Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 161
Filtrer
1.
Sci Rep ; 10(1): 7469, 2020 05 04.
Article de Anglais | MEDLINE | ID: mdl-32366958

RÉSUMÉ

The genetic contribution to different aspects of empathy is now established, although the exact loci are unknown. We undertook a genome-wide association study of emotional empathy (EE) as measured by emotion recognition skills in 4,780 8-year old children from the ALSPAC cohort who were genotyped and imputed to Phase 1 version 3 of the 1000 Genomes Project. We failed to find any genome-wide significant signal in either our unstratified analysis or analysis stratified according to sex. A gene-based association analysis similarly failed to find any significant loci. In contrast, our transcriptome-wide association study (TWAS) with a whole blood reference panel identified two significant loci in the unstratified analysis, residualised for the effects of age, sex and IQ. One signal was for CD93 on chromosome 20; this gene is not strongly expressed in the brain, however. The other signal was for AL118508, a non-protein coding pseudogene, which completely lies within CD93's genomic coordinates, thereby explaining its signal. Neither are obvious candidates for involvement in the brain processes that underlie emotion recognition and its developmental pathways.


Sujet(s)
Chromosomes humains de la paire 20/génétique , Émotions , Empathie/génétique , Génotype , Hérédité multifactorielle , Transcriptome , Enfant , Chromosomes humains de la paire 20/métabolisme , Femelle , Étude d'association pangénomique , Humains , Études longitudinales , Mâle
2.
Nat Commun ; 10(1): 5519, 2019 12 05.
Article de Anglais | MEDLINE | ID: mdl-31801954

RÉSUMÉ

Identification of genetic biomarkers associated with autism spectrum disorders (ASDs) could improve recurrence prediction for families with a child with ASD. Here, we describe clinical microarray findings for 253 longitudinally phenotyped ASD families from the Baby Siblings Research Consortium (BSRC), encompassing 288 infant siblings. By age 3, 103 siblings (35.8%) were diagnosed with ASD and 54 (18.8%) were developing atypically. Thirteen siblings have copy number variants (CNVs) involving ASD-relevant genes: 6 with ASD, 5 atypically developing, and 2 typically developing. Within these families, an ASD-related CNV in a sibling has a positive predictive value (PPV) for ASD or atypical development of 0.83; the Simons Simplex Collection of ASD families shows similar PPVs. Polygenic risk analyses suggest that common genetic variants may also contribute to ASD. CNV findings would have been pre-symptomatically predictive of ASD or atypical development in 11 (7%) of the 157 BSRC siblings who were eventually diagnosed clinically.


Sujet(s)
Trouble du spectre autistique/génétique , Variations de nombre de copies de segment d'ADN , Prédisposition génétique à une maladie/génétique , Génome humain/génétique , Génomique/méthodes , Fratrie , Trouble du spectre autistique/diagnostic , Enfant d'âge préscolaire , Santé de la famille , Femelle , Humains , Mâle , Pedigree , Phénotype , Facteurs de risque
3.
Clin Epigenetics ; 11(1): 103, 2019 07 16.
Article de Anglais | MEDLINE | ID: mdl-31311581

RÉSUMÉ

BACKGROUND: Autism spectrum disorder (ASD) is a common and etiologically heterogeneous neurodevelopmental disorder. Although many genetic causes have been identified (> 200 ASD-risk genes), no single gene variant accounts for > 1% of all ASD cases. A role for epigenetic mechanisms in ASD etiology is supported by the fact that many ASD-risk genes function as epigenetic regulators and evidence that epigenetic dysregulation can interrupt normal brain development. Gene-specific DNAm profiles have been shown to assist in the interpretation of variants of unknown significance. Therefore, we investigated the epigenome in patients with ASD or two of the most common genomic variants conferring increased risk for ASD. Genome-wide DNA methylation (DNAm) was assessed using the Illumina Infinium HumanMethylation450 and MethylationEPIC arrays in blood from individuals with ASD of heterogeneous, undefined etiology (n = 52), and individuals with 16p11.2 deletions (16p11.2del, n = 9) or pathogenic variants in the chromatin modifier CHD8 (CHD8+/-, n = 7). RESULTS: DNAm patterns did not clearly distinguish heterogeneous ASD cases from controls. However, the homogeneous genetically-defined 16p11.2del and CHD8+/- subgroups each exhibited unique DNAm signatures that distinguished 16p11.2del or CHD8+/- individuals from each other and from heterogeneous ASD and control groups with high sensitivity and specificity. These signatures also classified additional 16p11.2del (n = 9) and CHD8 (n = 13) variants as pathogenic or benign. Our findings that DNAm alterations in each signature target unique genes in relevant biological pathways including neural development support their functional relevance. Furthermore, genes identified in our CHD8+/- DNAm signature in blood overlapped differentially expressed genes in CHD8+/- human-induced pluripotent cell-derived neurons and cerebral organoids from independent studies. CONCLUSIONS: DNAm signatures can provide clinical utility complementary to next-generation sequencing in the interpretation of variants of unknown significance. Our study constitutes a novel approach for ASD risk-associated molecular classification that elucidates the vital cross-talk between genetics and epigenetics in the etiology of ASD.


Sujet(s)
Trouble du spectre autistique/génétique , Trouble autistique/génétique , Maladies chromosomiques/génétique , Méthylation de l'ADN , Protéines de liaison à l'ADN/génétique , Étude d'association pangénomique/méthodes , Déficience intellectuelle/génétique , Facteurs de transcription/génétique , Adolescent , Études cas-témoins , Enfant , Enfant d'âge préscolaire , Délétion de segment de chromosome , Chromosomes humains de la paire 16/génétique , Épigenèse génétique , Femelle , Réseaux de régulation génique , Prédisposition génétique à une maladie , Séquençage nucléotidique à haut débit , Humains , Nourrisson , Mâle , Sensibilité et spécificité , Analyse de séquence d'ADN
4.
Mol Psychiatry ; 22(6): 836-849, 2017 06.
Article de Anglais | MEDLINE | ID: mdl-27240531

RÉSUMÉ

Copy number variants (CNVs) are major contributors to genomic imbalance disorders. Phenotyping of 137 unrelated deletion and reciprocal duplication carriers of the distal 16p11.2 220 kb BP2-BP3 interval showed that these rearrangements are associated with autism spectrum disorders and mirror phenotypes of obesity/underweight and macrocephaly/microcephaly. Such phenotypes were previously associated with rearrangements of the non-overlapping proximal 16p11.2 600 kb BP4-BP5 interval. These two CNV-prone regions at 16p11.2 are reciprocally engaged in complex chromatin looping, as successfully confirmed by 4C-seq, fluorescence in situ hybridization and Hi-C, as well as coordinated expression and regulation of encompassed genes. We observed that genes differentially expressed in 16p11.2 BP4-BP5 CNV carriers are concomitantly modified in their chromatin interactions, suggesting that disruption of chromatin interplays could participate in the observed phenotypes. We also identified cis- and trans-acting chromatin contacts to other genomic regions previously associated with analogous phenotypes. For example, we uncovered that individuals with reciprocal rearrangements of the trans-contacted 2p15 locus similarly display mirror phenotypes on head circumference and weight. Our results indicate that chromosomal contacts' maps could uncover functionally and clinically related genes.


Sujet(s)
Trouble autistique/génétique , Cartographie chromosomique/méthodes , Chromosomes humains de la paire 16/physiologie , Obésité/génétique , Adolescent , Adulte , Sujet âgé , Trouble du spectre autistique/génétique , Indice de masse corporelle , Enfant , Enfant d'âge préscolaire , Chromatine/métabolisme , Chromatine/physiologie , Délétion de segment de chromosome , Duplication chromosomique , Chromosomes humains de la paire 16/génétique , Variations de nombre de copies de segment d'ADN/génétique , Femelle , Humains , Hybridation fluorescente in situ , Nourrisson , Déficience intellectuelle/génétique , Mâle , Mégalencéphalie/génétique , Microcéphalie/génétique , Adulte d'âge moyen , Phénotype
5.
Mol Psychiatry ; 22(4): 625-633, 2017 04.
Article de Anglais | MEDLINE | ID: mdl-27166760

RÉSUMÉ

Contactin genes CNTN5 and CNTN6 code for neuronal cell adhesion molecules that promote neurite outgrowth in sensory-motor neuronal pathways. Mutations of CNTN5 and CNTN6 have previously been reported in individuals with autism spectrum disorders (ASDs), but very little is known on their prevalence and clinical impact. In this study, we identified CNTN5 and CNTN6 deleterious variants in individuals with ASD. Among the carriers, a girl with ASD and attention-deficit/hyperactivity disorder was carrying five copies of CNTN5. For CNTN6, both deletions (6/1534 ASD vs 1/8936 controls; P=0.00006) and private coding sequence variants (18/501 ASD vs 535/33480 controls; P=0.0005) were enriched in individuals with ASD. Among the rare CNTN6 variants, two deletions were transmitted by fathers diagnosed with ASD, one stop mutation CNTN6W923X was transmitted by a mother to her two sons with ASD and one variant CNTN6P770L was found de novo in a boy with ASD. Clinical investigations of the patients carrying CNTN5 or CNTN6 variants showed that they were hypersensitive to sounds (a condition called hyperacusis) and displayed changes in wave latency within the auditory pathway. These results reinforce the hypothesis of abnormal neuronal connectivity in the pathophysiology of ASD and shed new light on the genes that increase risk for abnormal sensory perception in ASD.


Sujet(s)
Perception auditive/génétique , Trouble du spectre autistique/génétique , Contactines/génétique , Adolescent , Adulte , Trouble déficitaire de l'attention avec hyperactivité/génétique , Trouble du spectre autistique/métabolisme , Enfant , Contactines/métabolisme , Variations de nombre de copies de segment d'ADN , Femelle , Prédisposition génétique à une maladie , Humains , Mâle , Mutation , Polymorphisme de nucléotide simple
6.
Clin Genet ; 91(4): 640-646, 2017 Apr.
Article de Anglais | MEDLINE | ID: mdl-27874174

RÉSUMÉ

〈 We report on an infant with Opitz trigonocephaly C syndrome (OTCS), who also had manifestations of ciliopathy, including short ribs (non-asphyxiating), trident acetabular roofs, postaxial polydactyly cone-shaped epiphyses, and dysplasia of the renal, hepatic and pancreatic tissues. To investigate the molecular cause, we used an exome sequencing strategy followed by Sanger sequencing. Two rare variants, both predicted to result in loss of functional protein, were identified in the IFT140 gene; a substitution at the splice donor site of exon 24 (c.723 + 1 G > T) and a 17 bp deletion, impacting the first coding exon (c.-11_6del). The variants were confirmed as being biallelic using Sanger sequencing, showing that the splice variant was inherited from the propositus mother and the deletion from the father. To date, Mainzer-Saldino syndrome, Jeune syndrome, and a form of nonsyndromic retinal dystrophy, have been identified as ciliopathies caused by IFT140 mutations. We provide the first description of an OTCS phenotype that appears to result from IFT140 mutations. The presentation of this patient is consistent with previous reports showing that OTCS already exhibited skeleletal and nonskeletal features of a ciliopathy.


Sujet(s)
Protéines de transport/génétique , Ciliopathies/génétique , Craniosynostoses/génétique , Prédisposition génétique à une maladie , Déficience intellectuelle/génétique , Ciliopathies/diagnostic , Ciliopathies/physiopathologie , Craniosynostoses/diagnostic , Craniosynostoses/physiopathologie , Exome/génétique , Femelle , Hétérozygote , Séquençage nucléotidique à haut débit , Humains , Déficience intellectuelle/diagnostic , Déficience intellectuelle/physiopathologie , Mâle , Pedigree , Sites d'épissage d'ARN/génétique , Délétion de séquence/génétique
7.
Mol Psychiatry ; 21(9): 1202-7, 2016 09.
Article de Anglais | MEDLINE | ID: mdl-26573769

RÉSUMÉ

A strong motivation for undertaking psychiatric gene discovery studies is to provide novel insights into unknown biology. Although attention-deficit hyperactivity disorder (ADHD) is highly heritable, and large, rare copy number variants (CNVs) contribute to risk, little is known about its pathogenesis and it remains commonly misunderstood. We assembled and pooled five ADHD and control CNV data sets from the United Kingdom, Ireland, United States of America, Northern Europe and Canada. Our aim was to test for enrichment of neurodevelopmental gene sets, implicated by recent exome-sequencing studies of (a) schizophrenia and (b) autism as a means of testing the hypothesis that common pathogenic mechanisms underlie ADHD and these other neurodevelopmental disorders. We also undertook hypothesis-free testing of all biological pathways. We observed significant enrichment of individual genes previously found to harbour schizophrenia de novo non-synonymous single-nucleotide variants (SNVs; P=5.4 × 10(-4)) and targets of the Fragile X mental retardation protein (P=0.0018). No enrichment was observed for activity-regulated cytoskeleton-associated protein (P=0.23) or N-methyl-D-aspartate receptor (P=0.74) post-synaptic signalling gene sets previously implicated in schizophrenia. Enrichment of ADHD CNV hits for genes impacted by autism de novo SNVs (P=0.019 for non-synonymous SNV genes) did not survive Bonferroni correction. Hypothesis-free testing yielded several highly significantly enriched biological pathways, including ion channel pathways. Enrichment findings were robust to multiple testing corrections and to sensitivity analyses that excluded the most significant sample. The findings reveal that CNVs in ADHD converge on biologically meaningful gene clusters, including ones now established as conferring risk of other neurodevelopmental disorders.


Sujet(s)
Trouble déficitaire de l'attention avec hyperactivité/génétique , Psychiatrie biologique/méthodes , Adolescent , Trouble autistique/génétique , Canada , Enfant , Enfant d'âge préscolaire , Variations de nombre de copies de segment d'ADN/génétique , Bases de données d'acides nucléiques , Europe , Femelle , Études d'associations génétiques/méthodes , Prédisposition génétique à une maladie/génétique , Étude d'association pangénomique , Humains , Irlande , Mâle , Troubles du développement neurologique/génétique , Polymorphisme de nucléotide simple/génétique , Schizophrénie/génétique , Royaume-Uni
9.
Mol Psychiatry ; 20(11): 1366-72, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-25421404

RÉSUMÉ

Significant evidence exists for the association between copy number variants (CNVs) and Autism Spectrum Disorder (ASD); however, most of this work has focused solely on the diagnosis of ASD. There is limited understanding of the impact of CNVs on the 'sub-phenotypes' of ASD. The objective of this paper is to evaluate associations between CNVs in differentially brain expressed (DBE) genes or genes previously implicated in ASD/intellectual disability (ASD/ID) and specific sub-phenotypes of ASD. The sample consisted of 1590 cases of European ancestry from the Autism Genome Project (AGP) with a diagnosis of an ASD and at least one rare CNV impacting any gene and a core set of phenotypic measures, including symptom severity, language impairments, seizures, gait disturbances, intelligence quotient (IQ) and adaptive function, as well as paternal and maternal age. Classification analyses using a non-parametric recursive partitioning method (random forests) were employed to define sets of phenotypic characteristics that best classify the CNV-defined groups. There was substantial variation in the classification accuracy of the two sets of genes. The best variables for classification were verbal IQ for the ASD/ID genes, paternal age at birth for the DBE genes and adaptive function for de novo CNVs. CNVs in the ASD/ID list were primarily associated with communication and language domains, whereas CNVs in DBE genes were related to broader manifestations of adaptive function. To our knowledge, this is the first study to examine the associations between sub-phenotypes and CNVs genome-wide in ASD. This work highlights the importance of examining the diverse sub-phenotypic manifestations of CNVs in ASD, including the specific features, comorbid conditions and clinical correlates of ASD that comprise underlying characteristics of the disorder.


Sujet(s)
Trouble du spectre autistique/génétique , Trouble du spectre autistique/physiopathologie , Variations de nombre de copies de segment d'ADN/génétique , Prédisposition génétique à une maladie/génétique , Phénotype , Adolescent , Adulte , Sujet âgé , Analyse de variance , Enfants handicapés , Femelle , Humains , Modèles logistiques , Mâle , Adulte d'âge moyen , Parents , Échelles d'évaluation en psychiatrie , Jeune adulte
10.
Clin Genet ; 88(3): 224-33, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-25131214

RÉSUMÉ

Studies of genomic copy number variants (CNVs) have identified genes associated with autism spectrum disorder (ASD) and intellectual disability (ID) such as NRXN1, SHANK2, SHANK3 and PTCHD1. Deletions have been reported in PTCHD1 however there has been little information available regarding the clinical presentation of these individuals. Herein we present 23 individuals with PTCHD1 deletions or truncating mutations with detailed phenotypic descriptions. The results suggest that individuals with disruption of the PTCHD1 coding region may have subtle dysmorphic features including a long face, prominent forehead, puffy eyelids and a thin upper lip. They do not have a consistent pattern of associated congenital anomalies or growth abnormalities. They have mild to moderate global developmental delay, variable degrees of ID, and many have prominent behavioral issues. Over 40% of subjects have ASD or ASD-like behaviors. The only consistent neurological findings in our cohort are orofacial hypotonia and mild motor incoordination. Our findings suggest that hemizygous PTCHD1 loss of function causes an X-linked neurodevelopmental disorder with a strong propensity to autistic behaviors. Detailed neuropsychological studies are required to better define the cognitive and behavioral phenotype.


Sujet(s)
Trouble du spectre autistique/diagnostic , Trouble du spectre autistique/génétique , Déficience intellectuelle/diagnostic , Déficience intellectuelle/génétique , Protéines membranaires/génétique , Mutation , Phénotype , Délétion de séquence , Adolescent , Adulte , Enfant , Enfant d'âge préscolaire , Exons , Faciès , Femelle , Humains , Nourrisson , Mâle , Jeune adulte
11.
Clin Genet ; 83(5): 399-407, 2013 May.
Article de Anglais | MEDLINE | ID: mdl-23425232

RÉSUMÉ

Autism spectrum disorders (ASDs) are a heterogeneous group of neurodevelopmental disorders affecting social communication, language and behavior. The underlying cause(s) in a given individual is often elusive, with the exception of clinically recognizable genetic syndromes with readily available molecular diagnosis, such as fragile X syndrome. Clinical geneticists approach patients with ASDs by ruling out known genetic and genomic syndromes, leaving more than 80% of families without a definitive diagnosis and an uncertain risk of recurrence. Advances in microarray technology and next-generation sequencing are revealing rare variants in genes with important roles in synapse formation, function and maintenance. This review will focus on the clinical approach to ASDs, given the current state of knowledge about their complex genetic architecture.


Sujet(s)
Troubles généralisés du développement de l'enfant/diagnostic , Troubles généralisés du développement de l'enfant/génétique , Enfant , Aberrations des chromosomes , Conseil génétique , Dépistage génétique , Humains , Analyse sur microréseau , Mutation , Phosphohydrolase PTEN/génétique , Récidive
12.
Mol Psychiatry ; 18(2): 183-9, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-22042228

RÉSUMÉ

Large, rare copy number variants (CNVs) have been implicated in a variety of psychiatric disorders, but the role of CNVs in recurrent depression is unclear. We performed a genome-wide analysis of large, rare CNVs in 3106 cases of recurrent depression, 459 controls screened for lifetime-absence of psychiatric disorder and 5619 unscreened controls from phase 2 of the Wellcome Trust Case Control Consortium (WTCCC2). We compared the frequency of cases with CNVs against the frequency observed in each control group, analysing CNVs over the whole genome, genic, intergenic, intronic and exonic regions. We found that deletion CNVs were associated with recurrent depression, whereas duplications were not. The effect was significant when comparing cases with WTCCC2 controls (P=7.7 × 10(-6), odds ratio (OR) =1.25 (95% confidence interval (CI) 1.13-1.37)) and to screened controls (P=5.6 × 10(-4), OR=1.52 (95% CI 1.20-1.93). Further analysis showed that CNVs deleting protein coding regions were largely responsible for the association. Within an analysis of regions previously implicated in schizophrenia, we found an overall enrichment of CNVs in our cases when compared with screened controls (P=0.019). We observe an ordered increase of samples with deletion CNVs, with the lowest proportion seen in screened controls, the next highest in unscreened controls and the highest in cases. This may suggest that the absence of deletion CNVs, especially in genes, is associated with resilience to recurrent depression.


Sujet(s)
Variations de nombre de copies de segment d'ADN/génétique , Trouble dépressif/génétique , Prédisposition génétique à une maladie , Loi du khi-deux , Études de cohortes , Femelle , Étude d'association pangénomique , Génotype , Humains , Mâle , Récidive
13.
Neurology ; 76(9): 774-80, 2011 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-21288980

RÉSUMÉ

OBJECTIVE: We investigated mitochondrial DNA (mtDNA) variants in children with a first episode of acquired demyelinating syndromes (PD-ADS) of the CNS and their relationship to disease phenotype, including subsequent diagnosis of multiple sclerosis (MS). METHODS: This exploratory analysis included the initial 213 children with PD-ADS in the prospective Canadian Pediatric Demyelinating Study and 166 matched healthy sibling controls from the Canadian Autism Genome Project. A total of 31 single nucleotide polymorphisms (SNPs) were analyzed, including haplogroup-defining SNPs and mtDNA variants previously reported to be associated with MS. RESULTS: Primary Leber hereditary optic neuropathy (LHON) mutations and other known pathogenic mtDNA mutations were absent in both patients with pediatric acquired demyelinating syndromes and controls. The 13708A haplogroup J-associated variant, previously linked to adult MS, was more frequent among subjects with PD-ADS (13.0%) compared to controls (6.2%; odds ratio [OR] 2.27; 95% confidence interval [CI] 1.06 to 4.83) and haplogroup M was associated with an earlier age at onset of PD-ADS (-1.74 years; 95% CI -3.33 to -0.07). In contrast, the haplogroup cluster UKJT, as well as 3 other SNPs, were each associated with a lower risk of PD-ADS. A total of 33 subjects with PD-ADS were diagnosed with MS during a mean follow-up period of 3.11 ± 1.14 (SD) years. No single SNP was associated with the risk of subsequent diagnosis of MS. However, haplogroup H was associated with an increased risk of MS (OR 2.60; 95% CI 1.21 to 5.55). CONCLUSION: These data suggest an association between mtDNA variants and the risk of PD-ADS and of a subsequent MS diagnosis. Replication of these findings in an independent population of subjects with PD-ADS is required.


Sujet(s)
ADN mitochondrial/génétique , Maladies démyélinisantes/diagnostic , Maladies démyélinisantes/génétique , Variation génétique/génétique , Haplotypes/génétique , Mutation/génétique , Adolescent , Adulte , Enfant , Enfant d'âge préscolaire , Études de cohortes , Femelle , Études de suivi , Humains , Nourrisson , Mâle , Polymorphisme de nucléotide simple/génétique , Études prospectives , Jeune adulte
14.
Clin Genet ; 80(5): 435-43, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21114665

RÉSUMÉ

We describe the identification and clinical presentation of four individuals from three unrelated families with hemizygous deletions involving the DPYD gene at chromosome 1p21.3. DPYD encodes dihydropyrimidine dehydrogenase, which is the initial and rate-limiting enzyme in the catabolism of pyrimidine bases. All four individuals described met diagnostic criteria for autism spectrum disorder with severe speech delay. Patient 1's deletion was originally reported in 2008, and more detailed clinical information is provided. Subsequently, this male individual was found to have a missense mutation in the X-linked PTCHD1 autism susceptibility gene, which may also contribute to the phenotype. Patients 2 and 3 are siblings with a novel deletion encompassing the DPYD gene. In their mother, the genomic region deleted from chromosome 1p21.3 was inserted into chromosome 10. A fourth proband had a novel 10-kb intragenic deletion of exon 6 of the DPYD gene detected on a higher resolution microarray. Our study suggests that hemizygous deletions involving the DPYD locus present with variable phenotypes which can include speech delay and autistic features, and may also be influenced by additional mutations in other genes, issues which need to be considered in genetic counseling.


Sujet(s)
Troubles généralisés du développement de l'enfant/génétique , Chromosomes humains de la paire 1/génétique , Dihydrouracil dehydrogenase (NADP)/génétique , Adolescent , Adulte , Enfant , Enfant d'âge préscolaire , Délétion de segment de chromosome , Chromosomes humains de la paire 10/génétique , Femelle , Humains , Mâle , Pedigree
15.
Clin Genet ; 78(5): 478-83, 2010 Nov.
Article de Anglais | MEDLINE | ID: mdl-20345473

RÉSUMÉ

To date, of 13 loci with linkage to non-syndromic autosomal recessive mental retardation (NS-ARMR), only six genes have been established with associated mutations. Here we present our study on NS-ARMR among the Pakistani population, where people are traditionally bound to marry within the family or the wider clan. In an exceptional, far-reaching genetic survey we have collected more than 50 consanguineous families exhibiting clinical symptoms/phenotypes of NS-ARMR. In the first step, nine families (MR2-9 and MR11) with multiple affected individuals were selected for molecular genetic studies. Two families (MR3, MR4) showed linkage to already know NS-ARMR loci. Fifteen affected and 10 unaffected individuals from six (MR2, MR6, MR7, MR8, MR9 and MR11) families were genotyped by using Affymetrix 5.0 or 6.0 single-nucleotide polymorphism (SNP) microarrays. SNP microarray data was visually inspected by dChip and genome-wide homozygosity analysis was performed by HomozygosityMapper. Additional mapping was performed (to exclude false-positive regions of homozygosity called by HomozygosityMapper and dChip) on all available affected and unaffected members in seven NS-ARMR families, using microsatellite markers. In this manner we were able to map three novel loci in seven different families originating from different areas of Pakistan. Two families (MR2, MR5) showed linkage on chromosome 2p25.3-p25.2. Three families (MR7, MR8, and MR9) that have been collected from the same village and belong to the same clan were mapped on chromosome 9q34.3. MR11 maps to a locus on 9p23-p13.3. Analysis of MR6 showed two positive loci, on chromosome 1q23.2-q23.3 and 8q24.21-q24.23. Genotyping in additional family members has so far narrowed, but not excluded the 1q locus. In summary, through this study we have identified three new loci for NS-ARMR, namely MRT14, 15 and 16.


Sujet(s)
Consanguinité , Déficience intellectuelle/génétique , Cartographie chromosomique , Femelle , Gènes récessifs , Étude d'association pangénomique , Humains , Mâle , Répétitions microsatellites , Pakistan , Pedigree , Polymorphisme de nucléotide simple
16.
Genomics ; 95(2): 73-83, 2010 Feb.
Article de Anglais | MEDLINE | ID: mdl-20005943

RÉSUMÉ

Recent research suggests that epigenetic alterations involving DNA methylation can be causative for neurodevelopmental, growth and metabolic disorders. Although lymphoblastoid cell lines have been an invaluable resource for the study of both genetic and epigenetic disorders, the impact of EBV transformation, cell culturing and freezing on epigenetic patterns is unknown. We compared genome-wide DNA methylation patterns of four white blood cell samples, four low-passage lymphoblastoid cell lines pre and post freezing and four high-passage lymphobastoid cell lines, using two microarray platforms: Illumina HumanMethylation27 platform containing 27,578 CpG sites and Agilent Human CpG island Array containing 27,800 CpG islands. Comparison of genome-wide methylation profiles between white blood cells and lymphoblastoid cell lines demonstrated methylation alterations in lymphoblastoid cell lines occurring at random genomic locations. These changes were more profound in high-passage cells. Freezing at low-passages did not have a significant effect on DNA methylation. Methylation changes were observed in several imprinted differentially methylated regions, including DIRAS3, NNAT, H19, MEG3, NDN and MKRN3, but not in known imprinting centers. Our results suggest that lymphoblastoid cell lines should be used with caution for the identification of disease-associated DNA methylation changes or for discovery of new imprinted genes, as the methylation patterns seen in these cell lines may not always be representative of DNA methylation present in the original B-lymphocytes of the patient.


Sujet(s)
Lymphocytes B/métabolisme , Lymphocytes B/virologie , Méthylation de l'ADN , Herpèsvirus humain de type 4/génétique , Transformation génétique , Techniques de culture cellulaire , Ilots CpG , Épigenèse génétique , Analyse de profil d'expression de gènes/méthodes , Génome humain , Humains
17.
Cytogenet Genome Res ; 123(1-4): 65-78, 2008.
Article de Anglais | MEDLINE | ID: mdl-19287140

RÉSUMÉ

Microdeletions of 3q29 have previously been reported, but the postulated reciprocal microduplication has only recently been observed. Here, cases from four families, two ascertained in Toronto (Canada) and one each from Edinburgh (UK) and Leiden (Netherlands), carrying microduplications of 3q29 are presented. These families have been characterized by cytogenetic and molecular techniques, and all individuals have been further characterized with genome-wide, high density single nucleotide polymorphism (SNP) arrays run at a single centre (The Centre for Applied Genomics, Toronto). In addition to polymorphic copy-number variants (CNV), all carry duplications of 3q29 ranging in size from 1.9 to 2.4 Mb, encompassing multiple genes and defining a minimum region of overlap of about 1.6 Mb bounded by clusters of segmental duplications that is remarkably similar in location to previously reported 3q29 microdeletions. Consistent with other reports, the phenotype is variable, although developmental delay and significant ophthalmological findings were recurrent, suggesting that dosage sensitivity of genes located within 3q29 is important for eye and CNS development. We also consider CNVs found elsewhere in the genome for their contribution to the phenotype. We conclude by providing preliminary guidelines for management and anticipatory care of families with this microduplication, thereby establishing a standard for CNV reporting.


Sujet(s)
Chromosomes humains/génétique , Dosage génique/génétique , Duplication de gène , Prédisposition génétique à une maladie/génétique , Femelle , Recommandations comme sujet , Humains , Mâle
19.
Cytogenet Genome Res ; 115(3-4): 205-14, 2006.
Article de Anglais | MEDLINE | ID: mdl-17124402

RÉSUMÉ

The discovery of an abundance of copy number variants (CNVs; gains and losses of DNA sequences >1 kb) and other structural variants in the human genome is influencing the way research and diagnostic analyses are being designed and interpreted. As such, comprehensive databases with the most relevant information will be critical to fully understand the results and have impact in a diverse range of disciplines ranging from molecular biology to clinical genetics. Here, we describe the development of bioinformatics resources to facilitate these studies. The Database of Genomic Variants (http://projects.tcag.ca/variation/) is a comprehensive catalogue of structural variation in the human genome. The database currently contains 1,267 regions reported to contain copy number variation or inversions in apparently healthy human cases. We describe the current contents of the database and how it can serve as a resource for interpretation of array comparative genomic hybridization (array CGH) and other DNA copy imbalance data. We also present the structure of the database, which was built using a new data modeling methodology termed Cross-Referenced Tables (XRT). This is a generic and easy-to-use platform, which is strong in handling textual data and complex relationships. Web-based presentation tools have been built allowing publication of XRT data to the web immediately along with rapid sharing of files with other databases and genome browsers. We also describe a novel tool named eFISH (electronic fluorescence in situ hybridization) (http://projects.tcag.ca/efish/), a BLAST-based program that was developed to facilitate the choice of appropriate clones for FISH and CGH experiments, as well as interpretation of results in which genomic DNA probes are used in hybridization-based experiments.


Sujet(s)
Biologie informatique/méthodes , Génome humain , Algorithmes , Cartographie chromosomique , Interprétation statistique de données , Bases de données génétiques , Techniques génétiques , Variation génétique , Humains , Internet , Hybridation d'acides nucléiques , Langages de programmation , Statistiques comme sujet , Interface utilisateur
20.
J Med Genet ; 43(5): 429-34, 2006 May.
Article de Anglais | MEDLINE | ID: mdl-16556609

RÉSUMÉ

INTRODUCTION: We describe the case of two brothers diagnosed with autism who both carry a paracentic inversion of the short arm of chromosome 4 (46,XY, inv(4)(p12-p15.3)). We have determined that this inversion is inherited from an apparently unaffected mother and unaffected maternal grandfather. Methods/ RESULTS: Using fluorescence in situ hybridisation analysis and Southern blot hybridisation we identified the breakpoints. The proximal breakpoint (4p12) maps to a region containing a cluster of gamma-aminobutyric acid A (GABA(A)) receptor genes, and directly interrupts the GABRG1 gene, the distal-most gene of the cluster. We also identified an insertion/deletion polymorphism for a approximately 2 kb LINE1 (L1) element that occurs within intron 7 of GABRG1. Our genotype analysis amongst autism families indicated that the L1 deletion allele did not show increased transmission to affected individuals. No linkage disequilibrium was evident between the L1 and single nucleotide polymorphisms in adjacent GABA(A) receptor genes on 4p, where a recent study has identified significant association with autism. DISCUSSION: Despite this, the identification of an inversion breakpoint disrupting GABRG1 provides solid support for the genetic involvement of the short arm of chromosome 4 in the genetic aetiology of autism, and for the hypothesis of disrupted GABA neurotransmission in autism.


Sujet(s)
Trouble autistique/génétique , Inversion chromosomique , Chromosomes humains de la paire 4 , Récepteurs GABA/génétique , Adulte , Trouble autistique/diagnostic , Enfant , Enfant d'âge préscolaire , Analyse de mutations d'ADN , Étiquettes de séquences exprimées , Femelle , Dépistage génétique , Humains , Nourrisson , Caryotypage , Mâle , Pedigree , Cartographie physique de chromosome
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...