Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 14(2): 106-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23334822

RESUMEN

Therapies that target molecular pathways do not provide uniform benefits for all patients at present. New transformative therapies for autoimmune and inflammatory diseases require greater molecular understanding of patient subsets and the ability to personalize targeted therapies for each subset.


Asunto(s)
Enfermedades Autoinmunes/terapia , Inflamación/terapia , Terapia Molecular Dirigida , Neoplasias/terapia , Medicina de Precisión , Antineoplásicos/economía , Antineoplásicos/uso terapéutico , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Heterogeneidad Genética , Humanos , Inmunosupresores/economía , Inmunosupresores/uso terapéutico , Inflamación/genética , Inflamación/inmunología , Neoplasias/genética , Neoplasias/inmunología
2.
J Allergy Clin Immunol ; 150(4): 972-978.e7, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35487308

RESUMEN

BACKGROUND: Clinical studies of type 2 (T2) cytokine-related neutralizing antibodies in asthma have identified a substantial subset of patients with low levels of T2 inflammation who do not benefit from T2 cytokine neutralizing antibody treatment. Non-T2 mechanisms are poorly understood in asthma but represent a redefined unmet medical need. OBJECTIVE: We sought to gain a better understanding of genetic contributions to T2-low asthma. METHODS: We utilized an unbiased genome-wide association study of patients with moderate to severe asthma stratified by T2 serum biomarker periostin. We also performed additional expression and biological analysis for the top genetic hits. RESULTS: We identified a novel protective single nucleotide polymorphism at chr19q13.41, which is selectively associated with T2-low asthma and establishes Kallikrein-related peptidase 5 (KLK5) as the causal gene mediating this association. Heterozygous carriers of the single nucleotide polymorphisms have reduced KLK5 expression. KLK5 is secreted by human bronchial epithelial cells and elevated in asthma bronchial alveolar lavage. T2 cytokines IL-4 and IL-13 downregulate KLK5 in human bronchial epithelial cells. KLK5, dependent on its catalytic function, induces epithelial chemokine/cytokine expression. Finally, overexpression of KLK5 in airway or lack of an endogenous KLK5 inhibitor, SPINK5, leads to spontaneous airway neutrophilic inflammation. CONCLUSION: Our data identify KLK5 to be the causal gene at a novel locus at chr19q13.41 associated with T2-low asthma.


Asunto(s)
Asma , Estudio de Asociación del Genoma Completo , Anticuerpos Neutralizantes/genética , Asma/genética , Quimiocinas/genética , Citocinas/metabolismo , Humanos , Inflamación/genética , Interleucina-13/genética , Interleucina-4/genética , Calicreínas/genética , Calicreínas/metabolismo
3.
J Immunol ; 202(1): 183-193, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30510070

RESUMEN

Both common and rare genetic variants of laccase domain-containing 1 (LACC1, previously C13orf31) are associated with inflammatory bowel disease, leprosy, Behcet disease, and systemic juvenile idiopathic arthritis. However, the functional relevance of these variants is unclear. In this study, we use LACC1-deficient mice to gain insight into the role of LACC1 in regulating inflammation. Following oral administration of Citrobacter rodentium, LACC1 knockout (KO) mice had more severe colon lesions compared with wildtype (WT) controls. Immunization with collagen II, a collagen-induced arthritis (CIA) model, resulted in an accelerated onset of arthritis and significantly worse arthritis and inflammation in LACC1 KO mice. Similar results were obtained in a mannan-induced arthritis model. Serum and local TNF in CIA paws and C. rodentium colons were significantly increased in LACC1 KO mice compared with WT controls. The percentage of IL-17A-producing CD4+ T cells was elevated in LACC1 KO mice undergoing CIA as well as aged mice compared with WT controls. Neutralization of IL-17, but not TNF, prevented enhanced mannan-induced arthritis in LACC1 KO mice. These data provide new mechanistic insight into the function of LACC1 in regulating TNF and IL-17 during inflammatory responses. We hypothesize that these effects contribute to immune-driven pathologies observed in individuals carrying LACC1 variants.


Asunto(s)
Artritis Experimental/inmunología , Artritis Juvenil/inmunología , Citrobacter rodentium/fisiología , Infecciones por Enterobacteriaceae/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Oxidorreductasas/metabolismo , Células Th17/inmunología , Alelos , Animales , Artritis Experimental/microbiología , Artritis Juvenil/genética , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/genética , Interleucina-17/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidorreductasas/genética , Polimorfismo Genético , Factores de Necrosis Tumoral/metabolismo
4.
PLoS Genet ; 14(11): e1007427, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30388101

RESUMEN

Paired Immunoglobulin-like Type 2 Receptor Alpha (PILRA) is a cell surface inhibitory receptor that recognizes specific O-glycosylated proteins and is expressed on various innate immune cell types including microglia. We show here that a common missense variant (G78R, rs1859788) of PILRA is the likely causal allele for the confirmed Alzheimer's disease risk locus at 7q21 (rs1476679). The G78R variant alters the interaction of residues essential for sialic acid engagement, resulting in >50% reduced binding for several PILRA ligands including a novel ligand, complement component 4A, and herpes simplex virus 1 (HSV-1) glycoprotein B. PILRA is an entry receptor for HSV-1 via glycoprotein B, and macrophages derived from R78 homozygous donors showed significantly decreased levels of HSV-1 infection at several multiplicities of infection compared to homozygous G78 macrophages. We propose that PILRA G78R protects individuals from Alzheimer's disease risk via reduced inhibitory signaling in microglia and reduced microglial infection during HSV-1 recurrence.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Variación Genética , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Sustitución de Aminoácidos , Animales , Sitios Genéticos , Humanos , Ligandos , Glicoproteínas de Membrana/química , Ratones , Modelos Biológicos , Conformación Molecular , Unión Proteica , Sitios de Carácter Cuantitativo , Receptores Inmunológicos/química , Relación Estructura-Actividad
5.
Genes Immun ; 20(2): 172-179, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29550837

RESUMEN

In clinical trials, a placebo response refers to improvement in disease symptoms arising from the psychological effect of receiving a treatment rather than the actual treatment under investigation. Previous research has reported genomic variation associated with the likelihood of observing a placebo response, but these studies have been limited in scope and have not been validated. Here, we analyzed whole-genome sequencing data from 784 patients undergoing placebo treatment in Phase III Asthma or Rheumatoid Arthritis trials to assess the impact of previously reported variation on patient outcomes in the placebo arms and to identify novel variants associated with the placebo response. Contrary to expectations based on previous reports, we did not observe any statistically significant associations between genomic variants and placebo treatment outcome. Our findings suggest that the biological origin of the placebo response is complex and likely to be variable between disease areas.


Asunto(s)
Ensayos Clínicos Fase III como Asunto/normas , Efecto Placebo , Polimorfismo de Nucleótido Simple , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/genética , Asma/tratamiento farmacológico , Asma/genética , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Persona de Mediana Edad
6.
J Immunol ; 199(4): 1238-1249, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28696253

RESUMEN

Ox40 ligand (Ox40L) locus genetic variants are associated with the risk for systemic lupus erythematosus (SLE); however, it is unclear how Ox40L contributes to SLE pathogenesis. In this study, we evaluated the contribution of Ox40L and its cognate receptor, Ox40, using in vivo agonist and antagonist approaches in the NZB × NZW (NZB/W) F1 mouse model of SLE. Ox40 was highly expressed on several CD4 Th cell subsets in the spleen and kidney of diseased mice, and expression correlated with disease severity. Treatment of aged NZB/W F1 mice with agonist anti-Ox40 mAbs potently exacerbated renal disease, which was accompanied by activation of kidney-infiltrating T cells and cytokine production. The agonist mAbs also induced activation and inflammatory gene expression in splenic CD4 T cells, including IFN-regulated genes, increased the number of follicular helper T cells and plasmablasts in the spleen, and led to elevated levels of serum IgM and enhanced renal glomerular IgM deposition. In a type I IFN-accelerated lupus model, treatment with an antagonist Ox40:Fc fusion protein significantly delayed the onset of severe proteinuria and improved survival. These data support the hypothesis that the Ox40/Ox40L pathway drives cellular and humoral autoimmune responses during lupus nephritis in NZB/W F1 mice and emphasize the potential clinical value of targeting this pathway in human lupus.


Asunto(s)
Nefritis Lúpica/inmunología , Glicoproteínas de Membrana/metabolismo , Células Plasmáticas/inmunología , Receptores OX40/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Factores de Necrosis Tumoral/metabolismo , Animales , Anticuerpos Antinucleares/inmunología , Anticuerpos Monoclonales/inmunología , Autoinmunidad , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Riñón/inmunología , Riñón/patología , Glomérulos Renales/inmunología , Glomérulos Renales/patología , Nefritis Lúpica/fisiopatología , Ratones , Ratones Endogámicos NZB , Ligando OX40 , Proteinuria/inmunología , Transducción de Señal , Linfocitos T Colaboradores-Inductores/metabolismo
7.
Bioinformatics ; 33(4): 599-600, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28035029

RESUMEN

Motivation: We have developed geneAttribution, an R package that assigns candidate causal gene(s) to a risk variant identified by a genetic association study such as a GWAS. The method combines user-supplied functional annotation such as expression quantitative trait loci (eQTL) or Hi-C genome conformation data and reports the most likely candidate genes. In the absence of annotation data, geneAttribution relies on the distances between the genes and the input variant. Availability and Implementation: The package is freely available from http://www.bioconductor.org/ . A quick-start vignette is included with the package. Contact: wustera@gene.com.


Asunto(s)
Estudios de Asociación Genética/métodos , Polimorfismo Genético , Regiones Promotoras Genéticas , Programas Informáticos , Genoma Humano , Humanos , Fenotipo , Sitios de Carácter Cuantitativo
8.
Nature ; 488(7409): 96-9, 2012 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-22801501

RESUMEN

The prevalence of dementia in the Western world in people over the age of 60 has been estimated to be greater than 5%, about two-thirds of which are due to Alzheimer's disease. The age-specific prevalence of Alzheimer's disease nearly doubles every 5 years after age 65, leading to a prevalence of greater than 25% in those over the age of 90 (ref. 3). Here, to search for low-frequency variants in the amyloid-ß precursor protein (APP) gene with a significant effect on the risk of Alzheimer's disease, we studied coding variants in APP in a set of whole-genome sequence data from 1,795 Icelanders. We found a coding mutation (A673T) in the APP gene that protects against Alzheimer's disease and cognitive decline in the elderly without Alzheimer's disease. This substitution is adjacent to the aspartyl protease ß-site in APP, and results in an approximately 40% reduction in the formation of amyloidogenic peptides in vitro. The strong protective effect of the A673T substitution against Alzheimer's disease provides proof of principle for the hypothesis that reducing the ß-cleavage of APP may protect against the disease. Furthermore, as the A673T allele also protects against cognitive decline in the elderly without Alzheimer's disease, the two may be mediated through the same or similar mechanisms.


Asunto(s)
Envejecimiento/genética , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Mutación/genética , Alelos , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/prevención & control , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/química , Ácido Aspártico Endopeptidasas/metabolismo , Cognición/fisiología , Trastornos del Conocimiento/prevención & control , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Placa Amiloide/genética , Placa Amiloide/metabolismo
9.
BMC Bioinformatics ; 18(1): 351, 2017 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-28738841

RESUMEN

BACKGROUND: Large sample sets of whole genome sequencing with deep coverage are being generated, however assembling datasets from different sources inevitably introduces batch effects. These batch effects are not well understood and can be due to changes in the sequencing protocol or bioinformatics tools used to process the data. No systematic algorithms or heuristics exist to detect and filter batch effects or remove associations impacted by batch effects in whole genome sequencing data. RESULTS: We describe key quality metrics, provide a freely available software package to compute them, and demonstrate that identification of batch effects is aided by principal components analysis of these metrics. To mitigate batch effects, we developed new site-specific filters that identified and removed variants that falsely associated with the phenotype due to batch effect. These include filtering based on: a haplotype based genotype correction, a differential genotype quality test, and removing sites with missing genotype rate greater than 30% after setting genotypes with quality scores less than 20 to missing. This method removed 96.1% of unconfirmed genome-wide significant SNP associations and 97.6% of unconfirmed genome-wide significant indel associations. We performed analyses to demonstrate that: 1) These filters impacted variants known to be disease associated as 2 out of 16 confirmed associations in an AMD candidate SNP analysis were filtered, representing a reduction in power of 12.5%, 2) In the absence of batch effects, these filters removed only a small proportion of variants across the genome (type I error rate of 3%), and 3) in an independent dataset, the method removed 90.2% of unconfirmed genome-wide SNP associations and 89.8% of unconfirmed genome-wide indel associations. CONCLUSIONS: Researchers currently do not have effective tools to identify and mitigate batch effects in whole genome sequencing data. We developed and validated methods and filters to address this deficiency.


Asunto(s)
Estudio de Asociación del Genoma Completo/métodos , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Degeneración Macular/genética , Degeneración Macular/patología , Fenotipo , Polimorfismo de Nucleótido Simple , Análisis de Componente Principal , Análisis de Secuencia de ADN , Programas Informáticos
10.
Am J Hum Genet ; 94(4): 586-98, 2014 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-24702955

RESUMEN

Efforts to identify lupus-associated causal variants in the FAM167A/BLK locus on 8p21 are hampered by highly associated noncausal variants. In this report, we used a trans-population mapping and sequencing strategy to identify a common variant (rs922483) in the proximal BLK promoter and a tri-allelic variant (rs1382568) in the upstream alternative BLK promoter as putative causal variants for association with systemic lupus erythematosus. The risk allele (T) at rs922483 reduced proximal promoter activity and modulated alternative promoter usage. Allelic differences at rs1382568 resulted in altered promoter activity in B progenitor cell lines. Thus, our results demonstrated that both lupus-associated functional variants contribute to the autoimmune disease association by modulating transcription of BLK in B cells and thus potentially altering immune responses.


Asunto(s)
Lupus Eritematoso Sistémico/genética , Regiones Promotoras Genéticas , Transcripción Genética , Familia-src Quinasas/genética , Alelos , Cromosomas Humanos Par 8 , Ensayo de Cambio de Movilidad Electroforética , Femenino , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Masculino , Polimorfismo de Nucleótido Simple
11.
Nat Genet ; 40(9): 1062-4, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19165919

RESUMEN

The TNFAIP3 (tumor necrosis factor alpha-induced protein 3) gene encodes a ubiquitin editing enzyme, A20, that restricts NF-kappaB-dependent signaling and prevents inflammation. We show that three independent SNPs in the TNFAIP3 region (rs13192841, rs2230926 and rs6922466) are associated with systemic lupus erythematosus (SLE) among individuals of European ancestry. These findings provide critical links between A20 and the etiology of SLE.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Lupus Eritematoso Sistémico/genética , Proteínas Nucleares/genética , Polimorfismo de Nucleótido Simple , Proteínas de Unión al ADN , Predisposición Genética a la Enfermedad , Humanos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Población Blanca/genética
12.
Nat Genet ; 40(9): 1059-61, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19165918

RESUMEN

Systemic lupus erythematosus (SLE) is an autoimmune disease influenced by genetic and environmental factors. We carried out a genome-wide association scan and replication study and found an association between SLE and a variant in TNFAIP3 (rs5029939, meta-analysis P = 2.89 x 10(-12), OR = 2.29). We also found evidence of two independent signals near TNFAIP3 associated with SLE, including one previously associated with rheumatoid arthritis (RA). These results establish that variants near TNFAIP3 contribute to differential risk of SLE and RA.


Asunto(s)
Cromosomas Humanos Par 6 , Péptidos y Proteínas de Señalización Intracelular/genética , Lupus Eritematoso Sistémico/genética , Proteínas Nucleares/genética , Artritis Reumatoide/genética , Proteínas de Unión al ADN , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Polimorfismo de Nucleótido Simple , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
13.
Nature ; 466(7303): 243-7, 2010 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-20555325

RESUMEN

Sialic acid acetylesterase (SIAE) is an enzyme that negatively regulates B lymphocyte antigen receptor signalling and is required for the maintenance of immunological tolerance in mice. Heterozygous loss-of-function germline rare variants and a homozygous defective polymorphic variant of SIAE were identified in 24/923 subjects of European origin with relatively common autoimmune disorders and in 2/648 controls of European origin. All heterozygous loss-of-function SIAE mutations tested were capable of functioning in a dominant negative manner. A homozygous secretion-defective polymorphic variant of SIAE was catalytically active, lacked the ability to function in a dominant negative manner, and was seen in eight autoimmune subjects but in no control subjects. The odds ratio for inheriting defective SIAE alleles was 8.6 in all autoimmune subjects, 8.3 in subjects with rheumatoid arthritis, and 7.9 in subjects with type I diabetes. Functionally defective SIAE rare and polymorphic variants represent a strong genetic link to susceptibility in relatively common human autoimmune disorders.


Asunto(s)
Acetilesterasa/genética , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/genética , Autoinmunidad/genética , Hidrolasas de Éster Carboxílico/genética , Predisposición Genética a la Enfermedad/genética , Mutación de Línea Germinal/genética , Ácido N-Acetilneuramínico/metabolismo , Acetilación , Acetilesterasa/metabolismo , Alelos , Animales , Anticuerpos Antinucleares/sangre , Artritis Reumatoide/enzimología , Artritis Reumatoide/genética , Linfocitos B/metabolismo , Biocatálisis , Hidrolasas de Éster Carboxílico/metabolismo , Estudios de Casos y Controles , Línea Celular , Diabetes Mellitus Tipo 1/enzimología , Diabetes Mellitus Tipo 1/genética , Europa (Continente)/etnología , Exones/genética , Humanos , Ratones , Oportunidad Relativa , Polimorfismo de Nucleótido Simple/genética , Tamaño de la Muestra , Análisis de Secuencia de ADN
14.
Am J Hum Genet ; 91(5): 778-93, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-23084292

RESUMEN

We have performed a meta-analysis of the major-histocompatibility-complex (MHC) region in systemic lupus erythematosus (SLE) to determine the association with both SNPs and classical human-leukocyte-antigen (HLA) alleles. More specifically, we combined results from six studies and well-known out-of-study control data sets, providing us with 3,701 independent SLE cases and 12,110 independent controls of European ancestry. This study used genotypes for 7,199 SNPs within the MHC region and for classical HLA alleles (typed and imputed). Our results from conditional analysis and model choice with the use of the Bayesian information criterion show that the best model for SLE association includes both classical loci (HLA-DRB1(∗)03:01, HLA-DRB1(∗)08:01, and HLA-DQA1(∗)01:02) and two SNPs, rs8192591 (in class III and upstream of NOTCH4) and rs2246618 (MICB in class I). Our approach was to perform a stepwise search from multiple baseline models deduced from a priori evidence on HLA-DRB1 lupus-associated alleles, a stepwise regression on SNPs alone, and a stepwise regression on HLA alleles. With this approach, we were able to identify a model that was an overwhelmingly better fit to the data than one identified by simple stepwise regression either on SNPs alone (Bayes factor [BF] > 50) or on classical HLA alleles alone (BF > 1,000).


Asunto(s)
Alelos , Antígenos HLA/genética , Lupus Eritematoso Sistémico/genética , Complejo Mayor de Histocompatibilidad/genética , Población Blanca/genética , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Genotipo , Antígenos HLA-B/genética , Antígenos HLA-C/genética , Cadenas alfa de HLA-DQ/genética , Cadenas HLA-DRB1/genética , Haplotipos , Humanos , Polimorfismo de Nucleótido Simple
15.
PLoS Genet ; 8(1): e1002476, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22291608

RESUMEN

Selective IgA deficiency (IgAD; serum IgA<0.07 g/l) is the most common form of human primary immune deficiency, affecting approximately 1∶600 individuals in populations of Northern European ancestry. The polygenic nature of IgAD is underscored by the recent identification of several new risk genes in a genome-wide association study. Among the characterized susceptibility loci, the association with specific HLA haplotypes represents the major genetic risk factor for IgAD. Despite the robust association, the nature and location of the causal variants in the HLA region remains unknown. To better characterize the association signal in this region, we performed a high-density SNP mapping of the HLA locus and imputed the genotypes of common HLA-B, -DRB1, and -DQB1 alleles in a combined sample of 772 IgAD patients and 1,976 matched controls from 3 independent European populations. We confirmed the complex nature of the association with the HLA locus, which is the result of multiple effects spanning the entire HLA region. The primary association signal mapped to the HLA-DQB1*02 allele in the HLA Class II region (combined P = 7.69×10(-57); OR = 2.80) resulting from the combined independent effects of the HLA-B*0801-DRB1*0301-DQB1*02 and -DRB1*0701-DQB1*02 haplotypes, while additional secondary signals were associated with the DRB1*0102 (combined P = 5.86×10(-17); OR = 4.28) and the DRB1*1501 (combined P = 2.24×10(-35); OR = 0.13) alleles. Despite the strong population-specific frequencies of HLA alleles, we found a remarkable conservation of these effects regardless of the ethnic background, which supports the use of large multi-ethnic populations to characterize shared genetic association signals in the HLA region. We also provide evidence for the location of association signals within the specific extended haplotypes, which will guide future sequencing studies aimed at characterizing the precise functional variants contributing to disease pathogenesis.


Asunto(s)
Antígenos HLA-B/genética , Cadenas beta de HLA-DQ/genética , Cadenas HLA-DRB1/genética , Deficiencia de IgA/genética , Alelos , Estudios de Casos y Controles , Mapeo Cromosómico , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Haplotipos , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Desequilibrio de Ligamiento , Polimorfismo de Nucleótido Simple , Población Blanca/genética
16.
Hum Hered ; 78(2): 94-103, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25096029

RESUMEN

BACKGROUND/AIMS: Genome-wide association (GWA) studies have reported susceptible regions in the human genome for many common diseases and traits; however, these loci only explain a minority of trait heritability. To boost the power of a GWA study, substantial research endeavors have been focused on integrating other available genomic information in the analysis. Advances in high through-put technologies have generated a wealth of genomic data and made combining SNP and gene expression data become feasible. RESULTS: In this paper, we propose a novel procedure to incorporate gene expression information into GWA analysis. This procedure utilizes weights constructed by gene expression measurements to adjust p values from a GWA analysis. RESULTS from simulation analyses indicate that the proposed procedures may achieve substantial power gains, while controlling family-wise type I error rates at the nominal level. To demonstrate the implementation of our proposed approach, we apply the weight adjustment procedure to a GWA study on serum interferon-regulated chemokine levels in systemic lupus erythematosus patients. The study results can provide valuable insights for the functional interpretation of GWA signals. AVAILABILITY: The R source code for implementing the proposed weighting procedure is available at http://www.biostat.umn.edu/∼yho/research.html.


Asunto(s)
Expresión Génica , Estudio de Asociación del Genoma Completo , Lupus Eritematoso Sistémico/genética , Biomarcadores/sangre , Quimiocina CCL19/sangre , Quimiocina CCL2/sangre , Quimiocina CXCL10/sangre , Simulación por Computador , Femenino , Genoma Humano , Humanos , Lupus Eritematoso Sistémico/sangre , Masculino , Polimorfismo de Nucleótido Simple , Población Blanca/genética
17.
J Am Soc Nephrol ; 25(12): 2859-70, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24925725

RESUMEN

Lupus nephritis is a manifestation of SLE resulting from glomerular immune complex deposition and inflammation. Lupus nephritis demonstrates familial aggregation and accounts for significant morbidity and mortality. We completed a meta-analysis of three genome-wide association studies of SLE to identify lupus nephritis-predisposing loci. Through genotyping and imputation, >1.6 million markers were assessed in 2000 unrelated women of European descent with SLE (588 patients with lupus nephritis and 1412 patients with lupus without nephritis). Tests of association were computed using logistic regression adjusting for population substructure. The strongest evidence for association was observed outside the MHC and included markers localized to 4q11-q13 (PDGFRA, GSX2; P=4.5×10(-7)), 16p12 (SLC5A11; P=5.1×10(-7)), 6p22 (ID4; P=7.4×10(-7)), and 8q24.12 (HAS2, SNTB1; P=1.1×10(-6)). Both HLA-DR2 and HLA-DR3, two well established lupus susceptibility loci, showed evidence of association with lupus nephritis (P=0.06 and P=3.7×10(-5), respectively). Within the class I region, rs9263871 (C6orf15-HCG22) had the strongest evidence of association with lupus nephritis independent of HLA-DR2 and HLA-DR3 (P=8.5×10(-6)). Consistent with a functional role in lupus nephritis, intra-renal mRNA levels of PDGFRA and associated pathway members showed significant enrichment in patients with lupus nephritis (n=32) compared with controls (n=15). Results from this large-scale genome-wide investigation of lupus nephritis provide evidence of multiple biologically relevant lupus nephritis susceptibility loci.


Asunto(s)
Predisposición Genética a la Enfermedad , Lupus Eritematoso Sistémico/genética , Nefritis Lúpica/genética , Adolescente , Adulto , Anciano , Niño , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Antígeno HLA-DR2/genética , Antígeno HLA-DR3/genética , Humanos , Lupus Eritematoso Sistémico/epidemiología , Lupus Eritematoso Sistémico/fisiopatología , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple , ARN Mensajero/metabolismo , Población Blanca/genética , Adulto Joven
18.
Nat Genet ; 38(5): 550-5, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16642019

RESUMEN

Systemic lupus erythematosus (SLE) is a complex autoimmune disease characterized by activation of the type I interferon (IFN) pathway. Here we convincingly replicate association of the IFN regulatory factor 5 (IRF5) rs2004640 T allele with SLE in four independent case-control cohorts (P = 4.4 x 10(-16)) and by family-based transmission disequilibrium test analysis (P = 0.0006). The rs2004640 T allele creates a 5' donor splice site in an alternate exon 1 of IRF5, allowing expression of several unique IRF5 isoforms. We also identify an independent cis-acting variant associated with elevated expression of IRF5 and linked to the exon 1B splice site. Haplotypes carrying the variant associated with elevated expression and lacking the exon 1B donor site do not confer risk of SLE. Thus, a common IRF5 haplotype driving elevated expression of multiple unique isoforms of IRF5 is an important genetic risk factor for SLE, establishing a causal role for type I IFN pathway genes in human autoimmunity.


Asunto(s)
Factores Reguladores del Interferón/fisiología , Lupus Eritematoso Sistémico/genética , Empalme del ARN/fisiología , Estudios de Casos y Controles , Estudios de Cohortes , Genotipo , Haplotipos , Humanos , Factores Reguladores del Interferón/genética , Datos de Secuencia Molecular , Polimorfismo Genético
19.
PLoS Genet ; 7(10): e1002341, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22046141

RESUMEN

Systemic lupus erythematosus (SLE) is a complex trait characterised by the production of a range of auto-antibodies and a diverse set of clinical phenotypes. Currently, ~8% of the genetic contribution to SLE in Europeans is known, following publication of several moderate-sized genome-wide (GW) association studies, which identified loci with a strong effect (OR>1.3). In order to identify additional genes contributing to SLE susceptibility, we conducted a replication study in a UK dataset (870 cases, 5,551 controls) of 23 variants that showed moderate-risk for lupus in previous studies. Association analysis in the UK dataset and subsequent meta-analysis with the published data identified five SLE susceptibility genes reaching genome-wide levels of significance (P(comb)<5×10(-8)): NCF2 (P(comb) = 2.87×10(-11)), IKZF1 (P(comb) = 2.33×10(-9)), IRF8 (P(comb) = 1.24×10(-8)), IFIH1 (P(comb) = 1.63×10(-8)), and TYK2 (P(comb) = 3.88×10(-8)). Each of the five new loci identified here can be mapped into interferon signalling pathways, which are known to play a key role in the pathogenesis of SLE. These results increase the number of established susceptibility genes for lupus to ~30 and validate the importance of using large datasets to confirm associations of loci which moderately increase the risk for disease.


Asunto(s)
ARN Helicasas DEAD-box/genética , Factor de Transcripción Ikaros/genética , Factores Reguladores del Interferón/genética , Interferones/metabolismo , Lupus Eritematoso Sistémico/genética , NADPH Oxidasas/genética , TYK2 Quinasa/genética , Estudios de Casos y Controles , Estudios de Cohortes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Helicasa Inducida por Interferón IFIH1 , Interferones/genética , Polimorfismo de Nucleótido Simple , Transducción de Señal
20.
PLoS Genet ; 7(3): e1001323, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21408207

RESUMEN

Systemic lupus erythematosus (SLE) is a clinically heterogeneous, systemic autoimmune disease characterized by autoantibody formation. Previously published genome-wide association studies (GWAS) have investigated SLE as a single phenotype. Therefore, we conducted a GWAS to identify genetic factors associated with anti-dsDNA autoantibody production, a SLE-related autoantibody with diagnostic and clinical importance. Using two independent datasets, over 400,000 single nucleotide polymorphisms (SNPs) were studied in a total of 1,717 SLE cases and 4,813 healthy controls. Anti-dsDNA autoantibody positive (anti-dsDNA +, n = 811) and anti-dsDNA autoantibody negative (anti-dsDNA -, n = 906) SLE cases were compared to healthy controls and to each other to identify SNPs associated specifically with these SLE subtypes. SNPs in the previously identified SLE susceptibility loci STAT4, IRF5, ITGAM, and the major histocompatibility complex were strongly associated with anti-dsDNA + SLE. Far fewer and weaker associations were observed for anti-dsDNA - SLE. For example, rs7574865 in STAT4 had an OR for anti-dsDNA + SLE of 1.77 (95% CI 1.57-1.99, p = 2.0E-20) compared to an OR for anti-dsDNA - SLE of 1.26 (95% CI 1.12-1.41, p = 2.4E-04), with p(heterogeneity)<0.0005. SNPs in the SLE susceptibility loci BANK1, KIAA1542, and UBE2L3 showed evidence of association with anti-dsDNA + SLE and were not associated with anti-dsDNA - SLE. In conclusion, we identified differential genetic associations with SLE based on anti-dsDNA autoantibody production. Many previously identified SLE susceptibility loci may confer disease risk through their role in autoantibody production and be more accurately described as autoantibody propensity loci. Lack of strong SNP associations may suggest that other types of genetic variation or non-genetic factors such as environmental exposures have a greater impact on susceptibility to anti-dsDNA - SLE.


Asunto(s)
Autoanticuerpos , ADN/inmunología , Predisposición Genética a la Enfermedad , Lupus Eritematoso Sistémico , Autoanticuerpos/genética , Autoanticuerpos/inmunología , Antígeno CD11b/genética , Estudios de Casos y Controles , Variación Genética , Estudio de Asociación del Genoma Completo , Humanos , Factores Reguladores del Interferón/genética , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Complejo Mayor de Histocompatibilidad/genética , Polimorfismo de Nucleótido Simple , Factor de Transcripción STAT4/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA