Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Orphanet J Rare Dis ; 18(1): 360, 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37974153

RESUMEN

BACKGROUND: Hypoketotic hypoglycaemia with suppressed plasma fatty acids and detectable insulin suggests congenital hyperinsulinism (CHI). Severe hypoketotic hypoglycaemia mimicking hyperinsulinism but without detectable insulin has recently been described in syndromic individuals with mosaic genetic activation of post-receptor insulin signalling. We set out to expand understanding of this entity focusing on metabolic phenotypes. METHODS: Metabolic profiling, candidate gene and exome sequencing were performed in six infants with hypoketotic, hypoinsulinaemic hypoglycaemia, with or without syndromic features. Additional signalling studies were carried out in dermal fibroblasts from two individuals. RESULTS: Two infants had no syndromic features. One was mistakenly diagnosed with CHI. One had mild features of megalencephaly-capillary malformation-polymicrogyria (MCAP) syndrome, one had non-specific macrosomia, and two had complex syndromes. All required intensive treatment to maintain euglycaemia, with CHI-directed therapies being ineffective. Pathogenic PIK3CA variants were found in two individuals - de novo germline c.323G>A (p.Arg108His) in one non-syndromic infant and postzygotic mosaic c.2740G>A (p.Gly914Arg) in the infant with MCAP. No causal variants were proven in the other individuals despite extensive investigation, although rare variants in mTORC components were identified in one. No increased PI3K signalling in fibroblasts of two individuals was seen. CONCLUSIONS: We expand the spectrum of PI3K-related hypoinsulinaemic hypoketotic hypoglycaemia. We demonstrate that pathogenic germline variants activating post-insulin-receptor signalling may cause non-syndromic hypoinsulinaemic hypoketotic hypoglycaemia closely resembling CHI. This distinct biochemical footprint should be sought and differentiated from CHI in infantile hypoglycaemia. To facilitate adoption of this differential diagnosis, we propose the term "pseudohyperinsulinism".


Asunto(s)
Hiperinsulinismo Congénito , Proteínas Proto-Oncogénicas c-akt , Lactante , Humanos , Proteínas Proto-Oncogénicas c-akt/genética , Insulina , Hiperinsulinismo Congénito/genética , Fosfatidilinositol 3-Quinasas/metabolismo
2.
Cell Mol Gastroenterol Hepatol ; 14(6): 1295-1310, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36038072

RESUMEN

BACKGROUND & AIMS: Human intestinal epithelial organoids (IEOs) are a powerful tool to model major aspects of intestinal development, health, and diseases because patient-derived cultures retain many features found in vivo. A necessary aspect of the organoid model is the requirement to expand cultures in vitro through several rounds of passaging. This is of concern because the passaging of cells has been shown to affect cell morphology, ploidy, and function. METHODS: Here, we analyzed 173 human IEO lines derived from the small and large bowel and examined the effect of culture duration on DNA methylation (DNAm). Furthermore, we tested the potential impact of DNAm changes on gene expression and cellular function. RESULTS: Our analyses show a reproducible effect of culture duration on DNAm in a large discovery cohort as well as 2 publicly available validation cohorts generated in different laboratories. Although methylation changes were seen in only approximately 8% of tested cytosine-phosphate-guanine dinucleotides (CpGs) and global cellular function remained stable, a subset of methylation changes correlated with altered gene expression at baseline as well as in response to inflammatory cytokine exposure and withdrawal of Wnt agonists. Importantly, epigenetic changes were found to be enriched in genomic regions associated with colonic cancer and distant to the site of replication, indicating similarities to malignant transformation. CONCLUSIONS: Our study shows distinct culture-associated epigenetic changes in mucosa-derived human IEOs, some of which appear to impact gene transcriptomic and cellular function. These findings highlight the need for future studies in this area and the importance of considering passage number as a potentially confounding factor.


Asunto(s)
Metilación de ADN , Organoides , Humanos , Intestinos , Epigénesis Genética , Mucosa Intestinal
3.
Clin Epigenetics ; 13(1): 81, 2021 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-33865442

RESUMEN

Eosinophilic esophagitis (EoE) is a leading cause of dysphagia and food impaction in children and adults. The diagnosis relies on histological examination of esophageal mucosal biopsies and requires the presence of > 15 eosinophils per high-powered field. Potential pitfalls include the impact of biopsy sectioning as well as regional variations of eosinophil density. We performed genome-wide DNA methylation analyses on 30 esophageal biopsies obtained from children diagnosed with EoE (n = 7) and matched controls (n = 13) at the time of diagnosis as well as following first-line treatment. Analyses revealed striking disease-associated differences in mucosal DNA methylation profiles in children diagnosed with EoE, highlighting the potential for these epigenetic signatures to be developed into clinically applicable biomarkers.


Asunto(s)
Metilación de ADN/genética , Esofagitis Eosinofílica/genética , Esofagitis Eosinofílica/patología , Estudio de Asociación del Genoma Completo/métodos , Adolescente , Biopsia , Niño , Preescolar , Esófago/patología , Femenino , Humanos , Italia , Masculino , Estudios Prospectivos
4.
Sci Rep ; 7(1): 17593, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29242557

RESUMEN

Common genetic variants at the ARL15 locus are associated with plasma adiponectin, insulin and HDL cholesterol concentrations, obesity, and coronary atherosclerosis. The ARL15 gene encodes a small GTP-binding protein whose function is currently unknown. In this study adipocyte-autonomous roles for ARL15 were investigated using conditional knockdown of Arl15 in murine 3T3-L1 (pre)adipocytes. Arl15 knockdown in differentiated adipocytes impaired adiponectin secretion but not adipsin secretion or insulin action, while in preadipocytes it impaired adipogenesis. In differentiated adipocytes GFP-tagged ARL15 localized predominantly to the Golgi with lower levels detected at the plasma membrane and intracellular vesicles, suggesting involvement in intracellular trafficking. Sequencing of ARL15 in 375 severely insulin resistant patients identified four rare heterozygous variants, including an early nonsense mutation in a proband with femorogluteal lipodystrophy and non classical congenital adrenal hyperplasia, and an essential splice site mutation in a proband with partial lipodystrophy and a history of childhood yolk sac tumour. No nonsense or essential splice site mutations were found in 2,479 controls, while five such variants were found in the ExAC database. These findings provide evidence that ARL15 plays a role in adipocyte differentiation and adiponectin secretion, and raise the possibility that human ARL15 haploinsufficiency predisposes to lipodystrophy.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Adipocitos/metabolismo , Adipocitos/patología , Adiponectina/metabolismo , Diferenciación Celular , Síndrome Metabólico/metabolismo , Síndrome Metabólico/patología , Células 3T3-L1 , Factores de Ribosilacion-ADP/genética , Adipogénesis , Adulto , Animales , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Aparato de Golgi/metabolismo , Células HEK293 , Haploinsuficiencia , Humanos , Resistencia a la Insulina , Lipodistrofia/genética , Lipodistrofia/metabolismo , Masculino , Síndrome Metabólico/genética , Ratones , Persona de Mediana Edad , Transporte de Proteínas , Adulto Joven
5.
Eur J Endocrinol ; 177(2): 175-186, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28566443

RESUMEN

OBJECTIVE: Genetic activation of the insulin signal-transducing kinase AKT2 causes syndromic hypoketotic hypoglycaemia without elevated insulin. Mosaic activating mutations in class 1A phospatidylinositol-3-kinase (PI3K), upstream from AKT2 in insulin signalling, are known to cause segmental overgrowth, but the metabolic consequences have not been systematically reported. We assess the metabolic phenotype of 22 patients with mosaic activating mutations affecting PI3K, thereby providing new insight into the metabolic function of this complex node in insulin signal transduction. METHODS: Three patients with megalencephaly, diffuse asymmetric overgrowth, hypoketotic, hypoinsulinaemic hypoglycaemia and no AKT2 mutation underwent further genetic, clinical and metabolic investigation. Signalling in dermal fibroblasts from one patient and efficacy of the mTOR inhibitor Sirolimus on pathway activation were examined. Finally, the metabolic profile of a cohort of 19 further patients with mosaic activating mutations in PI3K was assessed. RESULTS: In the first three patients, mosaic mutations in PIK3CA (p.Gly118Asp or p.Glu726Lys) or PIK3R2 (p.Gly373Arg) were found. In different tissue samples available from one patient, the PIK3CA p.Glu726Lys mutation was present at burdens from 24% to 42%, with the highest level in the liver. Dermal fibroblasts showed increased basal AKT phosphorylation which was potently suppressed by Sirolimus. Nineteen further patients with mosaic mutations in PIK3CA had neither clinical nor biochemical evidence of hypoglycaemia. CONCLUSIONS: Mosaic mutations activating class 1A PI3K cause severe non-ketotic hypoglycaemia in a subset of patients, with the metabolic phenotype presumably related to the extent of mosaicism within the liver. mTOR or PI3K inhibitors offer the prospect for future therapy.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/genética , Hipoglucemia/genética , Insulina/genética , Megalencefalia/genética , Mosaicismo , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Preescolar , Femenino , Humanos , Hipoglucemia/diagnóstico , Hipoglucemia/metabolismo , Insulina/metabolismo , Masculino , Megalencefalia/diagnóstico , Megalencefalia/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
6.
JCI Insight ; 1(17): e88766, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27766312

RESUMEN

Obesity-related insulin resistance is associated with fatty liver, dyslipidemia, and low plasma adiponectin. Insulin resistance due to insulin receptor (INSR) dysfunction is associated with none of these, but when due to dysfunction of the downstream kinase AKT2 phenocopies obesity-related insulin resistance. We report 5 patients with SHORT syndrome and C-terminal mutations in PIK3R1, encoding the p85α/p55α/p50α subunits of PI3K, which act between INSR and AKT in insulin signaling. Four of 5 patients had extreme insulin resistance without dyslipidemia or hepatic steatosis. In 3 of these 4, plasma adiponectin was preserved, as in insulin receptor dysfunction. The fourth patient and her healthy mother had low plasma adiponectin associated with a potentially novel mutation, p.Asp231Ala, in adiponectin itself. Cells studied from one patient with the p.Tyr657X PIK3R1 mutation expressed abundant truncated PIK3R1 products and showed severely reduced insulin-stimulated association of mutant but not WT p85α with IRS1, but normal downstream signaling. In 3T3-L1 preadipocytes, mutant p85α overexpression attenuated insulin-induced AKT phosphorylation and adipocyte differentiation. Thus, PIK3R1 C-terminal mutations impair insulin signaling only in some cellular contexts and produce a subphenotype of insulin resistance resembling INSR dysfunction but unlike AKT2 dysfunction, implicating PI3K in the pathogenesis of key components of the metabolic syndrome.


Asunto(s)
Resistencia a la Insulina/genética , Mutación , Fosfatidilinositol 3-Quinasas/genética , Células 3T3-L1 , Adipocitos , Adolescente , Animales , Niño , Fosfatidilinositol 3-Quinasa Clase Ia , Dislipidemias , Hígado Graso , Femenino , Células HEK293 , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Masculino , Ratones , Persona de Mediana Edad , Fosforilación
7.
J Mol Endocrinol ; 55(2): 147-58, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26336158

RESUMEN

We describe a female proband with primordial dwarfism, skeletal dysplasia, facial dysmorphism, extreme dyslipidaemic insulin resistance and fatty liver associated with a novel homozygous frameshift mutation in POC1A, predicted to affect two of the three protein products of the gene. POC1A encodes a protein associated with centrioles throughout the cell cycle and implicated in both mitotic spindle and primary ciliary function. Three homozygous mutations affecting all isoforms of POC1A have recently been implicated in a similar syndrome of primordial dwarfism, although no detailed metabolic phenotypes were described. Primary cells from the proband we describe exhibited increased centrosome amplification and multipolar spindle formation during mitosis, but showed normal DNA content, arguing against mitotic skipping, cleavage failure or cell fusion. Despite evidence of increased DNA damage in cells with supernumerary centrosomes, no aneuploidy was detected. Extensive centrosome clustering both at mitotic spindles and in primary cilia mitigated the consequences of centrosome amplification, and primary ciliary formation was normal. Although further metabolic studies of patients with POC1A mutations are warranted, we suggest that POC1A may be added to ALMS1 and PCNT as examples of centrosomal or pericentriolar proteins whose dysfunction leads to extreme dyslipidaemic insulin resistance. Further investigation of links between these molecular defects and adipose tissue dysfunction is likely to yield insights into mechanisms of adipose tissue maintenance and regeneration that are critical to metabolic health.


Asunto(s)
Estatura/genética , Centriolos/genética , Enanismo/genética , Resistencia a la Insulina/genética , Proteínas/genética , Adulto , Secuencia de Aminoácidos , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Centrosoma/fisiología , Proteínas del Citoesqueleto , Facies , Hígado Graso/genética , Femenino , Mutación del Sistema de Lectura/genética , Humanos , Mitosis/genética , Datos de Secuencia Molecular , Isoformas de Proteínas/genética , Alineación de Secuencia , Huso Acromático/fisiología
8.
Nat Genet ; 44(8): 928-33, 2012 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-22729222

RESUMEN

The phosphatidylinositol 3-kinase (PI3K)-AKT signaling pathway is critical for cellular growth and metabolism. Correspondingly, loss of function of PTEN, a negative regulator of PI3K, or activating mutations in AKT1, AKT2 or AKT3 have been found in distinct disorders featuring overgrowth or hypoglycemia. We performed exome sequencing of DNA from unaffected and affected cells from an individual with an unclassified syndrome of congenital progressive segmental overgrowth of fibrous and adipose tissue and bone and identified the cancer-associated mutation encoding p.His1047Leu in PIK3CA, the gene that encodes the p110α catalytic subunit of PI3K, only in affected cells. Sequencing of PIK3CA in ten additional individuals with overlapping syndromes identified either the p.His1047Leu alteration or a second cancer-associated alteration, p.His1047Arg, in nine cases. Affected dermal fibroblasts showed enhanced basal and epidermal growth factor (EGF)-stimulated phosphatidylinositol 3,4,5-trisphosphate (PIP(3)) generation and concomitant activation of downstream signaling relative to their unaffected counterparts. Our findings characterize a distinct overgrowth syndrome, biochemically demonstrate activation of PI3K signaling and thereby identify a rational therapeutic target.


Asunto(s)
Tejido Adiposo/enzimología , Tejido Adiposo/patología , Tejido Conectivo/enzimología , Tejido Conectivo/patología , Mutación , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Adolescente , Adulto , Secuencia de Bases , Huesos/enzimología , Huesos/patología , Niño , Preescolar , Fosfatidilinositol 3-Quinasa Clase I , Análisis Mutacional de ADN , Activación Enzimática/genética , Femenino , Humanos , Hiperplasia , Lactante , Masculino , Persona de Mediana Edad , Mosaicismo , Fenotipo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Síndrome
9.
Diabetes ; 59(5): 1266-75, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20185807

RESUMEN

OBJECTIVE Recent genome-wide association studies have revealed loci associated with glucose and insulin-related traits. We aimed to characterize 19 such loci using detailed measures of insulin processing, secretion, and sensitivity to help elucidate their role in regulation of glucose control, insulin secretion and/or action. RESEARCH DESIGN AND METHODS We investigated associations of loci identified by the Meta-Analyses of Glucose and Insulin-related traits Consortium (MAGIC) with circulating proinsulin, measures of insulin secretion and sensitivity from oral glucose tolerance tests (OGTTs), euglycemic clamps, insulin suppression tests, or frequently sampled intravenous glucose tolerance tests in nondiabetic humans (n = 29,084). RESULTS The glucose-raising allele in MADD was associated with abnormal insulin processing (a dramatic effect on higher proinsulin levels, but no association with insulinogenic index) at extremely persuasive levels of statistical significance (P = 2.1 x 10(-71)). Defects in insulin processing and insulin secretion were seen in glucose-raising allele carriers at TCF7L2, SCL30A8, GIPR, and C2CD4B. Abnormalities in early insulin secretion were suggested in glucose-raising allele carriers at MTNR1B, GCK, FADS1, DGKB, and PROX1 (lower insulinogenic index; no association with proinsulin or insulin sensitivity). Two loci previously associated with fasting insulin (GCKR and IGF1) were associated with OGTT-derived insulin sensitivity indices in a consistent direction. CONCLUSIONS Genetic loci identified through their effect on hyperglycemia and/or hyperinsulinemia demonstrate considerable heterogeneity in associations with measures of insulin processing, secretion, and sensitivity. Our findings emphasize the importance of detailed physiological characterization of such loci for improved understanding of pathways associated with alterations in glucose homeostasis and eventually type 2 diabetes.


Asunto(s)
Sitios Genéticos/fisiología , Glucosa/genética , Insulina/genética , Alelos , delta-5 Desaturasa de Ácido Graso , Ácido Graso Desaturasas/genética , Sitios Genéticos/genética , Estudio de Asociación del Genoma Completo , Quinasas del Centro Germinal , Glucosa/metabolismo , Proteínas de Homeodominio/genética , Humanos , Insulina/metabolismo , Metaanálisis como Asunto , Polimorfismo de Nucleótido Simple/genética , Proteínas Serina-Treonina Quinasas/genética , Receptores de la Hormona Gastrointestinal/genética , Factores de Transcripción TCF/genética , Proteína 2 Similar al Factor de Transcripción 7 , Proteínas Supresoras de Tumor/genética
10.
BMC Med Genet ; 8: 71, 2007 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-18045485

RESUMEN

BACKGROUND: As genes associated with immune-mediated diseases have an increased prior probability of being associated with other immune-mediated diseases, we tested three such genes, IL23R, IRF5 and CD40, for an association with type 1 diabetes. In addition, we tested seven genes, TAF5L, PDCD1, TCF7, IL12B, IL6, ICAM1 and TBX21, with published marginal or inconsistent evidence of an association with type 1 diabetes. METHODS: We genotyped reported polymorphisms of the ten genes, nonsynonymous SNPs (nsSNPs) and, for the IL12B and IL6 regions, tag SNPs in up to 7,888 case, 8,858 control and 3,142 parent-child trio samples. In addition, we analysed data from the Wellcome Trust Case Control Consortium genome-wide association study to determine whether there was any further evidence of an association in each gene region. RESULTS: We found some evidence of associations between type 1 diabetes and TAF5L, PDCD1, TCF7 and IL6 (ORs = 1.05 - 1.13; P = 0.0291 - 4.16 x 10-4). No evidence of an association was obtained for IL12B, IRF5, IL23R, ICAM1, TBX21 and CD40, although there was some evidence of an association (OR = 1.10; P = 0.0257) from the genome-wide association study for the ICAM1 region. CONCLUSION: We failed to exclude the possibility of some effect in type 1 diabetes for TAF5L, PDCD1, TCF7, IL6 and ICAM1. Additional studies, of these and other candidate genes, employing much larger sample sizes and analysis of additional polymorphisms in each gene and its flanking region will be required to ascertain their contributions to type 1 diabetes susceptibility.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad , Antígenos CD/genética , Proteínas Reguladoras de la Apoptosis/genética , Estudios de Casos y Controles , Marcadores Genéticos , Genotipo , Humanos , Molécula 1 de Adhesión Intercelular/genética , Interleucina-6/genética , Modelos Logísticos , Polimorfismo de Nucleótido Simple , Receptor de Muerte Celular Programada 1 , Factor 1 de Transcripción de Linfocitos T/genética , Factores Asociados con la Proteína de Unión a TATA/genética , Factor de Transcripción TFIID/genética
11.
J Leukoc Biol ; 81(3): 581-3, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17209142

RESUMEN

Gene-gene interaction analyses have been suggested as a potential strategy to help identify common disease susceptibility genes. Recently, evidence of a statistical interaction between polymorphisms in two negative immunoregulatory genes, CBLB and CTLA4, has been reported in type 1 diabetes (T1D). This study, in 480 Danish families, reported an association between T1D and a synonymous coding SNP in exon 12 of the CBLB gene (rs3772534 G>A; minor allele frequency, MAF=0.24; derived relative risk, RR for G allele=1.78; P=0.046). Furthermore, evidence of a statistical interaction with the known T1D susceptibility-associated CTLA4 polymorphism rs3087243 (laboratory name CT60, G>A) was reported (P<0.0001), such that the CBLB SNP rs3772534 G allele was overtransmitted to offspring with the CTLA4 rs3087243 G/G genotype. We have, therefore, attempted to obtain additional support for this finding in both large family and case-control collections. In a primary analysis, no evidence for an association of the CBLB SNP rs3772534 with disease was found in either sample set (2162 parent-child trios, P=0.33; 3453 cases and 3655 controls, P=0.69). In the case-only statistical interaction analysis between rs3772534 and rs3087243, there was also no support for an effect (1994 T1D affected offspring, and 3215 cases, P=0.92). These data highlight the need for large, well-characterized populations, offering the possibility of obtaining additional support for initial observations owing to the low prior probability of identifying reproducible evidence of gene-gene interactions in the analysis of common disease-associated variants in human populations.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Antígenos CD/genética , Antígenos de Diferenciación/genética , Diabetes Mellitus Tipo 1/genética , Proteínas Proto-Oncogénicas c-cbl/genética , Animales , Antígeno CTLA-4 , Estudios de Casos y Controles , Dinamarca , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Humanos , Polimorfismo de Nucleótido Simple/genética , Ratas
12.
Diabetes ; 53(2): 505-9, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14747305

RESUMEN

The diabetes-prone BioBreeding (BB) and Komeda diabetes-prone (KDP) rats are both spontaneous animal models of human autoimmune, T-cell-associated type 1 diabetes. Both resemble the human disease, and consequently, susceptibility genes for diabetes found in these two strains can be considered as potential candidate genes in humans. Recently, a frameshift deletion in Ian4, a member of the immune-associated nucleotide (Ian)-related gene family, has been shown to map to BB rat Iddm1. In the KDP rat, a nonsense mutation in the T-cell regulatory gene, Cblb, has been described as a major susceptibility locus. Following a strategy of examining the human orthologues of susceptibility genes identified in animal models for association with type 1 diabetes, we identified single nucleotide polymorphisms (SNPs) from each gene by resequencing PCR product from at least 32 type 1 diabetic patients. Haplotype tag SNPs (htSNPs) were selected and genotyped in 754 affected sib-pair families from the U.K. and U.S. Evaluation of disease association by a multilocus transmission/disequilibrium test (TDT) gave a P value of 0.484 for IAN4L1 and 0.692 for CBLB, suggesting that neither gene influences susceptibility to common alleles of human type 1 diabetes in these populations.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Codón sin Sentido , Diabetes Mellitus Tipo 1/genética , Polimorfismo de Nucleótido Simple , Ubiquitina-Proteína Ligasas/genética , Animales , Familia , Mutación del Sistema de Lectura , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Desequilibrio de Ligamiento , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-cbl , Ratas , Ratas Endogámicas BB , Hermanos , Especificidad de la Especie
13.
Nature ; 423(6939): 506-11, 2003 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-12724780

RESUMEN

Genes and mechanisms involved in common complex diseases, such as the autoimmune disorders that affect approximately 5% of the population, remain obscure. Here we identify polymorphisms of the cytotoxic T lymphocyte antigen 4 gene (CTLA4)--which encodes a vital negative regulatory molecule of the immune system--as candidates for primary determinants of risk of the common autoimmune disorders Graves' disease, autoimmune hypothyroidism and type 1 diabetes. In humans, disease susceptibility was mapped to a non-coding 6.1 kb 3' region of CTLA4, the common allelic variation of which was correlated with lower messenger RNA levels of the soluble alternative splice form of CTLA4. In the mouse model of type 1 diabetes, susceptibility was also associated with variation in CTLA-4 gene splicing with reduced production of a splice form encoding a molecule lacking the CD80/CD86 ligand-binding domain. Genetic mapping of variants conferring a small disease risk can identify pathways in complex disorders, as exemplified by our discovery of inherited, quantitative alterations of CTLA4 contributing to autoimmune tissue destruction.


Asunto(s)
Antígenos de Diferenciación/genética , Enfermedades Autoinmunes/genética , Predisposición Genética a la Enfermedad/genética , Inmunoconjugados , Abatacept , Empalme Alternativo/genética , Animales , Antígenos CD , Secuencia de Bases , Antígeno CTLA-4 , Diabetes Mellitus Tipo 1/genética , Modelos Animales de Enfermedad , Genotipo , Enfermedad de Graves/genética , Humanos , Hipotiroidismo/genética , Ratones , Polimorfismo de Nucleótido Simple/genética , Isoformas de Proteínas/genética , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA