Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Autoimmun ; 68: 23-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26782299

RESUMEN

Genetic susceptibility to type 1 diabetes (T1D) is well supported by epidemiologic evidence; however, disease risk cannot be entirely explained by established genetic variants identified so far. This study addresses the question of whether epigenetic modification of the inherited DNA sequence may contribute to T1D susceptibility. Using the Infinium HumanMethylation450 BeadChip array (450k), a total of seven long-term disease-discordant monozygotic (MZ) twin pairs and five pairs of HLA-identical, disease-discordant non-twin siblings (NTS) were examined for associations between DNA methylation (DNAm) and T1D. Strong evidence for global hypomethylation of CpG sites within promoter regions in MZ twins with TID compared to twins without T1D was observed. DNA methylation data were then grouped into three categories of CpG sites for further analysis, including those within: 1) the major histocompatibility complex (MHC) region, 2) non-MHC genes with reported T1D association through genome wide association studies (GWAS), and 3) the epigenome, or remainder of sites that did not include MHC and T1D associated genes. Initial results showed modest methylation differences between discordant MZ twins for the MHC region and T1D-associated CpG sites, BACH2, INS-IGF2, and CLEC16A (DNAm difference range: 2.2%-5.0%). In the epigenome CpG set, the greatest methylation differences were observed in MAGI2, FANCC, and PCDHB16, (DNAm difference range: 6.9%-16.1%). These findings were not observed in the HLA-identical NTS pairs. Targeted pyrosequencing of five candidate CpG loci identified using the 450k array in the original discordant MZ twins produced similar results using control DNA samples, indicating strong agreement between the two DNA methylation profiling platforms. However, findings for the top five candidate CpG loci were not replicated in six additional T1D-discordant MZ twin pairs. Our results indicate global DNA hypomethylation within gene promoter regions may contribute to T1D; however, findings do not support the involvement of large DNAm differences at single CpG sites alone in T1D.


Asunto(s)
Metilación de ADN , Diabetes Mellitus Tipo 1/genética , Regiones Promotoras Genéticas , Gemelos Monocigóticos , Adolescente , Adulto , Niño , Preescolar , Islas de CpG , Epigénesis Genética , Femenino , Predisposición Genética a la Enfermedad , Humanos , Lactante , Masculino , Especificidad de Órganos/genética , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN , Adulto Joven
2.
Proc Natl Acad Sci U S A ; 110(8): 2952-6, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23382179

RESUMEN

Nuclear localization leucine-rich-repeat protein 1 (NLRP1) is a key regulator of the innate immune system, particularly in the skin where, in response to molecular triggers such as pathogen-associated or damage-associated molecular patterns, the NLRP1 inflammasome promotes caspase-1-dependent processing of bioactive interleukin-1ß (IL-1ß), resulting in IL-1ß secretion and downstream inflammatory responses. NLRP1 is genetically associated with risk of several autoimmune diseases including generalized vitiligo, Addison disease, type 1 diabetes, rheumatoid arthritis, and others. Here we identify a repertoire of variation in NLRP1 by deep DNA resequencing. Predicted functional variations in NLRP1 reside in several common high-risk haplotypes that differ from the reference by multiple nonsynonymous substitutions. The haplotypes that are high risk for disease share two substitutions, L155H and M1184V, and are inherited largely intact due to extensive linkage disequilibrium across the region. Functionally, we found that peripheral blood monocytes from healthy subjects homozygous for the predominant high-risk haplotype 2A processed significantly greater (P < 0.0001) amounts of the IL-1ß precursor to mature bioactive IL-1ß under basal (resting) conditions and in response to Toll-like receptor (TLR) agonists (TLR2 and TLR4) compared with monocytes from subjects homozygous for the reference haplotype 1. The increase in basal release was 1.8-fold greater in haplotype 2A monocytes, and these differences between the two haplotypes were consistently observed three times over a 3-mo period; no differences were observed for IL-1α or TNFα. NLRP1 RNA and protein levels were not altered by the predominant high-risk haplotype, indicating that altered function of the corresponding multivariant NLRP1 polypeptide predisposes to autoimmune diseases by activation of the NLRP1 inflammasome.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Autoinmunidad/genética , Haplotipos , Inflamasomas/inmunología , Interleucina-1beta/metabolismo , Vitíligo/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Humanos , Monocitos/inmunología , Monocitos/metabolismo , Proteínas NLR
3.
Diabetologia ; 58(9): 2079-86, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26070305

RESUMEN

AIMS/HYPOTHESIS: The discordance status of (autoimmune) type 1 diabetes within monozygotic twin pairs points to the importance of environmental factors. The aim of this study was to investigate whether the environmental events causing type 1 diabetes influence thyroid autoimmunity. METHODS: Monozygotic and dizygotic twins discordant for type 1 diabetes from the UK and USA were tested for thyroid peroxidase autoantibodies (TPOA) by radioimmunoassay. Using quantitative genetic model fitting of a liability-threshold model we estimated the contribution of genetic (heritability) and environmental factors to TPOA. RESULTS: TPOA positivity was higher in females than in males in both cohorts and was associated with later age at diagnosis in the UK and combined cohorts (p < 0.01). TPOA did not specifically segregate with type 1 diabetes in the twin pairs (p > 0.2 in all groups). The best-fitting models showed heritability (95% CI) estimates for TPOA of 63% (37%, 80%) for the UK and 80% (51%, 92%) for US twins, while the best-fitting meta-analysis model of the two twin cohorts combined included additive genetic and unique environmental factors with a heritability estimate of 69% (50%, 82%). CONCLUSIONS/INTERPRETATION: Risk of thyroid autoimmunity, defined by TPOA, in the context of autoimmune diabetes is, substantially, genetically determined in discordant twin pairs. Environmental factors leading to type 1 diabetes were not the same as those involved with thyroid autoimmunity. It follows that it is as important to investigate for thyroid autoimmunity in relatives of type 1 diabetes patients as it is in the patients themselves.


Asunto(s)
Autoanticuerpos/sangre , Autoantígenos/sangre , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/genética , Yoduro Peroxidasa/sangre , Proteínas de Unión a Hierro/sangre , Adolescente , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Autoinmunidad , Niño , Estudios de Cohortes , Diabetes Mellitus Tipo 1/inmunología , Ambiente , Femenino , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Humanos , Yoduro Peroxidasa/inmunología , Proteínas de Unión a Hierro/inmunología , Masculino , Radioinmunoensayo , Glándula Tiroides/inmunología , Gemelos Dicigóticos , Gemelos Monocigóticos , Reino Unido , Estados Unidos
4.
N Engl J Med ; 362(18): 1686-97, 2010 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-20410501

RESUMEN

BACKGROUND: Generalized vitiligo is an autoimmune disease characterized by melanocyte loss, which results in patchy depigmentation of skin and hair, and is associated with an elevated risk of other autoimmune diseases. METHODS: To identify generalized vitiligo susceptibility loci, we conducted a genomewide association study. We genotyped 579,146 single-nucleotide polymorphisms (SNPs) in 1514 patients with generalized vitiligo who were of European-derived white (CEU) ancestry and compared the genotypes with publicly available control genotypes from 2813 CEU persons. We then tested 50 SNPs in two replication sets, one comprising 677 independent CEU patients and 1106 CEU controls and the other comprising 183 CEU simplex trios with generalized vitiligo and 332 CEU multiplex families. RESULTS: We detected significant associations between generalized vitiligo and SNPs at several loci previously associated with other autoimmune diseases. These included genes encoding major-histocompatibility-complex class I molecules (P=9.05x10(-23)) and class II molecules (P=4.50x10(-34)), PTPN22 (P=1.31x10(-7)), LPP (P=1.01x10(-11)), IL2RA (P=2.78x10(-9)), UBASH3A (P=1.26x10(-9)), and C1QTNF6 (P=2.21x10(-16)). We also detected associations between generalized vitiligo and SNPs in two additional immune-related loci, RERE (P=7.07x10(-15)) and GZMB (P=3.44x10(-8)), and in a locus containing TYR (P=1.60x10(-18)), encoding tyrosinase. CONCLUSIONS: We observed associations between generalized vitiligo and markers implicating multiple genes, some associated with other autoimmune diseases and one (TYR) that may mediate target-cell specificity and indicate a mutually exclusive relationship between susceptibility to vitiligo and susceptibility to melanoma.


Asunto(s)
Enfermedades Autoinmunes/genética , Predisposición Genética a la Enfermedad , Complejo Mayor de Histocompatibilidad/genética , Monofenol Monooxigenasa/genética , Polimorfismo de Nucleótido Simple , Vitíligo/genética , Marcadores Genéticos , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Melanoma/genética , Vitíligo/inmunología
5.
J Transl Med ; 10: 32, 2012 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-22369243

RESUMEN

BACKGROUND: Historically, extended haplotypes have been defined using only a few data points, such as alleles for several HLA genes in the MHC. High-density SNP data, and the increasing affordability of whole genome SNP typing, creates the opportunity to define higher resolution extended haplotypes. This drives the need for new tools that support quantification and visualization of extended haplotypes as defined by as many as 2000 SNPs. Confronted with high-density SNP data across the major histocompatibility complex (MHC) for 2,300 complete families, compiled by the Type 1 Diabetes Genetics Consortium (T1DGC), we developed software for studying extended haplotypes. METHODS: The software, called ExHap (Extended Haplotype), uses a similarity measurement we term congruence to identify and quantify long-range allele identity. Using ExHap, we analyzed congruence in both the T1DGC data and family-phased data from the International HapMap Project. RESULTS: Congruent chromosomes from the T1DGC data have between 96.5% and 99.9% allele identity over 1,818 SNPs spanning 2.64 megabases of the MHC (HLA-DRB1 to HLA-A). Thirty-three of 132 DQ-DR-B-A defined haplotype groups have > 50% congruent chromosomes in this region. For example, 92% of chromosomes within the DR3-B8-A1 haplotype are congruent from HLA-DRB1 to HLA-A (99.8% allele identity). We also applied ExHap to all 22 autosomes for both CEU and YRI cohorts from the International HapMap Project, identifying multiple candidate extended haplotypes. CONCLUSIONS: Long-range congruence is not unique to the MHC region. Patterns of allele identity on phased chromosomes provide a simple, straightforward approach to visually and quantitatively inspect complex long-range structural patterns in the genome. Such patterns aid the biologist in appreciating genetic similarities and differences across cohorts, and can lead to hypothesis generation for subsequent studies.


Asunto(s)
Alelos , Genoma Humano/genética , Técnicas de Genotipaje/métodos , Haplotipos/genética , Algoritmos , Cromosomas Humanos/genética , Diabetes Mellitus Tipo 1/genética , Estudios de Asociación Genética , Proyecto Mapa de Haplotipos , Humanos , Complejo Mayor de Histocompatibilidad/genética , Recombinación Genética/genética , Programas Informáticos
6.
Proc Natl Acad Sci U S A ; 106(25): 10272-7, 2009 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-19509334

RESUMEN

Adequate responses by our innate immune system toward invading pathogens were of vital importance for surviving infections, especially before the antibiotic era. Recently, a polymorphism in Mal (Ser180Leu, TIRAP rs8177374), an important adaptor protein downstream of the Toll-like receptor (TLR) 2 and 4 pathways, has been described to provide protection against a broad range of infectious pathogens. We assessed the functional effects of this polymorphism in human experimental endotoxemia, and we demonstrate that individuals bearing the TIRAP 180L allele display an increased, innate immune response to TLR4 and TLR2 ligands, but not to TLR9 stimulation. This phenotype has been related to an increased resistance to infection. However, an overshoot in the release of proinflammatory cytokines by TIRAP 180L homozygous individuals suggests a scenario of balanced evolution. We have also investigated the worldwide distribution of the Ser180Leu polymorphism in 14 populations around the globe to correlate the genetic makeup of TIRAP with the local infectious pressures. Based on the immunological, clinical, and genetic data, we propose that this mutation might have been selected in West Eurasia during the early settlement of this region after the out-of-Africa migration of modern Homo sapiens. This combination of functional and genetic data provides unique insights to our understanding of the pathogenesis of sepsis.


Asunto(s)
Endotoxemia/genética , Endotoxemia/inmunología , Glicoproteínas de Membrana/fisiología , Receptores de Interleucina-1/fisiología , Selección Genética , Choque Séptico/genética , Choque Séptico/inmunología , Alelos , Humanos , Inmunidad Innata/genética , Leucina/genética , Glicoproteínas de Membrana/genética , Polimorfismo Genético , Receptores de Interleucina-1/genética , Serina/genética
7.
N Engl J Med ; 356(12): 1216-25, 2007 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-17377159

RESUMEN

BACKGROUND: Autoimmune and autoinflammatory diseases involve interactions between genetic risk factors and environmental triggers. We searched for a gene on chromosome 17p13 that contributes to a group of epidemiologically associated autoimmune and autoinflammatory diseases. The group includes various combinations of generalized vitiligo, autoimmune thyroid disease, latent autoimmune diabetes in adults, rheumatoid arthritis, psoriasis, pernicious anemia, systemic lupus erythematosus, and Addison's disease. METHODS: We tested 177 single-nucleotide polymorphisms (SNPs) spanning the 17p13 linkage peak for association with disease and identified a strong candidate gene. We then sequenced DNA in and around the gene to identify additional SNPs. We carried out a second round of tests of association using some of these additional SNPs, thus elucidating the association with disease in the gene and its extended promoter region in fine detail. RESULTS: Association analyses resulted in our identifying as a candidate gene NALP1, which encodes NACHT leucine-rich-repeat protein 1, a regulator of the innate immune system. Fine-scale association mapping with the use of DNA from affected families and additional SNPs in and around NALP1 showed an association of specific variants with vitiligo alone, with an extended autoimmune and autoinflammatory disease phenotype, or with both. Conditional logistic-regression analysis of NALP1 SNPs indicated that at least two variants contribute independently to the risk of disease. CONCLUSIONS: DNA sequence variants in the NALP1 region are associated with the risk of several epidemiologically associated autoimmune and autoinflammatory diseases, implicating the innate immune system in the pathogenesis of these disorders.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Enfermedades Autoinmunes/genética , Cromosomas Humanos Par 17 , Predisposición Genética a la Enfermedad , Vitíligo/genética , Secuencia de Aminoácidos , Ligamiento Genético , Variación Genética , Genotipo , Humanos , Patrón de Herencia , Modelos Logísticos , Datos de Secuencia Molecular , Proteínas NLR , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Alineación de Secuencia , Análisis de Secuencia de ADN , Vitíligo/inmunología
8.
Clin Cancer Res ; 15(8): 2666-74, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19351763

RESUMEN

PURPOSE: We have previously mapped a major susceptibility locus influencing familial lung cancer risk to chromosome 6q23-25. However, the causal gene at this locus remains undetermined. In this study, we further refined this locus to identify a single candidate gene, by fine mapping using microsatellite markers and association studies using high-density single nucleotide polymorphisms (SNP). EXPERIMENTAL DESIGN: Six multigenerational families with five or more affected members were chosen for fine-mapping the 6q linkage region using microsatellite markers. For association mapping, we genotyped 24 6q-linked cases and 72 unrelated noncancer controls from the Genetic Epidemiology of Lung Cancer Consortium resources using the Affymetrix 500K chipset. Significant associations were validated in two independent familial lung cancer populations: 226 familial lung cases and 313 controls from the Genetic Epidemiology of Lung Cancer Consortium, and 154 familial cases and 325 controls from Mayo Clinic. Each familial case was chosen from one high-risk lung cancer family that has three or more affected members. RESULTS: A region-wide scan across 6q23-25 found significant association between lung cancer susceptibility and three single nucleotide polymorphisms in the first intron of the RGS17 gene. This association was further confirmed in two independent familial lung cancer populations. By quantitative real-time PCR analysis of matched tumor and normal human tissues, we found that RGS17 transcript accumulation is highly and consistently increased in sporadic lung cancers. Human lung tumor cell proliferation and tumorigenesis in nude mice are inhibited upon knockdown of RGS17 levels. CONCLUSION: RGS17 is a major candidate for the familial lung cancer susceptibility locus on chromosome 6q23-25.


Asunto(s)
Cromosomas Humanos Par 6/genética , Predisposición Genética a la Enfermedad , Neoplasias Pulmonares/genética , Proteínas RGS/genética , Anciano , Animales , Línea Celular Tumoral , Mapeo Cromosómico , Femenino , Técnicas de Silenciamiento del Gen , Genotipo , Haplotipos/genética , Humanos , Pulmón/patología , Neoplasias Pulmonares/epidemiología , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Desnudos , Repeticiones de Microsatélite/genética , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple/genética , Proteínas RGS/metabolismo , ARN Interferente Pequeño/metabolismo , Trasplante Heterólogo/patología
9.
Cancer Res ; 67(10): 4665-70, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17510392

RESUMEN

The use of tyrosine kinase inhibitors (TKI) has yielded great success in treatment of lung adenocarcinomas. However, patients who develop resistance to TKI treatment often acquire a somatic resistance mutation (T790M) located in the catalytic cleft of the epidermal growth factor receptor (EGFR) enzyme. Recently, a report describing EGFR-T790M as a germ-line mutation suggested that this mutation may be associated with inherited susceptibility to lung cancer. Contrary to previous reports, our analysis indicates that the T790M mutation confers increased Y992 and Y1068 phosphorylation levels. In a human bronchial epithelial cell line, overexpression of EGFR-T790M displayed a growth advantage over wild-type (WT) EGFR. We also screened 237 lung cancer family probands, in addition to 45 bronchoalveolar tumors, and found that none of them contained the EGFR-T790M mutation. Our observations show that EGFR-T790M provides a proliferative advantage with respect to WT EGFR and suggest that the enhanced kinase activity of this mutant is the basis for rare cases of inherited susceptibility to lung cancer.


Asunto(s)
Alelos , Receptores ErbB/metabolismo , Genes erbB-1 , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Animales , Células COS , Chlorocebus aethiops , ADN de Neoplasias/genética , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Humanos , Linaje , Fosforilación
10.
Cancer Res ; 67(1): 93-9, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17210687

RESUMEN

In this study, we observed loss of heterozygosity (LOH) in human chromosomal fragment 6q25.1 in sporadic lung cancer patients. LOH was observed in 65% of the 26 lung tumors examined and was narrowed down to a 2.2-Mb region. Single-nucleotide polymorphism (SNP) analysis of genes located within this region identified a candidate gene, termed p34. This gene, also designated as ZC3H12D, C6orf95, FLJ46041, or dJ281H8.1, carries an A/G nonsynonymous SNP at codon 106, which alters the amino acid from lysine to arginine. Nearly 73% of heterozygous lung cancer tissues with LOH and the A/G SNP also exhibited loss of the A allele. In vitro clonogenic and in vivo nude mouse studies showed that overexpression of the A allele exerts tumor suppressor function compared with the G allele. p34 is located within a recently mapped human lung cancer susceptibility locus, and association of the p34 A/G SNP was tested among these families. No significant association between the less frequent G allele and lung cancer susceptibility was found. Our results suggest that p34 may be a novel tumor suppressor gene involved in sporadic lung cancer but it seems not to be the candidate familial lung cancer susceptibility gene linked to chromosomal region 6q23-25.


Asunto(s)
Cromosomas Humanos Par 6 , Genes Supresores de Tumor , Pérdida de Heterocigocidad , Neoplasias Pulmonares/genética , Alelos , Animales , Secuencia de Bases , Codón , Femenino , Predisposición Genética a la Enfermedad , Humanos , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple
11.
Am J Kidney Dis ; 52(6): 1042-50, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18640754

RESUMEN

BACKGROUND: At the University of Colorado Health Sciences Center, on detailed questioning, approximately 10% of patients with autosomal dominant polycystic kidney disease (ADPKD) gave no family history of ADPKD. There are several explanations for this observation, including occurrence of a de novo pathogenic sequence variant or extreme phenotypic variability. To confirm de novo sequence variants, we have undertaken clinical and genetic screening of affected offspring and their parents. STUDY DESIGN: Case series. SETTING & PARTICIPANTS: 24 patients with a well-documented ADPKD phenotype and no family history of polycystic kidney disease (PKD) and both parents of each patient. OUTCOME: Presence or absence of PKD1 or PKD2 pathogenic sequence variants in parents of affected offspring. MEASUREMENTS: Abdominal ultrasound of affected offspring and their parents for ADPKD diagnosis. Parentage testing by genotyping. Complete screening of PKD1 and PKD2 genes by using genomic DNA from affected offspring; analysis of genomic DNA from both parents to confirm the absence or presence of all DNA variants found. RESULTS: A positive diagnosis of ADPKD by means of ultrasound or genetic screening was made in 1 parent of 4 patients (17%). No PKD1 or PKD2 pathogenic sequence variants were identified in 10 patients (42%), whereas possible pathological DNA variants were identified in 4 patients (17%) and 1 of their respective parents. Parentage was confirmed in the remaining 6 patients (25%), and de novo sequence variants were documented. LIMITATIONS: Size of patient group. No direct examination of RNA. CONCLUSION: Causes other than de novo pathogenic sequence variants may explain the negative family history of ADPKD in certain families.


Asunto(s)
Mutación , Riñón Poliquístico Autosómico Dominante/genética , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Pruebas Genéticas , Humanos , Masculino , Persona de Mediana Edad , Canales Catiónicos TRPP/genética , Adulto Joven
12.
Diabetes ; 55(5): 1265-9, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16644681

RESUMEN

Technology has become available to cost-effectively analyze thousands of single nucleotide polymorphisms (SNPs). We recently confirmed by genotyping a small series of class I alleles and microsatellite markers that the extended haplotype HLA-A1-B8-DR3 (8.1 AH) at the major histocompatibility complex (MHC) is a common and conserved haplotype. To further evaluate the region of conservation of the DR3 haplotypes, we genotyped 31 8.1 AHs and 29 other DR3 haplotypes with a panel of 656 SNPs spanning 4.8 Mb in the MHC region. This multi-SNP evaluation revealed a 2.9-Mb region that was essentially invariable for all 31 8.1 AHs. The 31 8.1 AHs were >99.9% identical for 384 consecutive SNPs of the 656 SNPs analyzed. Future association studies of MHC-linked susceptibility to type 1 diabetes will need to account for the extensive conservation of the 8.1 AH, since individuals who carry this haplotype provide no information about the differential effects of the alleles that are present on this haplotype.


Asunto(s)
Antígeno HLA-A1/genética , Antígeno HLA-B8/genética , Antígeno HLA-DR3/genética , Complejo Mayor de Histocompatibilidad , Polimorfismo de Nucleótido Simple , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Predisposición Genética a la Enfermedad , Homocigoto , Humanos
13.
Circulation ; 112(1): 54-9, 2005 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-15998695

RESUMEN

BACKGROUND: Mutations in the beta-myosin heavy-chain (betaMyHC) gene cause hypertrophic (HCM) and dilated (DCM) forms of cardiomyopathy. In failing human hearts, downregulation of alphaMyHC mRNA or protein has been correlated with systolic dysfunction. We hypothesized that mutations in alphaMyHC could also lead to pleiotropic cardiac phenotypes, including HCM and DCM. METHODS AND RESULTS: A cohort of 434 subjects, 374 (134 affected, 214 unaffected, 26 unknown) belonging to 69 DCM families and 60 (29 affected, 30 unaffected, 1 unknown) in 21 HCM families, was screened for alphaMyHC gene (MYH6) mutations. Three heterozygous MYH6 missense mutations were identified in DCM probands (P830L, A1004S, and E1457K; 4.3% of probands). A Q1065H mutation was detected in 1 of 21 HCM probands and was absent in 2 unaffected offspring. All MYH6 mutations were distributed in highly conserved residues, were predicted to change the structure or chemical bonds of alphaMyHC, and were absent in at least 300 control chromosomes from an ethnically similar population. The DCM carrier phenotype was characterized by late onset, whereas the HCM phenotype was characterized by progression toward dilation, left ventricular dysfunction, and refractory heart failure. CONCLUSIONS: This study suggests that mutations in MYH6 may cause a spectrum of phenotypes ranging from DCM to HCM.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Hipertrófica/genética , Mutación Missense , Cadenas Pesadas de Miosina/genética , Miosinas Ventriculares/genética , Cardiomiopatía Dilatada/epidemiología , Cardiomiopatía Hipertrófica/epidemiología , Estudios de Casos y Controles , Secuencia Conservada , Análisis Mutacional de ADN , Salud de la Familia , Femenino , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/genética , Heterocigoto , Humanos , Masculino , Epidemiología Molecular , Linaje , Sarcómeros/genética , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/genética
14.
Diabetes ; 54(6): 1879-83, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15919812

RESUMEN

To evaluate potential differential diabetes risk of DR3 haplotypes we have evaluated class I alleles as well as two microsatellites previously associated with differential risk associated with DR3 haplotypes. We found that over one-third of patient DR3 chromosomes consisted of an extended DR3 haplotype, from DQ2 to D6S2223 (DQ2, DR3, D6S273-143, MIC-A5.1, HLA-B8, HLA-Cw7, HLA-A1, and D6S2223-177) with an identical extended haplotype in controls. The extended haplotype was present more frequently (35.1% of autoimmune-associated DR3 haplotypes, 39.4% of control DR3 haplotypes) than other haplotypes (no other haplotype >5% of DR3 haplotypes) and remarkably conserved, but it was not transmitted from parents to affected children more frequently than nonconserved DR3-bearing haplotypes. This suggests that if all alleles are truly identical for the major A1, B8, DR3 haplotype (between A1 and DR3), with different alleles on nonconserved haplotypes without differential diabetes risk, then in this region of the genome DR3-DQ2 may be the primary polymorphisms of common haplotypes contributing to diabetes risk.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad , Antígeno HLA-A1/genética , Antígeno HLA-B8/genética , Antígeno HLA-DR3/genética , Genotipo , Haplotipos , Humanos , Desequilibrio de Ligamiento , Polimorfismo Genético , Factores de Riesgo
16.
Hum Immunol ; 67(11): 940-50, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17145374

RESUMEN

We report the prevalence of celiac disease (CD) and its relationship with other autoimmune diseases and HLA haplotypes in a Bedouin kindred. Of 175 individuals sampled and typed for autoantibodies and HLA class II genotypes, six (3.4%) members had CD, and an additional 10 (5.7%) members tested positive for autoantibodies to transglutaminase (TgAA+). Several CD/TgAA+ relatives also had islet cell antigen or adrenal autoimmunity. Affected relatives are more closely related than expected from the pedigree relationships of all family members and were more often the offspring of consanguineous marriages. Individuals with CD or TgAA+ were enriched for DRB1*0301-DQA1*0501-DQB1*0201, a haplotype previously reported as high risk for CD. There was also an increased frequency of DQB1*0201/DQB1*0201 homozygotes among affected relatives. We found no evidence that DRB1*0701-DQA1*0201-DQB1*0201/DRB1*11-DQA1*0501-DQB1*0301 is a high-risk genotype, consistent with other studies of Arab communities. In addition, a nonparametric linkage analysis of 376 autosomal markers revealed suggestive evidence for linkage on chromosome 12p13 at marker D12S364 (NPL = 2.009, p = 0.0098). There were no other significant results, including the HLA region or any other previously reported regions. This could reflect the reduced power of family-based linkage and association analyses in isolated inbred populations.


Asunto(s)
Árabes , Enfermedad Celíaca/inmunología , Antígenos HLA/genética , Autoanticuerpos/sangre , Enfermedad Celíaca/etnología , Enfermedad Celíaca/genética , Ligamiento Genético , Homocigoto , Humanos , Transglutaminasas/inmunología
17.
Nat Genet ; 48(11): 1418-1424, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27723757

RESUMEN

Vitiligo is an autoimmune disease in which depigmented skin results from the destruction of melanocytes, with epidemiological association with other autoimmune diseases. In previous linkage and genome-wide association studies (GWAS1 and GWAS2), we identified 27 vitiligo susceptibility loci in patients of European ancestry. We carried out a third GWAS (GWAS3) in European-ancestry subjects, with augmented GWAS1 and GWAS2 controls, genome-wide imputation, and meta-analysis of all three GWAS, followed by an independent replication. The combined analyses, with 4,680 cases and 39,586 controls, identified 23 new significantly associated loci and 7 suggestive loci. Most encode immune and apoptotic regulators, with some also associated with other autoimmune diseases, as well as several melanocyte regulators. Bioinformatic analyses indicate a predominance of causal regulatory variation, some of which corresponds to expression quantitative trait loci (eQTLs) at these loci. Together, the identified genes provide a framework for the genetic architecture and pathobiology of vitiligo, highlight relationships with other autoimmune diseases and melanoma, and offer potential targets for treatment.


Asunto(s)
Enfermedades Autoinmunes/genética , Predisposición Genética a la Enfermedad , Vitíligo/genética , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Melanoma/genética , Sitios de Carácter Cuantitativo , Medición de Riesgo
18.
Hum Mutat ; 26(6): 566-74, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16247757

RESUMEN

Thymopoietin or TMPO (indicated by its alternative gene symbol, LAP2, in this work) has been proposed as a candidate disease gene for dilated cardiomyopathy (DCM), since a LAP2 product associates with nucleoplasmic lamins A/C, which are encoded by the DCM gene LMNA. We developed a study to screen for genetic mutations in LAP2 in a large collection of DCM patients and families. A total of 113 subjects from 88 families (56 with familial DCM (FDC) and 32 with sporadic DCM) were screened for LAP2 mutations using denaturing high-performance liquid chromatography and sequence analysis. We found a single putative mutation affecting the LAP2alpha isoform in one FDC pedigree. The mutation predicts an Arg690Cys substitution (c.2068C>T; p.R690C) located in the C-terminal domain of the LAP2alpha protein, a region that is known to interact with lamin A/C. RT-PCR, Western blot analyses, and immunolocalization revealed low-level LAP2alpha expression in adult cardiac muscle, and localization to a subset of nuclei. Mutated Arg690Cys LAP2alpha expressed in HeLa cells localized to the nucleoplasm like wild-type LAP2alpha, with no effect on peripheral and nucleoplasmic lamin A distribution. However, the in vitro interaction of mutated LAP2alpha with the pre-lamin A C-terminus was significantly compromised compared to the wild-type protein. LAP2 mutations may represent a rare cause of DCM. The Arg690Cys mutation altered the observed LAP2alpha interaction with A-type lamins. Our finding implicates a novel nuclear lamina-associated protein in the pathogenesis of genetic forms of dilated cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/genética , Proteínas de Unión al ADN/genética , Predisposición Genética a la Enfermedad , Proteínas de la Membrana/genética , Mutación Missense , Cromatografía Liquida , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Femenino , Pruebas Genéticas , Células HeLa , Humanos , Lamina Tipo A/química , Lamina Tipo A/metabolismo , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Miocardio/citología , Miocardio/metabolismo , Linaje , Isoformas de Proteínas/genética , Estructura Terciaria de Proteína
19.
J Clin Endocrinol Metab ; 90(1): 128-34, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15483092

RESUMEN

Individuals with type 1 diabetes mellitus (T1D) at risk for Addison's disease (AD) can be identified with RIAs for autoantibodies to the adrenal antigen 21-hydroxylase (21-OHAA). Screening individuals with T1D for 21OH-AA shows a relatively high prevalence of positive autoantibodies (1.4%, 38 of 2696 subjects). After detection of 21-OHAA, individuals were evaluated with endocrine testing, including baseline cortisol, ACTH, and plasma renin activity and low (1 mug) and high (250 mug) dose cortrosyn stimulation. Typing for DR and DQ alleles and for the major histocompatibility complex class I-related chain A (MICA) gene polymorphisms was performed. Six individuals were diagnosed with AD; five were identified on initial endocrine evaluation. Follow-up over 2.9 yr yielded one additional diagnosis of AD. Endocrine testing showed a correlation between baseline ACTH and peak cortisol (r = -0.61; P < 0.0001), baseline and peak cortisol (r = 0.70; P < 0.0001), and stimulated cortisol after low- and high-dose testing (r = 0.92; P < 0.0001). DR3-DQ2/DR4-DQ8 with DRB1* 0404 was associated with expression of 21-OHAA. At 2 yr, individuals homozygous for MICA5.1 had AD-free survival of 60% compared with 100% AD-free survival in those who were not homozygous for MICA5.1. Homozygosity for MICA5.1 may increase progression to overt AD among 21-OHAA-positive individuals.


Asunto(s)
Enfermedad de Addison/etiología , Autoanticuerpos/sangre , Diabetes Mellitus Tipo 1/complicaciones , Esteroide 21-Hidroxilasa/inmunología , Adolescente , Hormona Adrenocorticotrópica/sangre , Adulto , Anciano , Niño , Preescolar , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Antígenos de Histocompatibilidad Clase I , Prueba de Histocompatibilidad , Humanos , Hidrocortisona/sangre , Masculino , Persona de Mediana Edad , Proteínas/análisis , Sensibilidad y Especificidad
20.
J Am Coll Cardiol ; 41(5): 771-80, 2003 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-12628721

RESUMEN

OBJECTIVES: We examined the prevalence, genotype-phenotype correlation, and natural history of lamin A/C gene (LMNA) mutations in subjects with dilated cardiomyopathy (DCM). BACKGROUND: Mutations in LMNA have been found in patients with DCM with familial conduction defects and muscular dystrophy, but the clinical spectrum, prognosis, and clinical relevance of laminopathies in DCM are unknown. BACKGROUND: A cohort of 49 nuclear families, 40 with familial DCM and 9 with sporadic DCM (269 subjects, 105 affected), was screened for mutations in LMNA using denaturing high-performance liquid chromatography and sequence analysis. Bivariate analysis of clinical predictors of LMNA mutation carrier status and Kaplan-Meier survival analysis were performed. RESULTS: Mutations in LMNA were detected in four families (8%), three with familial (R89L, 959delT, R377H) and one with sporadic DCM (S573L). There was significant phenotypic variability, but the presence of skeletal muscle involvement (p < 0.001), supraventricular arrhythmia (p = 0.003), conduction defects (p = 0.01), and "mildly" DCM (p = 0.006) were predictors of LMNA mutations. The LMNA mutation carriers had a significantly poorer cumulative survival compared with non-carrier DCM patients: event-free survival at the age of 45 years was 31% versus 75% in non-carriers. CONCLUSIONS: Mutations in LMNA cause a severe and progressive DCM in a relevant proportion of patients. Mutation screening should be considered in patients with DCM, in particular when clinical predictors of LMNA mutation are present, regardless of family history.


Asunto(s)
Cardiomiopatía Dilatada/epidemiología , Cardiomiopatía Dilatada/genética , Predisposición Genética a la Enfermedad , Mutación Missense , Lámina Nuclear/genética , Secuencia de Aminoácidos , Cardiomiopatía Dilatada/diagnóstico , Preescolar , Cromatografía Líquida de Alta Presión , Estudios de Cohortes , Análisis Mutacional de ADN , Femenino , Estudios de Seguimiento , Genotipo , Humanos , Masculino , Linaje , Fenotipo , Reacción en Cadena de la Polimerasa , Prevalencia , Probabilidad , Medición de Riesgo , Índice de Severidad de la Enfermedad , Estadísticas no Paramétricas , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA