Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Psychol Med ; 51(13): 2260-2273, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33634770

RESUMEN

Autism spectrum disorder (autism) is a heterogeneous group of neurodevelopmental conditions characterized by early childhood-onset impairments in communication and social interaction alongside restricted and repetitive behaviors and interests. This review summarizes recent developments in human genetics research in autism, complemented by epigenetic and transcriptomic findings. The clinical heterogeneity of autism is mirrored by a complex genetic architecture involving several types of common and rare variants, ranging from point mutations to large copy number variants, and either inherited or spontaneous (de novo). More than 100 risk genes have been implicated by rare, often de novo, potentially damaging mutations in highly constrained genes. These account for substantial individual risk but a small proportion of the population risk. In contrast, most of the genetic risk is attributable to common inherited variants acting en masse, each individually with small effects. Studies have identified a handful of robustly associated common variants. Different risk genes converge on the same mechanisms, such as gene regulation and synaptic connectivity. These mechanisms are also implicated by genes that are epigenetically and transcriptionally dysregulated in autism. Major challenges to understanding the biological mechanisms include substantial phenotypic heterogeneity, large locus heterogeneity, variable penetrance, and widespread pleiotropy. Considerable increases in sample sizes are needed to better understand the hundreds or thousands of common and rare genetic variants involved. Future research should integrate common and rare variant research, multi-omics data including genomics, epigenomics, and transcriptomics, and refined phenotype assessment with multidimensional and longitudinal measures.


Asunto(s)
Trastorno del Espectro Autista , Variaciones en el Número de Copia de ADN/genética , Genómica , Herencia Multifactorial/genética , Trastorno del Espectro Autista/epidemiología , Trastorno del Espectro Autista/genética , Humanos , Fenotipo , Factores de Riesgo , Transcriptoma/genética , Estudios en Gemelos como Asunto
2.
Transl Psychiatry ; 9(1): 214, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31477683

RESUMEN

Depression is a severe and debilitating mental disorder diagnosed by evaluation of affective, cognitive and physical depression symptoms. Severity of these symptoms strongly impacts individual's quality of life and is influenced by a combination of genetic and environmental factors. One of the molecular mechanisms allowing for an interplay between these factors is DNA methylation, an epigenetic modification playing a pivotal role in regulation of brain functioning across lifespan. The aim of this study was to investigate if there are DNA methylation signatures associated with depression symptomatology in order to identify molecular mechanisms contributing to pathophysiology of depression. We performed an epigenome-wide association study (EWAS) of continuous depression symptomatology score measured in a cohort of 724 monozygotic Danish twins (346 males, 378 females). Through EWAS analyses adjusted for sex, age, flow-cytometry based blood cell composition, and twin relatedness structure in the data we identified depression symptomatology score to be associated with blood DNA methylation levels in promoter regions of neuropsin (KLK8, p-value = 4.7 × 10-7) and DAZ associated protein 2 (DAZAP2, p-value = 3.13 × 10-8) genes. Other top associated probes were located in gene bodies of MAD1L1 (p-value = 5.16 × 10-6), SLC29A2 (p-value = 6.15 × 10-6) and AKT1 (p-value = 4.47 × 10-6), all genes associated before with development of depression. Additionally, the following three measures (a) DNAmAge (calculated with Horvath and Hannum epigenetic clock estimators) adjusted for chronological age, (b) difference between DNAmAge and chronological age, and (c) DNAmAge acceleration were not associated with depression symptomatology score in our cohort. In conclusion, our data suggests that depression symptomatology score is associated with DNA methylation levels of genes implicated in response to stress, depressive-like behaviors, and recurrent depression in patients, but not with global DNA methylation changes across the genome.


Asunto(s)
Depresión/genética , Epigénesis Genética , Epigenoma , Gemelos Monocigóticos/genética , Anciano , Metilación de ADN , Dinamarca , Femenino , Interacción Gen-Ambiente , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Persona de Mediana Edad , Calidad de Vida
3.
Transl Psychiatry ; 7(8): e1221, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28850114

RESUMEN

Individuals with 22q11.2 deletion syndrome (DS) have an increased risk of comorbid mental disorders including schizophrenia, attention deficit hyperactivity disorder, depression, as well as intellectual disability. Although most 22q11.2 deletion carriers have the long 3-Mb form of the hemizygous deletion, there remains a large variation in the development and progression of psychiatric disorders, which suggests that alternative factors contribute to the pathogenesis. In this study we investigated whether neonatal DNA methylation signatures in individuals with the 22q11.2 deletion associate with mental disorder later in life. DNA methylation was measured genome-wide from neonatal dried blood spots in a cohort of 164 individuals with 22q11.2DS, including 48 individuals diagnosed with a psychiatric disorder. Among several CpG sites with P-value<10-6, we identified cg23546855 (P-value=2.15 × 10-7) mapping to STK32C to be associated with a later psychiatric diagnosis. Pathway analysis of the top findings resulted in the identification of several Gene Ontology pathways to be significantly enriched (P-value<0.05 after Benjamini-Hochberg correction); among them are the following: neurogenesis, neuron development, neuron projection development, astrocyte development, axonogenesis and axon guidance. In addition, we identified differentially methylated CpG sites in LRP2BP (P-value=5.37 × 10-8) to be associated with intellectual disability (F70-79), in TOP1 (P-value=1.86 × 10-7) with behavioral disorders (F90-98), in NOSIP (P-value=5.12 × 10-8) with disorders of psychological development (F80-89) and in SEMA4B (P-value=4.02 × 10-7) with schizophrenia spectrum disorders (F20-29). In conclusion, our study suggests an association of DNA methylation differences at birth with development of mental disorder later in life in 22q11.2DS individuals.


Asunto(s)
Metilación de ADN , Síndrome de DiGeorge/complicaciones , Síndrome de DiGeorge/genética , Trastornos Mentales/complicaciones , Trastornos Mentales/genética , Adolescente , Niño , Estudios de Cohortes , Islas de CpG , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Fenotipo
4.
Neurobiol Aging ; 50: 60-63, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27889677

RESUMEN

The epigenetic clock, also known as DNA methylation age (DNAmAge), represents age-related changes of DNA methylation at multiple sites of the genome and is suggested to be a biomarker for biological age. Elevated blood DNAmAge is associated with all-cause mortality, with the strongest effects reported in a recent intrapair twin study where epigenetically older twins had increased mortality risk in comparison to their co-twins. In the study presented here, we hypothesize that DNAmAge in blood is associated with cross-sectional and longitudinal cognitive abilities in middle-aged individuals. In 486 monozygotic twins, we investigated the association of DNAmAge, difference between DNAmAge and chronological age and age acceleration with cognition. Despite using a powerful paired twin design, we found no evidence for association of blood DNAmAge with cognitive abilities. This observation was confirmed in unpaired analyses, where DNAmAge initially correlated with cognitive abilities, until adjusting for chronological age. Overall, our study shows that for middle-aged individuals DNAmAge calculated in blood does not correlate with cognitive abilities.


Asunto(s)
Envejecimiento/genética , Cognición/fisiología , Metilación de ADN , Gemelos Monocigóticos , Estudios Transversales , Epigénesis Genética , Femenino , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad
5.
Transl Psychiatry ; 6(6): e831, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27271857

RESUMEN

The CACNA1C gene, encoding a subunit of the L-type voltage-gated calcium channel is one of the best-supported susceptibility genes for bipolar disorder (BD). Genome-wide association studies have identified a cluster of non-coding single-nucleotide polymorphisms (SNPs) in intron 3 to be highly associated with BD and schizophrenia. The mechanism by which these SNPs confer risk of BD appears to be through an altered regulation of CACNA1C expression. The role of CACNA1C DNA methylation in BD has not yet been addressed. The aim of this study was to investigate if CACNA1C DNA methylation is altered in BD. First, the methylation status of five CpG islands (CGIs) across CACNA1C in blood from BD subjects (n=40) and healthy controls (n=38) was determined. Four islands were almost completely methylated or completely unmethylated, while one island (CGI 3) in intron 3 displayed intermediate methylation levels. In the main analysis, the methylation status of CGI 3 was analyzed in a larger sample of BD subjects (n=582) and control individuals (n=319). Out of six CpG sites that were investigated, five sites showed significant hypermethylation in cases (lowest P=1.16 × 10(-7) for CpG35). Nearby SNPs were found to influence the methylation level, and we identified rs2238056 in intron 3 as the strongest methylation quantitative trait locus (P=2.6 × 10(-7)) for CpG35. In addition, we found an increased methylation in females, and no difference between bipolar I and II. In conclusion, we find that CACNA1C methylation is associated with BD and suggest that the regulatory effect of the non-coding risk variants involves a shift in DNA methylation.


Asunto(s)
Trastorno Bipolar/genética , Canales de Calcio Tipo L/genética , Metilación de ADN/genética , Islas de CpG/genética , Femenino , Regulación de la Expresión Génica/genética , Genotipo , Humanos , Intrones , Masculino , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo/genética , Valores de Referencia , Factores Sexuales
6.
Klin Monbl Augenheilkd ; 229(12): 1223-6, 2012 Dec.
Artículo en Alemán | MEDLINE | ID: mdl-23196811

RESUMEN

INTRODUCTION: The aim of this study was to compare individual accuracy and speed of laser application using the Navilas laser system in patients with diabetic and vascular macular oedema. METHODS: The Navilas laser system was used by an experienced retina specialist and a relatively inexperienced ophthalmologist. Obtained results were retrospectively assessed with regard to precision and speed of laser application. RESULTS: Precision of laser therapy was quite similar, 88 % and 84 % for experienced and inexperienced operators, respectively. The treatment procedure took on average 5.5 minutes and 5.34 minutes for experienced and inexperienced operators, respectively. CONCLUSIONS: Precision and duration of therapy using the Navilas laser system do not seem to be correlated with the individual experience of the treating physician.


Asunto(s)
Retinopatía Diabética/patología , Retinopatía Diabética/cirugía , Coagulación con Láser/métodos , Edema Macular/patología , Edema Macular/cirugía , Competencia Profesional , Terapia Asistida por Computador/métodos , Adulto , Anciano , Femenino , Humanos , Coagulación con Láser/instrumentación , Masculino , Persona de Mediana Edad , Terapia Asistida por Computador/instrumentación , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA