Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Psychiatry ; 26(7): 3004-3017, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33057169

RESUMEN

Developmental dyslexia (DD) is a learning disorder affecting the ability to read, with a heritability of 40-60%. A notable part of this heritability remains unexplained, and large genetic studies are warranted to identify new susceptibility genes and clarify the genetic bases of dyslexia. We carried out a genome-wide association study (GWAS) on 2274 dyslexia cases and 6272 controls, testing associations at the single variant, gene, and pathway level, and estimating heritability using single-nucleotide polymorphism (SNP) data. We also calculated polygenic scores (PGSs) based on large-scale GWAS data for different neuropsychiatric disorders and cortical brain measures, educational attainment, and fluid intelligence, testing them for association with dyslexia status in our sample. We observed statistically significant (p < 2.8 × 10-6) enrichment of associations at the gene level, for LOC388780 (20p13; uncharacterized gene), and for VEPH1 (3q25), a gene implicated in brain development. We estimated an SNP-based heritability of 20-25% for DD, and observed significant associations of dyslexia risk with PGSs for attention deficit hyperactivity disorder (at pT = 0.05 in the training GWAS: OR = 1.23[1.16; 1.30] per standard deviation increase; p = 8 × 10-13), bipolar disorder (1.53[1.44; 1.63]; p = 1 × 10-43), schizophrenia (1.36[1.28; 1.45]; p = 4 × 10-22), psychiatric cross-disorder susceptibility (1.23[1.16; 1.30]; p = 3 × 10-12), cortical thickness of the transverse temporal gyrus (0.90[0.86; 0.96]; p = 5 × 10-4), educational attainment (0.86[0.82; 0.91]; p = 2 × 10-7), and intelligence (0.72[0.68; 0.76]; p = 9 × 10-29). This study suggests an important contribution of common genetic variants to dyslexia risk, and novel genomic overlaps with psychiatric conditions like bipolar disorder, schizophrenia, and cross-disorder susceptibility. Moreover, it revealed the presence of shared genetic foundations with a neural correlate previously implicated in dyslexia by neuroimaging evidence.


Asunto(s)
Dislexia , Herencia Multifactorial , Polimorfismo de Nucleótido Simple , Trastorno por Déficit de Atención con Hiperactividad/genética , Dislexia/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética
2.
Am J Hum Genet ; 94(1): 23-32, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24360809

RESUMEN

Mutations in interferon regulatory factor 6 (IRF6) account for ∼70% of cases of Van der Woude syndrome (VWS), the most common syndromic form of cleft lip and palate. In 8 of 45 VWS-affected families lacking a mutation in IRF6, we found coding mutations in grainyhead-like 3 (GRHL3). According to a zebrafish-based assay, the disease-associated GRHL3 mutations abrogated periderm development and were consistent with a dominant-negative effect, in contrast to haploinsufficiency seen in most VWS cases caused by IRF6 mutations. In mouse, all embryos lacking Grhl3 exhibited abnormal oral periderm and 17% developed a cleft palate. Analysis of the oral phenotype of double heterozygote (Irf6(+/-);Grhl3(+/-)) murine embryos failed to detect epistasis between the two genes, suggesting that they function in separate but convergent pathways during palatogenesis. Taken together, our data demonstrated that mutations in two genes, IRF6 and GRHL3, can lead to nearly identical phenotypes of orofacial cleft. They supported the hypotheses that both genes are essential for the presence of a functional oral periderm and that failure of this process contributes to VWS.


Asunto(s)
Anomalías Múltiples/patología , Labio Leporino/patología , Fisura del Paladar/patología , Quistes/patología , Proteínas de Unión al ADN/genética , Labio/anomalías , Factores de Transcripción/genética , Anomalías Múltiples/genética , Alelos , Animales , Labio Leporino/genética , Fisura del Paladar/genética , Quistes/genética , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genotipo , Humanos , Hibridación Genética , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Labio/patología , Ratones , Ratones Noqueados , Mutación Missense , Linaje , Fenotipo , Análisis de Secuencia de ADN , Factores de Transcripción/metabolismo , Pez Cebra/embriología , Pez Cebra/genética
3.
J Neurosci ; 34(43): 14455-62, 2014 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-25339756

RESUMEN

Three genes, DYX1C1, DCDC2, and KIAA0319, have been previously associated with dyslexia, neuronal migration, and ciliary function. Three polymorphisms within these genes, rs3743204 (DYX1C1), rs793842 (DCDC2), and rs6935076 (KIAA0319) have also been linked to normal variability of left temporoparietal white matter volume connecting the middle temporal cortex to the angular and supramarginal gyri. Here, we assessed whether these polymorphisms are also related to the cortical thickness of the associated regions during childhood development using a longitudinal dataset of 76 randomly selected children and young adults who were scanned up to three times each, 2 years apart. rs793842 in DCDC2 was significantly associated with the thickness of left angular and supramarginal gyri as well as the left lateral occipital cortex. The cortex was significantly thicker for T-allele carriers, who also had lower white matter volume and lower reading comprehension scores. There was a negative correlation between white matter volume and cortical thickness, but only white matter volume predicted reading comprehension 2 years after scanning. These results show how normal variability in reading comprehension is related to gene, white matter volume, and cortical thickness in the inferior parietal lobe. Possibly, the variability of gray and white matter structures could both be related to the role of DCDC2 in ciliary function, which affects both neuronal migration and axonal outgrowth.


Asunto(s)
Desarrollo del Adolescente/fisiología , Desarrollo Infantil/fisiología , Proteínas Asociadas a Microtúbulos/genética , Lóbulo Parietal/crecimiento & desarrollo , Lóbulo Temporal/crecimiento & desarrollo , Sustancia Blanca/crecimiento & desarrollo , Adolescente , Adulto , Niño , Femenino , Humanos , Masculino , Polimorfismo de Nucleótido Simple/genética , Adulto Joven
4.
Hum Genet ; 134(11-12): 1239-48, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26400686

RESUMEN

Developmental dyslexia is the most common learning disorder in children. Problems in reading and writing are likely due to a complex interaction of genetic and environmental factors, resulting in reduced power of studies of the genetic factors underlying developmental dyslexia. Our approach in the current study was to perform exome sequencing of affected and unaffected individuals within an extended pedigree with a familial form of developmental dyslexia. We identified a two-base mutation, causing a p.R229L amino acid substitution in the centrosomal protein 63 kDa (CEP63), co-segregating with developmental dyslexia in this pedigree. This mutation is novel, and predicted to be highly damaging for the function of the protein. 3D modelling suggested a distinct conformational change caused by the mutation. CEP63 is localised to the centrosome in eukaryotic cells and is required for maintaining normal centriole duplication and control of cell cycle progression. We found that a common polymorphism in the CEP63 gene had a significant association with brain white matter volume. The brain regions were partly overlapping with the previously reported region influenced by polymorphisms in the dyslexia susceptibility genes DYX1C1 and KIAA0319. We hypothesise that CEP63 is particularly important for brain development and might control the proliferation and migration of cells when those two events need to be highly coordinated.


Asunto(s)
Dislexia/genética , Predisposición Genética a la Enfermedad , Proteínas de Neoplasias/genética , Secuencia de Aminoácidos , Secuencia de Bases , Encéfalo/crecimiento & desarrollo , Proteínas de Ciclo Celular , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Ligamiento Genético , Humanos , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Proteínas de Neoplasias/química , Linaje , Suecia
5.
J Hum Genet ; 60(7): 399-401, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25877001

RESUMEN

Genetic studies of complex traits have become increasingly successful as progress is made in next-generation sequencing. We aimed at discovering single nucleotide variation present in known and new candidate genes for developmental dyslexia: CYP19A1, DCDC2, DIP2A, DYX1C1, GCFC2 (also known as C2orf3), KIAA0319, MRPL19, PCNT, PRMT2, ROBO1 and S100B. We used next-generation sequencing to identify single-nucleotide polymorphisms in the exons of these 11 genes in pools of 100 DNA samples of Finnish individuals with developmental dyslexia. Subsequent individual genotyping of those 100 individuals, and additional cases and controls from the Finnish and German populations, validated 92 out of 111 different single-nucleotide variants. A nonsynonymous polymorphism in DCDC2 (corrected P = 0.002) and a noncoding variant in S100B (corrected P = 0.016) showed a significant association with spelling performance in families of German origin. No significant association was found for the variants neither in the Finnish case-control sample set nor in the Finnish family sample set. Our findings further strengthen the role of DCDC2 and implicate S100B, in the biology of reading and spelling.


Asunto(s)
Dislexia/genética , Proteínas Asociadas a Microtúbulos/genética , Subunidad beta de la Proteína de Unión al Calcio S100/genética , Estudios de Casos y Controles , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN
6.
J Cogn Neurosci ; 26(1): 54-62, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24001007

RESUMEN

Studying the effects of cognitive training can lead to finding better treatments, but it can also be a tool for investigating factors important for brain plasticity and acquisition of cognitive skills. In this study, we investigated how single-nucleotide polymorphisms (SNPs) and ratings of intrinsic motivation were associated to interindividual differences in improvement during working memory training. The study included 256 children aged 7-19 years who were genotyped for 13 SNPs within or near eight candidate genes previously implicated in learning: COMT, SLC6A3 (DAT1), DRD4, DRD2, PPP1R1B (DARPP32), MAOA, LMX1A, and BDNF. Ratings on the intrinsic motivation inventory were also available for 156 of these children. All participants performed at least 20 sessions of working memory training, and performance during the training was logged and used as the outcome variable. We found that two SNPs, rs1800497 and rs2283265, located near and within the dopamine receptor 2 (DRD2) gene, respectively, were significantly associated with improvements during training (p < .003 and p < .0004, respectively). Scores from a questionnaire regarding intrinsic motivation did not correlate with training outcome. However, we observed both the main effect of genotype at those two loci as well as the interaction between genotypes and ratings of intrinsic motivation (perceived competence). Both SNPs have previously been shown to affect DRD2 receptor density primarily in the BG. Our results suggest that genetic variation is accounting for some interindividual differences in how children acquire cognitive skills and that part of this effect is also seen on intrinsic motivation. Moreover, they suggest that dopamine D2 transmission in the BG is a key factor for cognitive plasticity.


Asunto(s)
Cognición/fisiología , Memoria a Corto Plazo/fisiología , Motivación/genética , Polimorfismo de Nucleótido Simple/genética , Desempeño Psicomotor/fisiología , Receptores de Dopamina D2/genética , Adolescente , Niño , Femenino , Humanos , Masculino , Adulto Joven
7.
Behav Genet ; 42(4): 509-27, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22426781

RESUMEN

Inspired by the localization, on 15q21.2 of the CYP19A1 gene in the linkage region of speech and language disorders, and a rare translocation in a dyslexic individual that was brought to our attention, we conducted a series of studies on the properties of CYP19A1 as a candidate gene for dyslexia and related conditions. The aromatase enzyme is a member of the cytochrome P450 super family, and it serves several key functions: it catalyzes the conversion of androgens into estrogens; during early mammalian development it controls the differentiation of specific brain areas (e.g. local estrogen synthesis in the hippocampus regulates synaptic plasticity and axonal growth); it is involved in sexual differentiation of the brain; and in songbirds and teleost fishes, it regulates vocalization. Our results suggest that variations in CYP19A1 are associated with dyslexia as a categorical trait and with quantitative measures of language and speech, such as reading, vocabulary, phonological processing and oral motor skills. Variations near the vicinity of its brain promoter region altered transcription factor binding, suggesting a regulatory role in CYP19A1 expression. CYP19A1 expression in human brain correlated with the expression of dyslexia susceptibility genes such as DYX1C1 and ROBO1. Aromatase-deficient mice displayed increased cortical neuronal density and occasional cortical heterotopias, also observed in Robo1-/- mice and human dyslexic brains, respectively. An aromatase inhibitor reduced dendritic growth in cultured rat neurons. From this broad set of evidence, we propose CYP19A1 as a candidate gene for human cognitive functions implicated in reading, speech and language.


Asunto(s)
Aromatasa/genética , Encéfalo/crecimiento & desarrollo , Dislexia/genética , Trastornos del Lenguaje/genética , ARN Mensajero/análisis , Trastornos del Habla/genética , Animales , Aromatasa/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Estudios de Cohortes , Proteínas del Citoesqueleto , Dislexia/metabolismo , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Trastornos del Lenguaje/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Sitios de Carácter Cuantitativo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Trastornos del Habla/metabolismo , Translocación Genética , Proteínas Roundabout
8.
Behav Genet ; 41(1): 134-40, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21203819

RESUMEN

Four genes, DYX1C1, ROBO1, DCDC2 and KIAA0319 have been studied both genetically and functionally as candidate genes for developmental dyslexia, a common learning disability in children. The identification of novel genes is crucial to better understand the molecular pathways affected in dyslectic individuals. Here, we report results from a fine-mapping approach involving linkage and association analysis in Finnish and German dyslexic cohorts. We restrict a candidate region to 0.3 Mb on chromosome 7q33. This region harbours the gene diacylglycerol kinase, iota (DGKI) which contains overlapping haplotypes associated with dyslexia in both Finnish and German sample sets.


Asunto(s)
Alelos , Mapeo Cromosómico , Cromosomas Humanos Par 7/genética , Diacilglicerol Quinasa/genética , Dislexia/genética , Estudios de Asociación Genética , Variación Genética/genética , Genética de Población , Polimorfismo de Nucleótido Simple/genética , Niño , Estudios de Cohortes , Femenino , Finlandia , Marcadores Genéticos/genética , Genotipo , Alemania , Haplotipos , Humanos , Estudios Longitudinales , Masculino , Fenotipo
9.
Am J Med Genet A ; 155A(6): 1314-21, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21574244

RESUMEN

Genetic variation in the transcription factor interferon regulatory factor 6 (IRF6) causes and contributes risk for oral clefting disorders. We hypothesized that genes regulated by IRF6 are also involved in oral clefting disorders. We used five criteria to identify potential IRF6 target genes; differential gene expression in skin taken from wild-type and Irf6-deficient murine embryos, localization to the Van der Woude syndrome 2 (VWS2) locus at 1p36-1p32, overlapping expression with Irf6, presence of a conserved predicted-binding site in the promoter region, and a mutant murine phenotype that was similar to the Irf6 mutant mouse. Previously, we observed altered expression for 573 genes; 13 were located in the murine region syntenic to the VWS2 locus. Two of these genes, Wdr65 and Stratifin, met 4 of 5 criteria. Wdr65 was a novel gene that encoded a predicted protein of 1,250 amino acids with two WD domains. As potential targets for Irf6 regulation, we hypothesized that disease-causing mutations will be found in WDR65 and Stratifin in individuals with VWS or VWS-like syndromes. We identified a potentially etiologic missense mutation in WDR65 in a person with VWS who does not have an exonic mutation in IRF6. The expression and mutation data were consistent with the hypothesis that WDR65 was a novel gene involved in oral clefting.


Asunto(s)
Anomalías Múltiples/genética , Cromosomas Humanos Par 1/genética , Labio Leporino/genética , Fisura del Paladar/genética , Quistes/genética , Regulación de la Expresión Génica/genética , Factores Reguladores del Interferón/genética , Mutación Missense/genética , Proteínas/genética , Animales , Secuencia de Bases , Clonación Molecular , Biología Computacional , ADN Complementario/genética , Humanos , Hibridación in Situ , Labio/anomalías , Ratones , Análisis por Micromatrices , Proteínas Asociadas a Microtúbulos , Datos de Secuencia Molecular , Proteínas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN
10.
Transl Psychiatry ; 9(1): 77, 2019 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-30741946

RESUMEN

Developmental dyslexia (DD) is one of the most prevalent learning disorders, with high impact on school and psychosocial development and high comorbidity with conditions like attention-deficit hyperactivity disorder (ADHD), depression, and anxiety. DD is characterized by deficits in different cognitive skills, including word reading, spelling, rapid naming, and phonology. To investigate the genetic basis of DD, we conducted a genome-wide association study (GWAS) of these skills within one of the largest studies available, including nine cohorts of reading-impaired and typically developing children of European ancestry (N = 2562-3468). We observed a genome-wide significant effect (p < 1 × 10-8) on rapid automatized naming of letters (RANlet) for variants on 18q12.2, within MIR924HG (micro-RNA 924 host gene; rs17663182 p = 4.73 × 10-9), and a suggestive association on 8q12.3 within NKAIN3 (encoding a cation transporter; rs16928927, p = 2.25 × 10-8). rs17663182 (18q12.2) also showed genome-wide significant multivariate associations with RAN measures (p = 1.15 × 10-8) and with all the cognitive traits tested (p = 3.07 × 10-8), suggesting (relational) pleiotropic effects of this variant. A polygenic risk score (PRS) analysis revealed significant genetic overlaps of some of the DD-related traits with educational attainment (EDUyears) and ADHD. Reading and spelling abilities were positively associated with EDUyears (p ~ [10-5-10-7]) and negatively associated with ADHD PRS (p ~ [10-8-10-17]). This corroborates a long-standing hypothesis on the partly shared genetic etiology of DD and ADHD, at the genome-wide level. Our findings suggest new candidate DD susceptibility genes and provide new insights into the genetics of dyslexia and its comorbities.


Asunto(s)
Cognición , Dislexia/genética , Dislexia/psicología , Adolescente , Adulto , Niño , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Herencia Multifactorial , Polimorfismo de Nucleótido Simple , Adulto Joven
11.
J Appl Physiol (1985) ; 103(1): 315-22, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17463303

RESUMEN

AMP deaminase (AMPD) deficiency is an inherited disorder of skeletal muscle found in approximately 2% of the Caucasian population. Although most AMPD-deficient individuals are asymptomatic, a small subset has exercise-related cramping and pain without any other identifiable neuromuscular complications. This heterogeneity has raised doubts about the physiological significance of AMPD in skeletal muscle, despite evidence for disrupted adenine nucleotide catabolism during exercise in deficient individuals. Previous studies have evaluated the effect of AMPD deficiency on exercise performance with mixed results. This study was designed to circumvent the perceived limitations in previous reports by measuring exercise performance during a 30-s Wingate test in 139 healthy, physically active subjects of both sexes, with different AMPD1 genotypes, including 12 AMPD-deficient subjects. Three of the deficient subjects were compound heterozygotes characterized by the common c.34C>T mutation in one allele and a newly discovered AMPD1 mutation, c.404delT, in the other. While there was no significant difference in peak power across AMPD1 genotypes, statistical analysis revealed a faster power decrease in the AMPD-deficient group and a difference in mean power across the genotypes (P = 0.0035). This divergence was most striking at 15 s of the 30-s cycling. Assessed by the fatigue index, the decrease in power output at 15 s of exercise was significantly greater in the deficient group compared with the other genotypes (P = 0.0006). The approximate 10% lower mean power in healthy AMPD-deficient subjects during a 30-s Wingate cycling test reveals a functional role for the AMPD1 enzyme in sprint exercise.


Asunto(s)
AMP Desaminasa/deficiencia , Ejercicio Físico/fisiología , Fatiga Muscular/genética , Fuerza Muscular/genética , Músculo Esquelético/enzimología , AMP Desaminasa/genética , Adulto , Amoníaco/sangre , Análisis Mutacional de ADN , Prueba de Esfuerzo/métodos , Femenino , Heterocigoto , Homocigoto , Humanos , Ácido Láctico/sangre , Masculino , Fibras Musculares Esqueléticas/enzimología , Músculo Esquelético/citología , Mutación , Fenotipo , Valores de Referencia , Reproducibilidad de los Resultados , Factores de Tiempo
12.
Brain Struct Funct ; 222(2): 707-716, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27240594

RESUMEN

The axon guidance receptor, Robo1, controls the pathfinding of callosal axons in mice. To determine whether the orthologous ROBO1 gene is involved in callosal development also in humans, we studied polymorphisms in the ROBO1 gene and variation in the white matter structure in the corpus callosum using both structural magnetic resonance imaging and diffusion tensor magnetic resonance imaging. We found that five polymorphisms in the regulatory region of ROBO1 were associated with white matter density in the posterior part of the corpus callosum pathways. One of the polymorphisms, rs7631357, was also significantly associated with the probability of connections to the parietal cortical regions. Our results demonstrate that human ROBO1 may be involved in the regulation of the structure and connectivity of posterior part of corpus callosum.


Asunto(s)
Cuerpo Calloso/anatomía & histología , Cuerpo Calloso/crecimiento & desarrollo , Proteínas del Tejido Nervioso/fisiología , Receptores Inmunológicos/fisiología , Sustancia Blanca/anatomía & histología , Sustancia Blanca/crecimiento & desarrollo , Adolescente , Adulto , Niño , Imagen de Difusión por Resonancia Magnética , Imagen de Difusión Tensora , Genotipo , Humanos , Proteínas del Tejido Nervioso/genética , Vías Nerviosas/anatomía & histología , Vías Nerviosas/crecimiento & desarrollo , Lóbulo Parietal/anatomía & histología , Lóbulo Parietal/crecimiento & desarrollo , Polimorfismo de Nucleótido Simple , Receptores Inmunológicos/genética , Adulto Joven , Proteínas Roundabout
13.
Sci Rep ; 7(1): 9294, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28839234

RESUMEN

A whole-genome linkage analysis in a Finnish pedigree of eight cases with developmental dyslexia (DD) revealed several regions shared by the affected individuals. Analysis of coding variants from two affected individuals identified rs146011974G > A (Ala1039Thr), a rare variant within the NCAN gene co-segregating with DD in the pedigree. This variant prompted us to consider this gene as a putative candidate for DD. The RNA expression pattern of the NCAN gene in human tissues was highly correlated (R > 0.8) with that of the previously suggested DD susceptibility genes KIAA0319, CTNND2, CNTNAP2 and GRIN2B. We investigated the association of common variation in NCAN to brain structures in two data sets: young adults (Brainchild study, Sweden) and infants (FinnBrain study, Finland). In young adults, we found associations between a common genetic variant in NCAN, rs1064395, and white matter volume in the left and right temporoparietal as well as the left inferior frontal brain regions. In infants, this same variant was found to be associated with cingulate and prefrontal grey matter volumes. Our results suggest NCAN as a new candidate gene for DD and indicate that NCAN variants affect brain structure.


Asunto(s)
Proteoglicanos Tipo Condroitín Sulfato/genética , Dislexia/genética , Predisposición Genética a la Enfermedad , Lectinas Tipo C/genética , Proteínas del Tejido Nervioso/genética , Adolescente , Adulto , Encéfalo/patología , Niño , Preescolar , Femenino , Finlandia , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Imagen por Resonancia Magnética , Masculino , Neurocano , Suecia , Adulto Joven
14.
FASEB J ; 19(7): 750-60, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15857889

RESUMEN

Global gene expression profiling is used to generate novel insight into a variety of disease states. Such studies yield a bewildering number of data points, making it a challenge to validate which genes specifically contribute to a disease phenotype. Aerobic exercise training represents a plausible model for identification of molecular mechanisms that cause metabolic-related changes in human skeletal muscle. We carried out the first transcriptome-wide characterization of human skeletal muscle responses to 6 wk of supervised aerobic exercise training in 8 sedentary volunteers. Biopsy samples before and after training allowed us to identify approximately 470 differentially regulated genes using the Affymetrix U95 platform (80 individual hybridization steps). Gene ontology analysis indicated that extracellular matrix and calcium binding gene families were most up-regulated after training. An electronic reanalysis of a Duchenne muscular dystrophy (DMD) transcript expression dataset allowed us to identify approximately 90 genes modulated in a nearly identical fashion to that observed in the endurance exercise dataset. Trophoblast noncoding RNA, an interfering RNA species, was the singular exception-being up-regulated by exercise and down-regulated in DMD. The common overlap between gene expression datasets may be explained by enhanced alpha7beta1 integrin signaling, and specific genes in this signaling pathway were up-regulated in both datasets. In contrast to these common features, OXPHOS gene expression is subdued in DMD yet elevated by exercise, indicating that more than one major mechanism must exist in human skeletal muscle to sense activity and therefore regulate gene expression. Exercise training modulated diabetes-related genes, suggesting our dataset may contain additional and novel gene expression changes relevant for the anti-diabetic properties of exercise. In conclusion, gene expression profiling after endurance exercise training identified a range of processes responsible for the physiological remodeling of human skeletal muscle tissue, many of which were similarly regulated in DMD. Furthermore, our analysis demonstrates that numerous genes previously suggested as being important for the DMD disease phenotype may principally reflect compensatory integrin signaling.


Asunto(s)
Perfilación de la Expresión Génica , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Resistencia Física/genética , Biopsia , Proteínas de Unión al Calcio/genética , Diabetes Mellitus/genética , Ejercicio Físico , Proteínas de la Matriz Extracelular/genética , Regulación de la Expresión Génica , Frecuencia Cardíaca , Humanos , Masculino , Músculo Esquelético/química , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación Oxidativa , Consumo de Oxígeno , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Suecia
15.
Eur J Hum Genet ; 13(12): 1261-7, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16160700

RESUMEN

The interferon regulatory factor 6 gene (IRF6) has been identified as the major Van der Woude (VWS) syndrome and popliteal pterygium (PPS) syndrome gene with mutations in the majority of the kindreds. We have studied altogether 17 kindreds from Sweden, Finland, Norway, Thailand and Singapore, and report here 10 mutations, six of them previously unseen. In two kindreds, we could document de novo mutations, both of them changing a codon for a glutamine residue to a stop. No mutation could be detected in the four VWS kindreds from Finland, suggesting a founder effect for a mutation in an atypical noncoding position. Our findings demonstrate that several distinct mutations occur in the Swedish population, and confirm the general notion of a broad spectrum of IRF6 mutations underlying the VWS/PPS phenotypes.


Asunto(s)
Labio Leporino/genética , Fisura del Paladar/genética , Factores Reguladores del Interferón/genética , Anomalías Cutáneas/genética , Análisis Mutacional de ADN , Femenino , Genes Dominantes , Genitales/anomalías , Humanos , Pierna/anomalías , Masculino , Mutación , Linaje , Polimorfismo Genético , Síndrome
16.
Eur J Hum Genet ; 13(1): 127-30, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15470369

RESUMEN

DYX1C1: was recently identified as a candidate gene for developmental dyslexia, which is characterized by an unexpected difficulty in learning to read and write despite adequate intelligence, motivation, and education. It will be important to clarify, whether the phenotype caused by DYX1C1 extends to other language-related or comorbid disorders. Impaired language development is one of the essential features in autism. Therefore, we analyzed the allelic distribution of the DYX1C1 gene by family-based association method in 100 Finnish autism families. No evidence for association was observed with any intragenic marker or with haplotypes constructed from alleles of several adjacent markers. No evidence for deviated allelic diversity was either observed: the frequency of expected dyslexia risk haplotype was comparable to its frequency in Finnish controls. Thus it seems unlikely that DYX1C1 gene would be involved in the genetic etiology of autism in Finnish patients.


Asunto(s)
Alelos , Trastorno Autístico/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Polimorfismo de Nucleótido Simple , Proteínas del Citoesqueleto , Familia , Femenino , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Genotipo , Haplotipos , Humanos , Desequilibrio de Ligamiento , Masculino
17.
PLoS One ; 9(2): e89544, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586859

RESUMEN

First, we explore the performance of nonword repetition (NWR) in children with specific language impairment (SLI) and typically developing children (TD) in order to investigate the accuracy of NWR as a clinical marker for SLI in Swedish-speaking school-age children. Second, we examine the relationship between NWR, family aggregation, and parental level of education in children with SLI. A sample of 61 children with SLI, and 86 children with TD, aged 8-12 years, were administered an NWR test. Family aggregation, measured as the prevalence of language and/or literacy problems (LLP) in parents of the children with SLI, was based on family history interviews. The sensitivity and specificity of nonword repetition was analyzed in a binary logistic regression, cut-off values were established with ROC curves, and positive and negative likelihood ratios reported. Results from the present study show that NWR distinguishes well between Swedish-speaking school-children with and without SLI. We found 90.2% sensitivity and 97.7% specificity at a cut-off level of -2 standard deviations for binary scoring of nonwords. Differences between the SLI and TD groups showed large effect sizes for the two scoring measures binary (d = 2.11) and percent correct consonants (PCC) (d = 1.79). The children with SLI were split into two subgroups: those with no parents affected with LLP (n = 12), and those with one or both parents affected (n = 49). The subgroup consisting of affected parents had a significantly lower score on NWR binary (p = .037), and there was a great difference between the subgroups (d = 0.7). When compared to the TD group, the difference from the subgroup with affected parents was almost one standard deviation larger (d = 2.47) than the difference from the TD to the subgroup consisting of non-affected parents (d = 1.57). Our study calls for further exploration of the complex interaction between family aggregation, language input, and phenotypes of SLI.


Asunto(s)
Biomarcadores/análisis , Trastornos del Desarrollo del Lenguaje/clasificación , Trastornos del Desarrollo del Lenguaje/diagnóstico , Lenguaje , Relaciones Padres-Hijo , Padres , Conducta Verbal , Niño , Femenino , Humanos , Masculino , Tamizaje Masivo , Percepción del Habla , Medición de la Producción del Habla , Análisis y Desempeño de Tareas
18.
Eur J Hum Genet ; 22(5): 675-80, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24022301

RESUMEN

Dyslexia is one of the most common childhood disorders with a prevalence of around 5-10% in school-age children. Although an important genetic component is known to have a role in the aetiology of dyslexia, we are far from understanding the molecular mechanisms leading to the disorder. Several candidate genes have been implicated in dyslexia, including DYX1C1, DCDC2, KIAA0319, and the MRPL19/C2ORF3 locus, each with reports of both positive and no replications. We generated a European cross-linguistic sample of school-age children - the NeuroDys cohort - that includes more than 900 individuals with dyslexia, sampled with homogenous inclusion criteria across eight European countries, and a comparable number of controls. Here, we describe association analysis of the dyslexia candidate genes/locus in the NeuroDys cohort. We performed both case-control and quantitative association analyses of single markers and haplotypes previously reported to be dyslexia-associated. Although we observed association signals in samples from single countries, we did not find any marker or haplotype that was significantly associated with either case-control status or quantitative measurements of word-reading or spelling in the meta-analysis of all eight countries combined. Like in other neurocognitive disorders, our findings underline the need for larger sample sizes to validate possibly weak genetic effects.


Asunto(s)
Dislexia/genética , Estudios de Asociación Genética , Estudio de Asociación del Genoma Completo , Estudios de Casos y Controles , Sitios Genéticos , Genotipo , Haplotipos , Humanos , Metaanálisis como Asunto , Fenotipo , Polimorfismo de Nucleótido Simple , Carácter Cuantitativo Heredable
19.
Biol Psychiatry ; 72(8): 671-6, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22683091

RESUMEN

BACKGROUND: Volume and integrity of white matter correlate with reading ability, but the underlying factors contributing to this variability are unknown. METHODS: We investigated single nucleotide polymorphisms in three genes previously associated with dyslexia and implicated in neuronal migration (DYX1C1, DCDC2, KIAA0319) and white matter volume in a cohort of 76 children and young adults from the general population. RESULTS: We found that all three genes contained polymorphisms that were significantly associated with white matter volume in the left temporo-parietal region and that white matter volume influenced reading ability. CONCLUSIONS: The identified region contained white matter pathways connecting the middle temporal gyrus with the inferior parietal lobe. The finding links previous neuroimaging and genetic results and proposes a mechanism underlying variability in reading ability in both normal and impaired readers.


Asunto(s)
Proteínas Asociadas a Microtúbulos/genética , Fibras Nerviosas Mielínicas , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Lóbulo Parietal/anatomía & histología , Polimorfismo de Nucleótido Simple/genética , Lóbulo Temporal/anatomía & histología , Adolescente , Adulto , Niño , Proteínas del Citoesqueleto , Imagen de Difusión Tensora , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Imagenología Tridimensional , Modelos Lineales , Masculino , Tiempo de Reacción/genética , Lectura , Suecia , Adulto Joven
20.
Dev Psychol ; 48(3): 836-43, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22103304

RESUMEN

Cognitive deficits and particularly deficits in working memory (WM) capacity are common features in neuropsychiatric disorders. Understanding the underlying mechanisms through which WM capacity can be improved is therefore of great importance. Several lines of research indicate that dopamine plays an important role not only in WM function but also for improving WM capacity. For example, pharmacological interventions acting on the dopaminergic system, such as methylphenidate, improve WM performance. In addition, behavioral interventions for improving WM performance in the form of intensive computerized training have recently been associated with changes in dopamine receptor density. These two different means of improving WM performance--pharmacological and behavioral--are thus associated with similar biological mechanisms in the brain involving dopaminergic systems. This article reviews some of the evidence for the role of dopamine in WM functioning, in particular concerning the link to WM development and cognitive plasticity. Novel data are presented showing that variation in the dopamine transporter gene (DAT1) influences improvements in WM and fluid intelligence in preschool-age children following cognitive training. Our results emphasize the importance of the role of dopamine in determining cognitive plasticity.


Asunto(s)
Dopamina/metabolismo , Aprendizaje/fisiología , Memoria a Corto Plazo/fisiología , Plasticidad Neuronal/fisiología , Transferencia de Experiencia en Psicología/fisiología , Animales , Niño , Desarrollo Infantil/fisiología , Preescolar , Cognición , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Femenino , Genotipo , Humanos , Inteligencia/genética , Masculino , Plasticidad Neuronal/genética , Polimorfismo de Nucleótido Simple/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA