Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genome Res ; 27(5): 677-685, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27895111

RESUMEN

In an effort to more fully understand the full spectrum of human genetic variation, we generated deep single-molecule, real-time (SMRT) sequencing data from two haploid human genomes. By using an assembly-based approach (SMRT-SV), we systematically assessed each genome independently for structural variants (SVs) and indels resolving the sequence structure of 461,553 genetic variants from 2 bp to 28 kbp in length. We find that >89% of these variants have been missed as part of analysis of the 1000 Genomes Project even after adjusting for more common variants (MAF > 1%). We estimate that this theoretical human diploid differs by as much as ∼16 Mbp with respect to the human reference, with long-read sequencing data providing a fivefold increase in sensitivity for genetic variants ranging in size from 7 bp to 1 kbp compared with short-read sequence data. Although a large fraction of genetic variants were not detected by short-read approaches, once the alternate allele is sequence-resolved, we show that 61% of SVs can be genotyped in short-read sequence data sets with high accuracy. Uncoupling discovery from genotyping thus allows for the majority of this missed common variation to be genotyped in the human population. Interestingly, when we repeat SV detection on a pseudodiploid genome constructed in silico by merging the two haploids, we find that ∼59% of the heterozygous SVs are no longer detected by SMRT-SV. These results indicate that haploid resolution of long-read sequencing data will significantly increase sensitivity of SV detection.


Asunto(s)
Mapeo Contig/métodos , Genoma Humano , Variación Estructural del Genoma , Haploidia , Análisis de Secuencia de ADN/métodos , Mapeo Contig/normas , Proyecto Genoma Humano , Humanos , Análisis de Secuencia de ADN/normas
2.
Nature ; 515(7526): 216-21, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25363768

RESUMEN

Whole exome sequencing has proven to be a powerful tool for understanding the genetic architecture of human disease. Here we apply it to more than 2,500 simplex families, each having a child with an autistic spectrum disorder. By comparing affected to unaffected siblings, we show that 13% of de novo missense mutations and 43% of de novo likely gene-disrupting (LGD) mutations contribute to 12% and 9% of diagnoses, respectively. Including copy number variants, coding de novo mutations contribute to about 30% of all simplex and 45% of female diagnoses. Almost all LGD mutations occur opposite wild-type alleles. LGD targets in affected females significantly overlap the targets in males of lower intelligence quotient (IQ), but neither overlaps significantly with targets in males of higher IQ. We estimate that LGD mutation in about 400 genes can contribute to the joint class of affected females and males of lower IQ, with an overlapping and similar number of genes vulnerable to contributory missense mutation. LGD targets in the joint class overlap with published targets for intellectual disability and schizophrenia, and are enriched for chromatin modifiers, FMRP-associated genes and embryonically expressed genes. Most of the significance for the latter comes from affected females.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Predisposición Genética a la Enfermedad/genética , Mutación/genética , Sistemas de Lectura Abierta/genética , Niño , Análisis por Conglomerados , Exoma/genética , Femenino , Genes , Humanos , Pruebas de Inteligencia , Masculino , Reproducibilidad de los Resultados
3.
Am J Hum Genet ; 98(3): 541-552, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26942287

RESUMEN

Intellectual disability (ID) and autism spectrum disorders (ASD) are genetically heterogeneous, and a significant number of genes have been associated with both conditions. A few mutations in POGZ have been reported in recent exome studies; however, these studies do not provide detailed clinical information. We collected the clinical and molecular data of 25 individuals with disruptive mutations in POGZ by diagnostic whole-exome, whole-genome, or targeted sequencing of 5,223 individuals with neurodevelopmental disorders (ID primarily) or by targeted resequencing of this locus in 12,041 individuals with ASD and/or ID. The rarity of disruptive mutations among unaffected individuals (2/49,401) highlights the significance (p = 4.19 × 10(-13); odds ratio = 35.8) and penetrance (65.9%) of this genetic subtype with respect to ASD and ID. By studying the entire cohort, we defined common phenotypic features of POGZ individuals, including variable levels of developmental delay (DD) and more severe speech and language delay in comparison to the severity of motor delay and coordination issues. We also identified significant associations with vision problems, microcephaly, hyperactivity, a tendency to obesity, and feeding difficulties. Some features might be explained by the high expression of POGZ, particularly in the cerebellum and pituitary, early in fetal brain development. We conducted parallel studies in Drosophila by inducing conditional knockdown of the POGZ ortholog row, further confirming that dosage of POGZ, specifically in neurons, is essential for normal learning in a habituation paradigm. Combined, the data underscore the pathogenicity of loss-of-function mutations in POGZ and define a POGZ-related phenotype enriched in specific features.


Asunto(s)
Trastorno del Espectro Autista/genética , Discapacidad Intelectual/genética , Transposasas/genética , Adolescente , Adulto , Animales , Trastorno del Espectro Autista/diagnóstico , Niño , Preescolar , Estudios de Cohortes , Regulación hacia Abajo , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Exoma , Femenino , Técnicas de Silenciamiento del Gen , Estudio de Asociación del Genoma Completo , Humanos , Lactante , Discapacidad Intelectual/diagnóstico , Trastornos del Desarrollo del Lenguaje/diagnóstico , Trastornos del Desarrollo del Lenguaje/genética , Modelos Lineales , Masculino , Microcefalia/diagnóstico , Microcefalia/genética , Mutación , Fenotipo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Nature ; 499(7459): 471-5, 2013 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-23823723

RESUMEN

Most great ape genetic variation remains uncharacterized; however, its study is critical for understanding population history, recombination, selection and susceptibility to disease. Here we sequence to high coverage a total of 79 wild- and captive-born individuals representing all six great ape species and seven subspecies and report 88.8 million single nucleotide polymorphisms. Our analysis provides support for genetically distinct populations within each species, signals of gene flow, and the split of common chimpanzees into two distinct groups: Nigeria-Cameroon/western and central/eastern populations. We find extensive inbreeding in almost all wild populations, with eastern gorillas being the most extreme. Inferred effective population sizes have varied radically over time in different lineages and this appears to have a profound effect on the genetic diversity at, or close to, genes in almost all species. We discover and assign 1,982 loss-of-function variants throughout the human and great ape lineages, determining that the rate of gene loss has not been different in the human branch compared to other internal branches in the great ape phylogeny. This comprehensive catalogue of great ape genome diversity provides a framework for understanding evolution and a resource for more effective management of wild and captive great ape populations.


Asunto(s)
Variación Genética , Hominidae/genética , África , Animales , Animales Salvajes/genética , Animales de Zoológico/genética , Asia Sudoriental , Evolución Molecular , Flujo Génico/genética , Genética de Población , Genoma/genética , Gorilla gorilla/clasificación , Gorilla gorilla/genética , Hominidae/clasificación , Humanos , Endogamia , Pan paniscus/clasificación , Pan paniscus/genética , Pan troglodytes/clasificación , Pan troglodytes/genética , Filogenia , Polimorfismo de Nucleótido Simple/genética , Densidad de Población
5.
Nature ; 485(7397): 246-50, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22495309

RESUMEN

It is well established that autism spectrum disorders (ASD) have a strong genetic component; however, for at least 70% of cases, the underlying genetic cause is unknown. Under the hypothesis that de novo mutations underlie a substantial fraction of the risk for developing ASD in families with no previous history of ASD or related phenotypes--so-called sporadic or simplex families--we sequenced all coding regions of the genome (the exome) for parent-child trios exhibiting sporadic ASD, including 189 new trios and 20 that were previously reported. Additionally, we also sequenced the exomes of 50 unaffected siblings corresponding to these new (n = 31) and previously reported trios (n = 19), for a total of 677 individual exomes from 209 families. Here we show that de novo point mutations are overwhelmingly paternal in origin (4:1 bias) and positively correlated with paternal age, consistent with the modest increased risk for children of older fathers to develop ASD. Moreover, 39% (49 of 126) of the most severe or disruptive de novo mutations map to a highly interconnected ß-catenin/chromatin remodelling protein network ranked significantly for autism candidate genes. In proband exomes, recurrent protein-altering mutations were observed in two genes: CHD8 and NTNG1. Mutation screening of six candidate genes in 1,703 ASD probands identified additional de novo, protein-altering mutations in GRIN2B, LAMC3 and SCN1A. Combined with copy number variant (CNV) data, these results indicate extreme locus heterogeneity but also provide a target for future discovery, diagnostics and therapeutics.


Asunto(s)
Trastorno Autístico/genética , Exoma/genética , Exones/genética , Mutación Puntual/genética , Mapas de Interacción de Proteínas/genética , Proteínas de Unión al ADN/genética , Proteínas Ligadas a GPI/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Laminina/genética , Canal de Sodio Activado por Voltaje NAV1.1 , Proteínas del Tejido Nervioso/genética , Netrinas , Padres , Receptores de N-Metil-D-Aspartato/genética , Reproducibilidad de los Resultados , Hermanos , Transducción de Señal , Canales de Sodio/genética , Procesos Estocásticos , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/metabolismo , beta Catenina/metabolismo
6.
Am J Hum Genet ; 93(4): 595-606, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-24035194

RESUMEN

We searched for disruptive, genic rare copy-number variants (CNVs) among 411 families affected by sporadic autism spectrum disorder (ASD) from the Simons Simplex Collection by using available exome sequence data and CoNIFER (Copy Number Inference from Exome Reads). Compared to high-density SNP microarrays, our approach yielded ∼2× more smaller genic rare CNVs. We found that affected probands inherited more CNVs than did their siblings (453 versus 394, p = 0.004; odds ratio [OR] = 1.19) and that the probands' CNVs affected more genes (921 versus 726, p = 0.02; OR = 1.30). These smaller CNVs (median size 18 kb) were transmitted preferentially from the mother (136 maternal versus 100 paternal, p = 0.02), although this bias occurred irrespective of affected status. The excess burden of inherited CNVs among probands was driven primarily by sibling pairs with discordant social-behavior phenotypes (p < 0.0002, measured by Social Responsiveness Scale [SRS] score), which contrasts with families where the phenotypes were more closely matched or less extreme (p > 0.5). Finally, we found enrichment of brain-expressed genes unique to probands, especially in the SRS-discordant group (p = 0.0035). In a combined model, our inherited CNVs, de novo CNVs, and de novo single-nucleotide variants all independently contributed to the risk of autism (p < 0.05). Taken together, these results suggest that small transmitted rare CNVs play a role in the etiology of simplex autism. Importantly, the small size of these variants aids in the identification of specific genes as additional risk factors associated with ASD.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Variaciones en el Número de Copia de ADN , Desequilibrio de Ligamiento , Niño , Exoma , Femenino , Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Masculino , Fenotipo , Polimorfismo de Nucleótido Simple , Riesgo , Factores de Riesgo , Hermanos
7.
Genome Res ; 23(11): 1763-73, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24077392

RESUMEN

Ape chromosomes homologous to human chromosomes 14 and 15 were generated by a fission event of an ancestral submetacentric chromosome, where the two chromosomes were joined head-to-tail. The hominoid ancestral chromosome most closely resembles the macaque chromosome 7. In this work, we provide insights into the evolution of human chromosomes 14 and 15, performing a comparative study between macaque boundary region 14/15 and the orthologous human regions. We construct a 1.6-Mb contig of macaque BAC clones in the region orthologous to the ancestral hominoid fission site and use it to define the structural changes that occurred on human 14q pericentromeric and 15q subtelomeric regions. We characterize the novel euchromatin-heterochromatin transition region (∼20 Mb) acquired during the neocentromere establishment on chromosome 14, and find it was mainly derived through pericentromeric duplications from ancestral hominoid chromosomes homologous to human 2q14-qter and 10. Further, we show a relationship between evolutionary hotspots and low-copy repeat loci for chromosome 15, revealing a possible role of segmental duplications not only in mediating but also in "stitching" together rearrangement breakpoints.


Asunto(s)
Cromosomas Humanos Par 14/genética , Cromosomas Humanos Par 15/genética , Cromosomas de los Mamíferos/genética , Evolución Molecular , Hominidae/genética , Duplicaciones Segmentarias en el Genoma , Animales , Puntos de Rotura del Cromosoma , Duplicación Cromosómica , Cromosomas Artificiales Bacterianos , Clonación Molecular , Eucromatina/genética , Heterocromatina/genética , Humanos , Datos de Secuencia Molecular , Filogenia
10.
Genome Res ; 22(6): 1036-49, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22419167

RESUMEN

Chimpanzee and gorilla chromosomes differ from human chromosomes by the presence of large blocks of subterminal heterochromatin thought to be composed primarily of arrays of tandem satellite sequence. We explore their sequence composition and organization and show a complex organization composed of specific sets of segmental duplications that have hyperexpanded in concert with the formation of subterminal satellites. These regions are highly copy number polymorphic between and within species, and copy number differences involving hundreds of copies can be accurately estimated by assaying read-depth of next-generation sequencing data sets. Phylogenetic and comparative genomic analyses suggest that the structures have arisen largely independently in the two lineages with the exception of a few seed sequences present in the common ancestor of humans and African apes. We propose a model where an ancestral human-chimpanzee pericentric inversion and the ancestral chromosome 2 fusion both predisposed and protected the chimpanzee and human genomes, respectively, to the formation of subtelomeric heterochromatin. Our findings highlight the complex interplay between duplicated sequences and chromosomal rearrangements that rapidly alter the cytogenetic landscape in a short period of evolutionary time.


Asunto(s)
Cromosomas Humanos Par 2 , Evolución Molecular , Heterocromatina/genética , Hominidae/genética , Modelos Genéticos , Secuencia de Aminoácidos , Animales , Análisis Citogenético , ADN Satélite , Duplicación de Gen , Gorilla gorilla/genética , Humanos , Datos de Secuencia Molecular , Pan troglodytes/genética , Filogenia , Telómero/genética
11.
Am J Hum Genet ; 88(3): 317-32, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21397061

RESUMEN

Copy-number variants (CNVs) can reach appreciable frequencies in the human population, and recent discoveries have shown that several of these copy-number polymorphisms (CNPs) are associated with human diseases, including lupus, psoriasis, Crohn disease, and obesity. Despite new advances, significant biases remain in terms of CNP discovery and genotyping. We developed a method based on single-channel intensity data and benchmarked against copy numbers determined from sequencing read depth to successfully obtain CNP genotypes for 1495 CNPs from 487 human DNA samples of diverse ethnic backgrounds. This microarray contained CNPs in segmental duplication-rich regions and insertions of sequences not represented in the reference genome assembly or on standard SNP microarray platforms. We observe that CNPs in segmental duplications are more likely to be population differentiated than CNPs in unique regions (p = 0.015) and that biallelic CNPs show greater stratification when compared to frequency-matched SNPs (p = 0.0026). Although biallelic CNPs show a strong correlation of copy number with flanking SNP genotypes, the majority of multicopy CNPs do not (40% with r > 0.8). We selected a subset of CNPs for further characterization in 1876 additional samples from 62 populations; this revealed striking population-differentiated structural variants in genes of clinical significance such as OCLN, a tight junction protein involved in hepatitis C viral entry. Our microarray design allows these variants to be rapidly tested for disease association and our results suggest that CNPs (especially those that cannot be imputed from SNP genotypes) might have contributed disproportionately to human diversity and selection.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Genética de Población , Hibridación Genómica Comparativa , Sitios Genéticos/genética , Genotipo , Geografía , Humanos , Desequilibrio de Ligamiento/genética , Mutagénesis Insercional/genética , Polimorfismo de Nucleótido Simple/genética
12.
Nat Methods ; 9(2): 176-8, 2011 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-22179552

RESUMEN

We report an algorithm to detect structural variation and indels from 1 base pair (bp) to 1 Mbp within exome sequence data sets. Splitread uses one end-anchored placements to cluster the mappings of subsequences of unanchored ends to identify the size, content and location of variants with high specificity and sensitivity. The algorithm discovers indels, structural variants, de novo events and copy number-polymorphic processed pseudogenes missed by other methods.


Asunto(s)
Exoma , Algoritmos , Seudogenes
13.
PLoS Genet ; 7(11): e1002334, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22102821

RESUMEN

While numerous studies have implicated copy number variants (CNVs) in a range of neurological phenotypes, the impact relative to disease severity has been difficult to ascertain due to small sample sizes, lack of phenotypic details, and heterogeneity in platforms used for discovery. Using a customized microarray enriched for genomic hotspots, we assayed for large CNVs among 1,227 individuals with various neurological deficits including dyslexia (376), sporadic autism (350), and intellectual disability (ID) (501), as well as 337 controls. We show that the frequency of large CNVs (>1 Mbp) is significantly greater for ID-associated phenotypes compared to autism (p = 9.58 × 10(-11), odds ratio = 4.59), dyslexia (p = 3.81 × 10(-18), odds ratio = 14.45), or controls (p = 2.75 × 10(-17), odds ratio = 13.71). There is a striking difference in the frequency of rare CNVs (>50 kbp) in autism (10%, p = 2.4 × 10(-6), odds ratio = 6) or ID (16%, p = 3.55 × 10(-12), odds ratio = 10) compared to dyslexia (2%) with essentially no difference in large CNV burden among dyslexia patients compared to controls. Rare CNVs were more likely to arise de novo (64%) in ID when compared to autism (40%) or dyslexia (0%). We observed a significantly increased large CNV burden in individuals with ID and multiple congenital anomalies (MCA) compared to ID alone (p = 0.001, odds ratio = 2.54). Our data suggest that large CNV burden positively correlates with the severity of childhood disability: ID with MCA being most severely affected and dyslexics being indistinguishable from controls. When autism without ID was considered separately, the increase in CNV burden was modest compared to controls (p = 0.07, odds ratio = 2.33).


Asunto(s)
Trastorno Autístico/genética , Variaciones en el Número de Copia de ADN/genética , Dislexia/genética , Discapacidad Intelectual/genética , Neurogénesis/inmunología , Adolescente , Trastorno Autístico/diagnóstico , Trastorno Autístico/patología , Niño , Hibridación Genómica Comparativa/métodos , Proteínas del Citoesqueleto , Dislexia/diagnóstico , Dislexia/patología , Endopeptidasas/genética , Femenino , Factores de Transcripción Forkhead/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/patología , Masculino , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/patología , Neurogénesis/genética , Fenotipo , Proteínas/genética , Proteínas Represoras/genética , Eliminación de Secuencia/genética , Factores de Transcripción
14.
Bone ; 168: 116654, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36584785

RESUMEN

OBJECTIVES: To estimate the incidence of clinical fragility fractures in postmenopausal women with rheumatoid arthritis (RA) and analyze risk factors for fracture. METHODS: Incidence of clinical fragility fractures in 330 postmenopausal women with RA was compared to that of a control population of 660 age-matched postmenopausal Spanish women. Clinical fractures during the previous five years were recorded. We analyzed associations with risk factors for fracture in both populations and with disease-related variables in RA patients. RESULTS: Median age of RA patients was 64 years; median RA duration was eight years. Sixty-nine percent were in remission or on low activity. Eighty-five percent had received glucocorticoids (GCs); 85 %, methotrexate; and 40 %, ≥1 biologic DMARD. Fifty-four patients and 47 controls had ≥1 major osteoporotic fracture (MOF). Incidence of MOFs was 3.55 per 100 patient-year in patients and 0.72 in controls (HR: 2.6). Risk factors for MOFs in RA patients were age, previous fracture, parental hip fracture, years since menopause, BMD, erosions, disease activity and disability, and cumulative dose of GCs. Previous fracture in RA patients was a strong risk for MOFs (HR: 10.37). CONCLUSION: Of every 100 postmenopausal Spanish women with RA, 3-4 have a MOF per year. This is more than double that of the general population. A previous fracture poses a high risk for a new fracture. Other classic risk factors for fracture, RA disease activity and disability, and the cumulative dose of GCs are associated with fracture development.


Asunto(s)
Artritis Reumatoide , Fracturas Osteoporóticas , Humanos , Femenino , Persona de Mediana Edad , Estudios de Casos y Controles , Posmenopausia , Incidencia , Artritis Reumatoide/complicaciones , Artritis Reumatoide/epidemiología , Fracturas Osteoporóticas/etiología , Factores de Riesgo , Densidad Ósea
15.
Proc Natl Acad Sci U S A ; 105(50): 19809-14, 2008 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-19060200

RESUMEN

Silencing of multiple cancer-related genes is associated with de novo methylation of linked CpG islands. Additionally, bivalent histone modification profiles characterized by the juxtaposition of active and inactive histone marks have been observed in genes that become hypermethylated in cancer. It is unknown how these ambiguous epigenetic states are maintained and how they interrelate with adjacent genomic regions with different epigenetic landscapes. Here, we present the analysis of a set of neighboring genes, including many frequently silenced in colon cancer cells, in a chromosomal region at 5q35.2 spanning 1.25 Mb. Promoter DNA methylation occurs only at genes maintained at a low transcriptional state and is characterized by the presence of bivalent histone marks, namely trimethylation of lysines 4 and 27 in histone 3. Chemically induced hyperacetylation and DNA demethylation lead to up-regulation of silenced genes in this locus yet do not resolve bivalent domains into a domain-wide active chromatin conformation. In contrast, active genes in the region become down-regulated after drug treatment, accompanied by a partial loss of chromatin domain boundaries and spreading of the inactive histone mark trimethylated lysine 27 in histone 3. Our results demonstrate that bivalent domains mark the promoters of genes that will become DNA methylated in adult tumor cells to enforce transcriptional silence. These bivalent domains not only remain upon drug induced gene reactivation, but also spread over adjacent CpG islands. These results may have important implications in understanding and managing epigenetic therapies of cancer.


Asunto(s)
Cromosomas Humanos Par 5/genética , Neoplasias del Colon/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Genes Relacionados con las Neoplasias , Secuencia de Bases , Transformación Celular Neoplásica/genética , Cromatina/metabolismo , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Metilación de ADN/efectos de los fármacos , Terapia Genética , Humanos , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Transcripción Genética
16.
RMD Open ; 7(3)2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34887346

RESUMEN

OBJECTIVES: To analyse the effect of targeted therapies, either biological (b) disease-modifying antirheumatic drugs (DMARDs), targeted synthetic (ts) DMARDs and other factors (demographics, comorbidities or COVID-19 symptoms) on the risk of COVID-19 related hospitalisation in patients with inflammatory rheumatic diseases. METHODS: The COVIDSER study is an observational cohort including 7782 patients with inflammatory rheumatic diseases. Multivariable logistic regression was used to estimate ORs and 95% CIs of hospitalisation. Antirheumatic medication taken immediately prior to infection, demographic characteristics, rheumatic disease diagnosis, comorbidities and COVID-19 symptoms were analysed. RESULTS: A total of 426 cases of symptomatic COVID-19 from 1 March 2020 to 13 April 2021 were included in the analyses: 106 (24.9%) were hospitalised and 19 (4.4%) died. In multivariate-adjusted models, bDMARDs and tsDMARDs in combination were not associated with hospitalisation compared with conventional synthetic DMARDs (OR 0.55, 95% CI 0.24 to 1.25 of b/tsDMARDs, p=0.15). Tumour necrosis factor inhibitors (TNF-i) were associated with a reduced likelihood of hospitalisation (OR 0.32, 95% CI 0.12 to 0.82, p=0.018), whereas rituximab showed a tendency to an increased risk of hospitalisation (OR 4.85, 95% CI 0.86 to 27.2). Glucocorticoid use was not associated with hospitalisation (OR 1.69, 95% CI 0.81 to 3.55). A mix of sociodemographic factors, comorbidities and COVID-19 symptoms contribute to patients' hospitalisation. CONCLUSIONS: The use of targeted therapies as a group is not associated with COVID-19 severity, except for rituximab, which shows a trend towards an increased risk of hospitalisation, while TNF-i was associated with decreased odds of hospitalisation in patients with rheumatic disease. Other factors like age, male gender, comorbidities and COVID-19 symptoms do play a role.


Asunto(s)
Antirreumáticos , COVID-19 , Enfermedades Reumáticas , Antirreumáticos/efectos adversos , Humanos , Masculino , Enfermedades Reumáticas/tratamiento farmacológico , Enfermedades Reumáticas/epidemiología , SARS-CoV-2 , Factores Sociodemográficos
17.
Nucleic Acids Res ; 36(3): 770-84, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18084025

RESUMEN

Methylation of the cytosine is the most frequent epigenetic modification of DNA in mammalian cells. In humans, most of the methylated cytosines are found in CpG-rich sequences within tandem and interspersed repeats that make up to 45% of the human genome, being Alu repeats the most common family. Demethylation of Alu elements occurs in aging and cancer processes and has been associated with gene reactivation and genomic instability. By targeting the unmethylated SmaI site within the Alu sequence as a surrogate marker, we have quantified and identified unmethylated Alu elements on the genomic scale. Normal colon epithelial cells contain in average 25 486 +/- 10 157 unmethylated Alu's per haploid genome, while in tumor cells this figure is 41 995 +/- 17 187 (P = 0.004). There is an inverse relationship in Alu families with respect to their age and methylation status: the youngest elements exhibit the highest prevalence of the SmaI site (AluY: 42%; AluS: 18%, AluJ: 5%) but the lower rates of unmethylation (AluY: 1.65%; AluS: 3.1%, AluJ: 12%). Data are consistent with a stronger silencing pressure on the youngest repetitive elements, which are closer to genes. Further insights into the functional implications of atypical unmethylation states in Alu elements will surely contribute to decipher genomic organization and gene regulation in complex organisms.


Asunto(s)
Elementos Alu , Carcinoma/genética , Neoplasias Colorrectales/genética , Metilación de ADN , Genómica/métodos , Línea Celular Tumoral , Cromosomas Humanos , Colon/citología , Biología Computacional , Islas de CpG , Epigénesis Genética , Genoma Humano , Humanos , Mucosa Intestinal/química , Reacción en Cadena de la Polimerasa
18.
Nat Genet ; 49(4): 515-526, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28191889

RESUMEN

Gene-disruptive mutations contribute to the biology of neurodevelopmental disorders (NDDs), but most of the related pathogenic genes are not known. We sequenced 208 candidate genes from >11,730 cases and >2,867 controls. We identified 91 genes, including 38 new NDD genes, with an excess of de novo mutations or private disruptive mutations in 5.7% of cases. Drosophila functional assays revealed a subset with increased involvement in NDDs. We identified 25 genes showing a bias for autism versus intellectual disability and highlighted a network associated with high-functioning autism (full-scale IQ >100). Clinical follow-up for NAA15, KMT5B, and ASH1L highlighted new syndromic and nonsyndromic forms of disease.


Asunto(s)
Trastorno Autístico/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Femenino , Humanos , Masculino , Mutación/genética , Fenotipo
19.
Nat Commun ; 7: 13316, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27824329

RESUMEN

Recurrent de novo (DN) and likely gene-disruptive (LGD) mutations contribute significantly to autism spectrum disorders (ASDs) but have been primarily investigated in European cohorts. Here, we sequence 189 risk genes in 1,543 Chinese ASD probands (1,045 from trios). We report an 11-fold increase in the odds of DN LGD mutations compared with expectation under an exome-wide neutral model of mutation. In aggregate, ∼4% of ASD patients carry a DN mutation in one of just 29 autism risk genes. The most prevalent gene for recurrent DN mutations is SCN2A (1.1% of patients) followed by CHD8, DSCAM, MECP2, POGZ, WDFY3 and ASH1L. We identify novel DN LGD recurrences (GIGYF2, MYT1L, CUL3, DOCK8 and ZNF292) and DN mutations in previous ASD candidates (ARHGAP32, NCOR1, PHIP, STXBP1, CDKL5 and SHANK1). Phenotypic follow-up confirms potential subtypes and highlights how large global cohorts might be leveraged to prove the pathogenic significance of individually rare mutations.


Asunto(s)
Trastorno del Espectro Autista/genética , Mutación/genética , Pueblo Asiatico/genética , Estudios de Cohortes , Análisis Mutacional de ADN , Exoma/genética , Predisposición Genética a la Enfermedad , Geografía , Humanos , Patrón de Herencia/genética , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo
20.
Nat Genet ; 47(6): 582-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25961944

RESUMEN

To assess the relative impact of inherited and de novo variants on autism risk, we generated a comprehensive set of exonic single-nucleotide variants (SNVs) and copy number variants (CNVs) from 2,377 families with autism. We find that private, inherited truncating SNVs in conserved genes are enriched in probands (odds ratio = 1.14, P = 0.0002) in comparison to unaffected siblings, an effect involving significant maternal transmission bias to sons. We also observe a bias for inherited CNVs, specifically for small (<100 kb), maternally inherited events (P = 0.01) that are enriched in CHD8 target genes (P = 7.4 × 10(-3)). Using a logistic regression model, we show that private truncating SNVs and rare, inherited CNVs are statistically independent risk factors for autism, with odds ratios of 1.11 (P = 0.0002) and 1.23 (P = 0.01), respectively. This analysis identifies a second class of candidate genes (for example, RIMS1, CUL7 and LZTR1) where transmitted mutations may create a sensitized background but are unlikely to be completely penetrant.


Asunto(s)
Trastorno Autístico/genética , Codón sin Sentido , Variaciones en el Número de Copia de ADN , Exoma , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Desequilibrio de Ligamiento , Masculino , Polimorfismo de Nucleótido Simple , Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA