Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 376(17): 1615-1626, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28445677

RESUMEN

BACKGROUND: Genomewide association studies of autoimmune diseases have mapped hundreds of susceptibility regions in the genome. However, only for a few association signals has the causal gene been identified, and for even fewer have the causal variant and underlying mechanism been defined. Coincident associations of DNA variants affecting both the risk of autoimmune disease and quantitative immune variables provide an informative route to explore disease mechanisms and drug-targetable pathways. METHODS: Using case-control samples from Sardinia, Italy, we performed a genomewide association study in multiple sclerosis followed by TNFSF13B locus-specific association testing in systemic lupus erythematosus (SLE). Extensive phenotyping of quantitative immune variables, sequence-based fine mapping, cross-population and cross-phenotype analyses, and gene-expression studies were used to identify the causal variant and elucidate its mechanism of action. Signatures of positive selection were also investigated. RESULTS: A variant in TNFSF13B, encoding the cytokine and drug target B-cell activating factor (BAFF), was associated with multiple sclerosis as well as SLE. The disease-risk allele was also associated with up-regulated humoral immunity through increased levels of soluble BAFF, B lymphocytes, and immunoglobulins. The causal variant was identified: an insertion-deletion variant, GCTGT→A (in which A is the risk allele), yielded a shorter transcript that escaped microRNA inhibition and increased production of soluble BAFF, which in turn up-regulated humoral immunity. Population genetic signatures indicated that this autoimmunity variant has been evolutionarily advantageous, most likely by augmenting resistance to malaria. CONCLUSIONS: A TNFSF13B variant was associated with multiple sclerosis and SLE, and its effects were clarified at the population, cellular, and molecular levels. (Funded by the Italian Foundation for Multiple Sclerosis and others.).


Asunto(s)
Factor Activador de Células B/genética , Mutación INDEL , Lupus Eritematoso Sistémico/genética , Esclerosis Múltiple/genética , Autoinmunidad , Factor Activador de Células B/metabolismo , Estudios de Casos y Controles , Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Italia , Lupus Eritematoso Sistémico/inmunología , MicroARNs , Esclerosis Múltiple/inmunología , Fenotipo , Polimorfismo de Nucleótido Simple , Riesgo , Análisis de Secuencia de ARN , Transcripción Genética
2.
Am J Hum Genet ; 99(1): 236-45, 2016 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-27392078

RESUMEN

Crisponi syndrome (CS)/cold-induced sweating syndrome type 1 (CISS1) is a very rare autosomal-recessive disorder characterized by a complex phenotype with high neonatal lethality, associated with the following main clinical features: hyperthermia and feeding difficulties in the neonatal period, scoliosis, and paradoxical sweating induced by cold since early childhood. CS/CISS1 can be caused by mutations in cytokine receptor-like factor 1 (CRLF1). However, the physiopathological role of CRLF1 is still poorly understood. A subset of CS/CISS1 cases remain yet genetically unexplained after CRLF1 sequencing. In five of them, exome sequencing and targeted Sanger sequencing identified four homozygous disease-causing mutations in kelch-like family member 7 (KLHL7), affecting the Kelch domains of the protein. KLHL7 encodes a BTB-Kelch-related protein involved in the ubiquitination of target proteins for proteasome-mediated degradation. Mono-allelic substitutions in other domains of KLHL7 have been reported in three families affected by a late-onset form of autosomal-dominant retinitis pigmentosa. Retinitis pigmentosa was also present in two surviving children reported here carrying bi-allelic KLHL7 mutations. KLHL7 mutations are thus associated with a more severe phenotype in recessive than in dominant cases. Although these data further support the pathogenic role of KLHL7 mutations in a CS/CISS1-like phenotype, they do not explain all their clinical manifestations and highlight the high phenotypic heterogeneity associated with mutations in KLHL7.


Asunto(s)
Alelos , Autoantígenos/genética , Deformidades Congénitas de la Mano/complicaciones , Deformidades Congénitas de la Mano/genética , Hiperhidrosis/complicaciones , Hiperhidrosis/genética , Mutación , Retinitis Pigmentosa/complicaciones , Retinitis Pigmentosa/genética , Trismo/congénito , Secuencia de Aminoácidos , Autoantígenos/química , Niño , Preescolar , Muerte Súbita , Facies , Femenino , Humanos , Lactante , Masculino , Modelos Moleculares , Linaje , Fenotipo , Síndrome , Trismo/complicaciones , Trismo/genética
3.
Dev Biol ; 416(1): 200-211, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27212026

RESUMEN

FOXL2 belongs to the evolutionarily conserved forkhead box (FOX) superfamily and is a master transcription factor in a spectrum of developmental pathways, including ovarian and eyelid development and bone, cartilage and uterine maturation. To analyse its action, we searched for proteins that interact with FOXL2. We found that FOXL2 interacts with specific C-terminal propeptides of several fibrillary collagens. Because these propeptides can participate in feedback regulation of collagen biosynthesis, we inferred that FOXL2 could thereby affect the transcription of the cognate collagen genes. Focusing on COL1A2, we found that FOXL2 indeed affects collagen synthesis, by binding to a DNA response element located about 65Kb upstream of this gene. According to our hypothesis we found that in Foxl2(-/-) mouse ovaries, Col1a2 was elevated from birth to adulthood. The extracellular matrix (ECM) compartmentalizes the ovary during folliculogenesis, (with type I, type III and type IV collagens as primary components), and ECM composition changes during the reproductive lifespan. In Foxl2(-/-) mouse ovaries, in addition to up-regulation of Col1a2, Col3a1, Col4a1 and fibronectin were also upregulated, while laminin expression was reduced. Thus, by regulating levels of extracellular matrix components, FOXL2 may contribute to both ovarian histogenesis and the fibrosis attendant on depletion of the follicle reserve during reproductive aging and menopause.


Asunto(s)
Colágeno Tipo I/genética , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Colágeno Tipo I/metabolismo , Secuencia de Consenso , Matriz Extracelular/metabolismo , Femenino , Proteína Forkhead Box L2 , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovario/metabolismo , Regiones Promotoras Genéticas , Unión Proteica
5.
BMC Dev Biol ; 15: 27, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26134413

RESUMEN

BACKGROUND: Haploinsufficiency of the FOXL2 transcription factor in humans causes Blepharophimosis/Ptosis/Epicanthus Inversus syndrome (BPES), characterized by eyelid anomalies and premature ovarian failure. Mice lacking Foxl2 recapitulate human eyelid/forehead defects and undergo female gonadal dysgenesis. We report here that mice lacking Foxl2 also show defects in postnatal growth and embryonic bone and cartilage formation. METHODS: Foxl2 (-/-) male mice at different stages of development have been characterized and compared to wild type. Body length and weight were measured and growth curves were created. Skeletons were stained with alcian blue and/or alizarin red. Bone and cartilage formation was analyzed by Von Kossa staining and immunofluorescence using anti-FOXL2 and anti-SOX9 antibodies followed by confocal microscopy. Genes differentially expressed in skull vaults were evaluated by microarray analysis. Analysis of the GH/IGF1 pathway was done evaluating the expression of several hypothalamic-pituitary-bone axis markers by RT-qPCR. RESULTS: Compared to wild-type, Foxl2 null mice are smaller and show skeletal abnormalities and defects in cartilage and bone mineralization, with down-regulation of the GH/IGF1 axis. Consistent with these effects, we find FOXL2 expressed in embryos at 9.5 dpc in neural tube epithelium, in head mesenchyme near the neural tube, and within the first branchial arch; then, starting at 12.5 dpc, expressed in cartilaginous tissue; and at PO and P7, in hypothalamus. CONCLUSIONS: Our results support FOXL2 as a master transcription factor in a spectrum of developmental processes, including growth, cartilage and bone formation. Its action overlaps that of SOX9, though they are antagonistic in female vs male gonadal sex determination but conjoint in cartilage and skeletal development.


Asunto(s)
Desarrollo Óseo , Cartílago/crecimiento & desarrollo , Factores de Transcripción Forkhead/metabolismo , Transducción de Señal , Animales , Blefarofimosis/metabolismo , Cartílago/metabolismo , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Anomalías Cutáneas/metabolismo , Anomalías Urogenitales/metabolismo
6.
Proc Natl Acad Sci U S A ; 105(5): 1620-5, 2008 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-18245381

RESUMEN

beta-Thalassemia and sickle cell disease both display a great deal of phenotypic heterogeneity, despite being generally thought of as simple Mendelian diseases. The reasons for this are not well understood, although the level of fetal hemoglobin (HbF) is one well characterized ameliorating factor in both of these conditions. To better understand the genetic basis of this heterogeneity, we carried out genome-wide scans with 362,129 common SNPs on 4,305 Sardinians to look for genetic linkage and association with HbF levels, as well as other red blood cell-related traits. Among major variants affecting HbF levels, SNP rs11886868 in the BCL11A gene was strongly associated with this trait (P < 10(-35)). The C allele frequency was significantly higher in Sardinian individuals with elevated HbF levels, detected by screening for beta-thalassemia, and patients with attenuated forms of beta-thalassemia vs. those with thalassemia major. We also show that the same BCL11A variant is strongly associated with HbF levels in a large cohort of sickle cell patients. These results indicate that BCL11A variants, by modulating HbF levels, act as an important ameliorating factor of the beta-thalassemia phenotype, and it is likely they could help ameliorate other hemoglobin disorders. We expect our findings will help to characterize the molecular mechanisms of fetal globin regulation and could eventually contribute to the development of new therapeutic approaches for beta-thalassemia and sickle cell anemia.


Asunto(s)
Proteínas Portadoras/genética , Hemoglobina Fetal/análisis , Hemoglobina Fetal/metabolismo , Ligamiento Genético , Proteínas Nucleares/genética , Talasemia beta/diagnóstico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Frecuencia de los Genes , Genoma Humano , Humanos , Italia , Masculino , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple , Proteínas Represoras
7.
PLoS Genet ; 2(8): e132, 2006 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-16934002

RESUMEN

In family studies, phenotypic similarities between relatives yield information on the overall contribution of genes to trait variation. Large samples are important for these family studies, especially when comparing heritability between subgroups such as young and old, or males and females. We recruited a cohort of 6,148 participants, aged 14-102 y, from four clustered towns in Sardinia. The cohort includes 34,469 relative pairs. To extract genetic information, we implemented software for variance components heritability analysis, designed to handle large pedigrees, analyze multiple traits simultaneously, and model heterogeneity. Here, we report heritability analyses for 98 quantitative traits, focusing on facets of personality and cardiovascular function. We also summarize results of bivariate analyses for all pairs of traits and of heterogeneity analyses for each trait. We found a significant genetic component for every trait. On average, genetic effects explained 40% of the variance for 38 blood tests, 51% for five anthropometric measures, 25% for 20 measures of cardiovascular function, and 19% for 35 personality traits. Four traits showed significant evidence for an X-linked component. Bivariate analyses suggested overlapping genetic determinants for many traits, including multiple personality facets and several traits related to the metabolic syndrome; but we found no evidence for shared genetic determinants that might underlie the reported association of some personality traits and cardiovascular risk factors. Models allowing for heterogeneity suggested that, in this cohort, the genetic variance was typically larger in females and in younger individuals, but interesting exceptions were observed. For example, narrow heritability of blood pressure was approximately 26% in individuals more than 42 y old, but only approximately 8% in younger individuals. Despite the heterogeneity in effect sizes, the same loci appear to contribute to variance in young and old, and in males and females. In summary, we find significant evidence for heritability of many medically important traits, including cardiovascular function and personality. Evidence for heterogeneity by age and sex suggests that models allowing for these differences will be important in mapping quantitative traits.


Asunto(s)
Enfermedades Cardiovasculares/genética , Fenómenos Fisiológicos Cardiovasculares , Personalidad/genética , Carácter Cuantitativo Heredable , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento , Análisis de Varianza , Cromosomas Humanos X/genética , Estudios de Cohortes , Femenino , Genes Mitocondriales , Humanos , Italia , Masculino , Persona de Mediana Edad , Modelos Genéticos , Herencia Multifactorial , Caracteres Sexuales , Hermanos
8.
Eur J Hum Genet ; 19(5): 525-33, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21326283

RESUMEN

Crisponi syndrome (CS) and cold-induced sweating syndrome type 1 (CISS1) are disorders caused by mutations in CRLF1. The two syndromes share clinical characteristics, such as dysmorphic features, muscle contractions, scoliosis and cold-induced sweating, with CS patients showing a severe clinical course in infancy involving hyperthermia, associated with death in most cases in the first years of life. To evaluate a potential genotype/phenotype correlation and whether CS and CISS1 represent two allelic diseases or manifestations at different ages of the same disorder, we carried out a detailed clinical analysis of 19 patients carrying mutations in CRLF1. We studied the functional significance of the mutations found in CRLF1, providing evidence that phenotypic severity of the two disorders mainly depends on altered kinetics of secretion of the mutated CRLF1 protein. On the basis of these findings, we believe that the two syndromes, CS and CISS1, represent manifestations of the same disorder, with different degrees of severity. We suggest renaming the two genetic entities CS and CISS1 with the broader term of Sohar-Crisponi syndrome.


Asunto(s)
Receptores de Citocinas/genética , Adolescente , Secuencia de Aminoácidos , Animales , Células COS , Niño , Preescolar , Chlorocebus aethiops , Análisis Mutacional de ADN , Muerte Súbita , Facies , Femenino , Fiebre/genética , Deformidades Congénitas de la Mano/genética , Humanos , Hiperhidrosis , Lactante , Masculino , Contracción Muscular/genética , Receptores de Citocinas/química , Alineación de Secuencia , Terminología como Asunto , Trismo/congénito , Trismo/genética
9.
PLoS One ; 5(3): e9477, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20209145

RESUMEN

The FOXL2 forkhead transcription factor is expressed in ovarian granulosa cells, and mutated FOXL2 causes the blepharophimosis, ptosis and epicanthus inversus syndrome (BPES) and predisposes to premature ovarian failure. Inactivation of Foxl2 in mice demonstrated its indispensability for female gonadal sex determination and ovary development and revealed its antagonism of Sox9, the effector of male testis development. To help to define the regulatory activities of FOXL2, we looked for interacting proteins. Based on yeast two-hybrid screening, we found that FOXL2 interacts with PIAS1 and UBC9, both parts of the sumoylation machinery. We showed that human FOXL2 is sumoylated in transfected cell lines, and that endogenous mouse Foxl2 is comparably sumoylated. This modification changes its cellular localization, stability and transcriptional activity. It is intriguing that similar sumoylation and regulatory consequences have also been reported for SOX9, the male counterpart of FOXL2 in somatic gonadal tissues.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Animales , Células COS , Chlorocebus aethiops , Proteína Forkhead Box L2 , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteína SUMO-1 , Enzimas Ubiquitina-Conjugadoras/metabolismo
10.
Genomics ; 83(5): 757-64, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15081106

RESUMEN

A translocation breakpoint 171 kb 5' of the transcription start of FOXL2 causes blepharophimosis/ptosis/epicanthus inversus syndrome (BPES) and associated premature ovarian failure. The breakpoint falls within another gene, MRPS22, that has been sequenced in 500 kb of continuous DNA. MRPS22 encodes 20 exons and a number of alternative transcripts. Three CpG islands (>91% identical) are followed by noncoding exons 4-12 and coding exons 13-20. The 3'UTR extends into the 3'UTR of COPB2. Based on the sequence, three reported translocations that cause BPES all fall within intron 6 of MRPS22. Comparisons reveal conserved segments in introns 6, 11, and 12 of human and mouse. Notably intron 11 sequence is also deleted in goat PIS syndrome (which combines craniofacial defects, female infertility, and XX sex reversal). The conserved sequences are candidates for models in which they are distant enhancers or otherwise affect higher order chromatin structure to impose long-range cis regulation of FOXL2 expression.


Asunto(s)
Rotura Cromosómica/genética , Cromosomas Humanos Par 3/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Transcripción/genética , Translocación Genética/genética , Animales , Secuencia de Bases , Proteína Coatómero/genética , Secuencia Conservada/genética , Islas de CpG/genética , Exones/genética , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead , Genómica , Cabras/genética , Humanos , Intrones/genética , Masculino , Ratones , Proteínas Mitocondriales/genética , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Ribosómicas/genética , Testículo/metabolismo
11.
Dev Biol ; 243(1): 185-206, 2002 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11846487

RESUMEN

The type 1 Simpson-Golabi-Behmel overgrowth syndrome (SGBS1) is caused by loss-of-function mutations of the X-linked GPC3 gene encoding glypican-3, a cell-surface heparan sulfate proteoglycan that apparently plays a negative role in growth control by an unknown mechanism. Mice carrying a Gpc3 gene knockout exhibited several phenotypic features that resemble clinical hallmarks of SGBS1, including somatic overgrowth, renal dysplasia, accessory spleens, polydactyly, and placentomegaly. In Gpc3/DeltaH19 double mutants (lacking GPC3 and also carrying a deletion around the H19 gene region that causes bialellic expression of the closely linked Igf2 gene by imprint relaxation), the Gpc3-null phenotype was exacerbated, while additional SGBS1 features (omphalocele and skeletal defects) were manifested. However, results from a detailed comparative analysis of growth patterns in double mutants lacking GPC3 and also IGF2, IGF1, or the type 1 IGF receptor (IGF1R) provided conclusive genetic evidence inconsistent with the hypothesis that GPC3 acts as a growth suppressor by sequestering or downregulating an IGF ligand. Nevertheless, our data are compatible with a model positing that there is downstream convergence of the independent signaling pathways in which either IGFs or (indirectly) GPC3 participate.


Asunto(s)
Anomalías Múltiples/genética , Proteoglicanos de Heparán Sulfato/genética , Factor II del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/genética , Receptor IGF Tipo 1/genética , Animales , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Regulación de la Expresión Génica , Glipicanos , Masculino , Ratones , Ratones Noqueados , Transducción de Señal/genética
12.
Hum Mol Genet ; 13(11): 1171-81, 2004 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15056605

RESUMEN

FOXL2 mutations cause gonadal dysgenesis or premature ovarian failure (POF) in women, as well as eyelid/forehead dysmorphology in both sexes (the 'blepharophimosis-ptosis-epicanthus inversus syndrome', BPES). Here we report that mice lacking Foxl2 recapitulate relevant features of human BPES: males and females are small and show distinctive craniofacial morphology with upper eyelids absent. Furthermore, in mice as in humans, sterility is confined to females. Features of Foxl2 null animals point toward a new mechanism of POF, with all major somatic cell lineages failing to develop around growing oocytes from the time of primordial follicle formation. Foxl2 disruption thus provides a model for histogenesis and reproductive competence of the ovary.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Ovario/patología , Insuficiencia Ovárica Primaria/etiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Blefarofimosis/etiología , Blefarofimosis/genética , Modelos Animales de Enfermedad , Embrión de Mamíferos/patología , Párpados/anomalías , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead , Eliminación de Gen , Inmunoquímica , Antígeno Ki-67/inmunología , Masculino , Ratones , Ratones Noqueados , Ovario/embriología , Ovario/crecimiento & desarrollo , Insuficiencia Ovárica Primaria/genética , Insuficiencia Ovárica Primaria/patología , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA