/genética , Depresión/genética , Estudios de Asociación Genética , Enfermedades Vasculares/genética , Depresión/fisiopatología , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo/genética , Enfermedades Vasculares/fisiopatología
17.
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
18.
J Rheumatol ; 44(1): 4-10, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27744395

RESUMEN

OBJECTIVE: Inflammatory diseases, specifically rheumatoid arthritis (RA), are assumed to increase the risk of coronary artery disease (CAD). More recently, multiple single-nucleotide polymorphisms (SNP) associated with RA risk were identified. If causal mechanisms affecting risks of RA and CAD are overlapping, risk alleles for RA might also increase the risk of CAD. METHODS: Sixty-one SNP associating with RA in genome-wide significant analyses were tested for association with CAD in CARDIoGRAM (Coronary ARtery DIsease Genome wide Replication and Meta-analysis), a metaanalysis including genome-wide association data (22,233 CAD cases, 64,762 controls). In parallel, a set of SNP being associated with low-density lipoprotein cholesterol (LDL-C) was tested as a positive control. RESULTS: Twenty-nine RA-associated SNP displayed a directionality-consistent association with CAD (OR range 1.002-1.073), whereas 32 RA-associated SNP were not associated with CAD (OR range 0.96-0.99 per RA risk-increasing allele). The proportion (48%) of directionality-consistent associated SNP equaled the proportion expected by chance (50%, p = 0.09). Of only 5 RA-associated SNP showing p values for CAD < 0.05, 4 loci (C5orf30, IL-6R, PTPN22, and RAD51B) showed directionality-consistent effects on CAD, and 1 (rs10774624, locus SH2B3) reached study-wide significance (p = 7.29E-06). By contrast, and as a proof of concept, 46 (74%) out of 62 LDL-C-associated SNP displayed a directionality-consistent association with CAD, a proportion that was significantly different from 50% (p = 5.9E-05). CONCLUSION: We found no evidence that RA-associated SNP as a group are associated with CAD. Even though we were not able to study potential effects of all genetic variants individually, shared nongenetic factors may more plausibly explain the observed coincidence of the 2 conditions.


Asunto(s)
Artritis Reumatoide/genética , Enfermedad de la Arteria Coronaria/genética , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , Alelos , LDL-Colesterol/genética , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo , Genotipo , Humanos
19.
PLoS One ; 12(8): e0182999, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28829817

RESUMEN

Glatiramer acetate is used therapeutically in multiple sclerosis but also known for adverse effects including elevated coronary artery disease (CAD) risk. The mechanisms underlying the cardiovascular side effects of the medication are unclear. Here, we made use of the chromosomal variation in the genes that are known to be affected by glatiramer treatment. Focusing on genes and gene products reported by drug-gene interaction database to interact with glatiramer acetate we explored a large meta-analysis on CAD genome-wide association studies aiming firstly, to investigate whether variants in these genes also affect cardiovascular risk and secondly, to identify new CAD risk genes. We traced association signals in a 200-kb region around genomic positions of genes interacting with glatiramer in up to 60 801 CAD cases and 123 504 controls. We validated the identified association in additional 21 934 CAD cases and 76 087 controls. We identified three new CAD risk alleles within the TGFB1 region on chromosome 19 that independently affect CAD risk. The lead SNP rs12459996 was genome-wide significantly associated with CAD in the extended meta-analysis (odds ratio 1.09, p = 1.58×10-12). The other two SNPs at the locus were not in linkage disequilibrium with the lead SNP and by a conditional analysis showed p-values of 4.05 × 10-10 and 2.21 × 10-6. Thus, studying genes reported to interact with glatiramer acetate we identified genetic variants that concordantly with the drug increase the risk of CAD. Of these, TGFB1 displayed signal for association. Indeed, the gene has been associated with CAD previously in both in vivo and in vitro studies. Here we establish genome-wide significant association with CAD in large human samples.


Asunto(s)
Enfermedades Cardiovasculares/genética , Predisposición Genética a la Enfermedad , Acetato de Glatiramer/efectos adversos , Enfermedades Cardiovasculares/inducido químicamente , Estudios de Casos y Controles , Humanos , Desequilibrio de Ligamiento , Polimorfismo de Nucleótido Simple
20.
Sci Rep ; 7(1): 10252, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28860667

RESUMEN

Cyclooxygenase-2 inhibitors (coxibs) are characterized by multiple molecular off-target effects and increased coronary artery disease (CAD) risk. Here, we systematically explored common variants of genes representing molecular targets of coxibs for association with CAD. Given a broad spectrum of pleiotropic effects of coxibs, our intention was to narrow potential mechanisms affecting CAD risk as we hypothesized that the affected genes may also display genomic signals of coronary disease risk. A Drug Gene Interaction Database search identified 47 gene products to be affected by coxibs. We traced association signals in 200-kb regions surrounding these genes in 84,813 CAD cases and 202,543 controls. Based on a threshold of 1 × 10-5 (Bonferroni correction for 3131 haplotype blocks), four gene loci yielded significant associations. The lead SNPs were rs7270354 (MMP9), rs4888383 (BCAR1), rs6905288 (VEGFA1), and rs556321 (CACNA1E). By additional genotyping, rs7270354 at MMP9 and rs4888383 at BCAR1 also reached the established GWAS threshold for genome-wide significance. The findings demonstrate overlap of genes affected by coxibs and those mediating CAD risk and points to further mechanisms, which are potentially responsible for coxib-associated CAD risk. The novel approach furthermore suggests that genetic studies may be useful to explore the clinical relevance of off-target drug effects.


Asunto(s)
Cardiotoxicidad/etiología , Inhibidores de la Ciclooxigenasa 2/efectos adversos , Farmacogenética , Mapeo Cromosómico , Biología Computacional/métodos , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Bases de Datos Genéticas , Bases de Datos Farmacéuticas , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Farmacogenética/métodos , Polimorfismo de Nucleótido Simple
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA