Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 158(2): 263-276, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-24998929

ABSTRACT

Autism spectrum disorder (ASD) is a heterogeneous disease in which efforts to define subtypes behaviorally have met with limited success. Hypothesizing that genetically based subtype identification may prove more productive, we resequenced the ASD-associated gene CHD8 in 3,730 children with developmental delay or ASD. We identified a total of 15 independent mutations; no truncating events were identified in 8,792 controls, including 2,289 unaffected siblings. In addition to a high likelihood of an ASD diagnosis among patients bearing CHD8 mutations, characteristics enriched in this group included macrocephaly, distinct faces, and gastrointestinal complaints. chd8 disruption in zebrafish recapitulates features of the human phenotype, including increased head size as a result of expansion of the forebrain/midbrain and impairment of gastrointestinal motility due to a reduction in postmitotic enteric neurons. Our findings indicate that CHD8 disruptions define a distinct ASD subtype and reveal unexpected comorbidities between brain development and enteric innervation.


Subject(s)
Child Development Disorders, Pervasive/genetics , Child Development Disorders, Pervasive/physiopathology , DNA-Binding Proteins/genetics , Transcription Factors/genetics , Adolescent , Amino Acid Sequence , Animals , Brain/growth & development , Brain/pathology , Child , Child Development Disorders, Pervasive/classification , Child Development Disorders, Pervasive/pathology , Child, Preschool , DNA-Binding Proteins/metabolism , Female , Gastrointestinal Tract/innervation , Gastrointestinal Tract/physiopathology , Humans , Macaca mulatta , Male , Megalencephaly/pathology , Molecular Sequence Data , Mutation , Sequence Alignment , Transcription Factors/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
2.
Am J Hum Genet ; 110(12): 2015-2028, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37979581

ABSTRACT

We examined more than 97,000 families from four neurodevelopmental disease cohorts and the UK Biobank to identify phenotypic and genetic patterns in parents contributing to neurodevelopmental disease risk in children. We identified within- and cross-disorder correlations between six phenotypes in parents and children, such as obsessive-compulsive disorder (R = 0.32-0.38, p < 10-126). We also found that measures of sub-clinical autism features in parents are associated with several autism severity measures in children, including biparental mean Social Responsiveness Scale scores and proband Repetitive Behaviors Scale scores (regression coefficient = 0.14, p = 3.38 × 10-4). We further describe patterns of phenotypic similarity between spouses, where spouses show correlations for six neurological and psychiatric phenotypes, including a within-disorder correlation for depression (R = 0.24-0.68, p < 0.001) and a cross-disorder correlation between anxiety and bipolar disorder (R = 0.09-0.22, p < 10-92). Using a simulated population, we also found that assortative mating can lead to increases in disease liability over generations and the appearance of "genetic anticipation" in families carrying rare variants. We identified several families in a neurodevelopmental disease cohort where the proband inherited multiple rare variants in disease-associated genes from each of their affected parents. We further identified parental relatedness as a risk factor for neurodevelopmental disorders through its inverse relationship with variant pathogenicity and propose that parental relatedness modulates disease risk by increasing genome-wide homozygosity in children (R = 0.05-0.26, p < 0.05). Our results highlight the utility of assessing parent phenotypes and genotypes toward predicting features in children who carry rare variably expressive variants and implicate assortative mating as a risk factor for increased disease severity in these families.


Subject(s)
Autistic Disorder , Bipolar Disorder , Child , Humans , Virulence , Parents , Family , Autistic Disorder/genetics , Bipolar Disorder/genetics
3.
Hum Genet ; 138(2): 187-198, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30656450

ABSTRACT

Developmental and epileptic encephalopathies (DEEs) are genetically heterogenous conditions, often characterized by early onset, EEG interictal epileptiform abnormalities, polymorphous and drug-resistant seizures, and neurodevelopmental impairments. In this study, we investigated the genetic defects in two siblings who presented with severe DEE, microcephaly, spastic tetraplegia, diffuse brain hypomyelination, cerebellar atrophy, short stature, and kyphoscoliosis. Whole exome next-generation sequencing (WES) identified in both siblings a homozygous non-sense variant in the ACTL6B gene (NM_016188:c.820C>T;p.Gln274*) coding for a subunit of the neuron-specific chromatin remodeling complex nBAF. To further support these findings, a targeted ACTL6B sequencing assay was performed on a cohort of 85 unrelated DEE individuals, leading to the identification of a homozygous missense variant (NM_016188:c.1045G>A;p.Gly349Ser) in a patient. This variant did not segregate in the unaffected siblings in this family and was classified as deleterious by several prediction softwares. Interestingly, in both families, homozygous patients shared a rather homogeneous phenotype. Very few patients with ACTL6B gene variants have been sporadically reported in WES cohort studies of patients with neurodevelopmental disorders and/or congenital brain malformations. However, the limited number of patients with incomplete clinical information yet reported in the literature did not allow to establish a strong gene-disease association. Here, we provide additional genetic and clinical data on three new cases that support the pathogenic role of ACTL6B gene mutation in a syndromic form of DEE.


Subject(s)
Actins/genetics , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Genetic Diseases, Inborn/diagnostic imaging , Microcephaly/genetics , Neurodevelopmental Disorders/genetics , Quadriplegia/genetics , Spasms, Infantile/genetics , Child , Child, Preschool , Chromatin/genetics , DNA Methylation/genetics , Female , Genetic Diseases, Inborn/genetics , Humans , Infant , Infant, Newborn , Male , Microcephaly/diagnostic imaging , Neurodevelopmental Disorders/diagnostic imaging , Pedigree , Quadriplegia/diagnostic imaging , Spasms, Infantile/diagnostic imaging
4.
Genet Med ; 21(4): 816-825, 2019 04.
Article in English | MEDLINE | ID: mdl-30190612

ABSTRACT

PURPOSE: To assess the contribution of rare variants in the genetic background toward variability of neurodevelopmental phenotypes in individuals with rare copy-number variants (CNVs) and gene-disruptive variants. METHODS: We analyzed quantitative clinical information, exome sequencing, and microarray data from 757 probands and 233 parents and siblings who carry disease-associated variants. RESULTS: The number of rare likely deleterious variants in functionally intolerant genes ("other hits") correlated with expression of neurodevelopmental phenotypes in probands with 16p12.1 deletion (n=23, p=0.004) and in autism probands carrying gene-disruptive variants (n=184, p=0.03) compared with their carrier family members. Probands with 16p12.1 deletion and a strong family history presented more severe clinical features (p=0.04) and higher burden of other hits compared with those with mild/no family history (p=0.001). The number of other hits also correlated with severity of cognitive impairment in probands carrying pathogenic CNVs (n=53) or de novo pathogenic variants in disease genes (n=290), and negatively correlated with head size among 80 probands with 16p11.2 deletion. These co-occurring hits involved known disease-associated genes such as SETD5, AUTS2, and NRXN1, and were enriched for cellular and developmental processes. CONCLUSION: Accurate genetic diagnosis of complex disorders will require complete evaluation of the genetic background even after a candidate disease-associated variant is identified.


Subject(s)
Autistic Disorder/genetics , Cell Adhesion Molecules, Neuronal/genetics , Genetic Carrier Screening , Methyltransferases/genetics , Nerve Tissue Proteins/genetics , Proteins/genetics , Autistic Disorder/physiopathology , Calcium-Binding Proteins , Chromosomes, Human, Pair 16/genetics , Cognition/physiology , Cytoskeletal Proteins , DNA Copy Number Variations/genetics , Female , Gene Expression Regulation/genetics , Genetic Background , Humans , Male , Neural Cell Adhesion Molecules , Parents , Pedigree , Phenotype , Sequence Deletion/genetics , Siblings , Transcription Factors
5.
Am J Med Genet A ; 173(6): 1649-1655, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28407444

ABSTRACT

Deletion of 18q12.2 is an increasingly recognized condition with a distinct neuropsychiatric phenotype. Twenty-two patients have been described with overlapping neurobehavioral disturbances including developmental delay, intellectual disability of variable degree, seizures, motor coordination disorder, behavioral/emotional disturbances, and autism spectrum disorders. The CUGBP Elav-like family member 4 (CELF4) gene at 18q12.2 encodes a RNA-binding protein that links to RNA subsets involved in pre- and postsynaptic neurotransmission including almost 30% of potential autism-related genes. Haploinsufficiency of CELF4 was associated with an autism or autistic behavior diagnosis in two adult patients with de novo 18q12.2 deletions. We report on a girl and her mildly affected mother with a 275 kb deletion at 18q12.2 involving CELF4 and KIAA1328 whose disruption is not associated with any known disease. The child was diagnosed with syndromic intellectual disability and autism at 6 years of age. Her mother had minor dysmorphisms, mild intellectual disability, and autistic behavior. The deleted region reported in this family is one of the smallest so far reported at 18q12.2. This is also the first full clinical description of maternally inherited CELF4 haploinsufficiency. The present study refines the molecular and neuropsychiatric phenotype associated with 18q12.2 deletion leading to CELF4 haploinsufficiency and provides evidence for a role for CELF4 in brain development and autism spectrum disorders.


Subject(s)
Autism Spectrum Disorder/genetics , CELF Proteins/genetics , Developmental Disabilities/genetics , Intellectual Disability/genetics , Adult , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 18/genetics , Developmental Disabilities/physiopathology , Female , Haploinsufficiency/genetics , Humans , Intellectual Disability/physiopathology , Phenotype , RNA-Binding Proteins/genetics
6.
PLoS Genet ; 10(7): e1004491, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25010521

ABSTRACT

Ocular coloboma is a sight-threatening malformation caused by failure of the choroid fissure to close during morphogenesis of the eye, and is frequently associated with additional anomalies, including microphthalmia and cataracts. Although Hedgehog signaling is known to play a critical role in choroid fissure closure, genetic regulation of this pathway remains poorly understood. Here, we show that the transcription factor Sox11 is required to maintain specific levels of Hedgehog signaling during ocular development. Sox11-deficient zebrafish embryos displayed delayed and abnormal lens formation, coloboma, and a specific reduction in rod photoreceptors, all of which could be rescued by treatment with the Hedgehog pathway inhibitor cyclopamine. We further demonstrate that the elevated Hedgehog signaling in Sox11-deficient zebrafish was caused by a large increase in shha transcription; indeed, suppressing Shha expression rescued the ocular phenotypes of sox11 morphants. Conversely, over-expression of sox11 induced cyclopia, a phenotype consistent with reduced levels of Sonic hedgehog. We screened DNA samples from 79 patients with microphthalmia, anophthalmia, or coloboma (MAC) and identified two novel heterozygous SOX11 variants in individuals with coloboma. In contrast to wild type human SOX11 mRNA, mRNA containing either variant failed to rescue the lens and coloboma phenotypes of Sox11-deficient zebrafish, and both exhibited significantly reduced transactivation ability in a luciferase reporter assay. Moreover, decreased gene dosage from a segmental deletion encompassing the SOX11 locus resulted in microphthalmia and related ocular phenotypes. Therefore, our study reveals a novel role for Sox11 in controlling Hedgehog signaling, and suggests that SOX11 variants contribute to pediatric eye disorders.


Subject(s)
Coloboma/genetics , Embryonic Development/genetics , Hedgehog Proteins/biosynthesis , Hedgehog Proteins/genetics , SOXC Transcription Factors/genetics , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/genetics , Animals , Choroid Diseases/genetics , Choroid Diseases/metabolism , Choroid Diseases/pathology , Coloboma/metabolism , Coloboma/pathology , Embryo, Nonmammalian , Eye/growth & development , Eye/metabolism , Humans , Morphogenesis/genetics , RNA, Messenger/biosynthesis , SOXC Transcription Factors/biosynthesis , Signal Transduction/genetics , Zebrafish/genetics
7.
Nat Methods ; 10(9): 903-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23892896

ABSTRACT

Over 900 genes have been annotated within duplicated regions of the human genome, yet their functions and potential roles in disease remain largely unknown. One major obstacle has been the inability to accurately and comprehensively assay genetic variation for these genes in a high-throughput manner. We developed a sequencing-based method for rapid and high-throughput genotyping of duplicated genes using molecular inversion probes designed to target unique paralogous sequence variants. We applied this method to genotype all members of two gene families, SRGAP2 and RH, among a diversity panel of 1,056 humans. The approach could accurately distinguish copy number in paralogs having up to ∼99.6% sequence identity, identify small gene-disruptive deletions, detect single-nucleotide variants, define breakpoints of unequal crossover and discover regions of interlocus gene conversion. The ability to rapidly and accurately genotype multiple gene families in thousands of individuals at low cost enables the development of genome-wide gene conversion maps and 'unlocks' many previously inaccessible duplicated genes for association with human traits.


Subject(s)
GTPase-Activating Proteins/genetics , Gene Conversion , Genes, Duplicate , Genotyping Techniques/methods , High-Throughput Nucleotide Sequencing/methods , Gene Dosage , Genetic Variation , Genome, Human , Genotyping Techniques/economics , Homologous Recombination , Humans , Molecular Probes , Rh-Hr Blood-Group System/genetics
8.
J Hum Genet ; 61(2): 95-101, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26490184

ABSTRACT

Methyl-CpG-binding protein 2 (MeCP2) is a nuclear protein highly expressed in neurons that is involved in transcriptional modulation and chromatin remodeling. Mutations in MECP2 in females are associated with Rett syndrome, a neurological disorder characterized by a normal neonatal period, followed by the arrest of development and regression of acquired skills. Although it was initially thought that MECP2 pathogenic mutations in males were not compatible with life, starting from 1999 about 60 male patients have been identified and their phenotype varies from severe neonatal encephalopathy to mild intellectual disability. Targeted next-generation sequencing of a panel of intellectual disability related genes was performed on two unrelated male patients, and two missense variants in MECP2 were identified (p.Gly185Val and p.Arg167Trp). These variants lie outside the canonical methyl-CpG-binding domain and transcription repression domain domains, where the pathogenicity of missense variants is more difficult to establish. In both families, variants were found in all affected siblings and were inherited from the asymptomatic mother, showing skewed X-chromosome inactivation. We report here the first missense variant located in AT-hook domain 1 and we underline the importance of MECP2 substitutions outside the canonical MeCP2 domains in X-linked intellectual disability.


Subject(s)
Genetic Diseases, X-Linked/genetics , Intellectual Disability/genetics , Methyl-CpG-Binding Protein 2/genetics , Mutation, Missense , Adult , Child , Child, Preschool , Female , Genetic Diseases, X-Linked/diagnosis , Humans , Intellectual Disability/diagnosis , Male , Phenotype , Protein Domains/genetics
9.
Anticancer Drugs ; 27(7): 609-19, 2016 08.
Article in English | MEDLINE | ID: mdl-27101258

ABSTRACT

In a previous study, we reported the cytotoxic activity against various tumour cells of the peptidoglycan of Lactobacillus casei. To isolate the most active components, we performed column-chromatography separation of the peptidoglycan complex and tested the related fractions for their cytotoxic activity. The most active fractions were then lyophilized and the residue was analysed by gas chromatography for its amino acid content and composition. On the basis of the known chemical formula of the basic peptidic component of the peptidoglycan complex of L. casei, a peptide was then synthesized [Europ. (CH-DE-FR-GB) Patent number 1217005; IT number 01320177] and its cytotoxicity was tested against tumoural and normal cells. The synthetic peptide was found to impair the entire metabolism of cultured tumour cells and to restore the apoptotic process. By contrast, normal cells appeared to be stimulated rather than inhibited by the peptide, whereas primary mouse embryo fibroblasts behaved similarly to tumour cells. On the basis of these results, L. casei peptidoglycan fragments and their constituent basic peptide might be applicable as potent antitumour agents.


Subject(s)
Hexokinase/metabolism , Lacticaseibacillus casei/chemistry , Mitochondria/enzymology , Neoplasms/drug therapy , Peptide Fragments/isolation & purification , Peptide Fragments/pharmacology , Peptidoglycan/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Humans , K562 Cells , Male , Mice , Mitochondria/drug effects , Peptide Fragments/chemical synthesis , Peptidoglycan/pharmacology , Rats , Tumor Hypoxia
10.
Am J Med Genet A ; 167A(12): 3038-45, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26420380

ABSTRACT

The ability to identify the clinical nature of the recurrent duplication of chromosome 17q12 has been limited by its rarity and the diverse range of phenotypes associated with this genomic change. In order to further define the clinical features of affected patients, detailed clinical information was collected in the largest series to date (30 patients and 2 of their siblings) through a multi-institutional collaborative effort. The majority of patients presented with developmental delays varying from mild to severe. Though dysmorphic features were commonly reported, patients do not have consistent and recognizable features. Cardiac, ophthalmologic, growth, behavioral, and other abnormalities were each present in a subset of patients. The newly associated features potentially resulting from 17q12 duplication include height and weight above the 95th percentile, cataracts, microphthalmia, coloboma, astigmatism, tracheomalacia, cutaneous mosaicism, pectus excavatum, scoliosis, hypermobility, hypospadias, diverticulum of Kommerell, pyloric stenosis, and pseudohypoparathryoidism. The majority of duplications were inherited with some carrier parents reporting learning disabilities or microcephaly. We identified additional, potentially contributory copy number changes in a subset of patients, including one patient each with 16p11.2 deletion and 15q13.3 deletion. Our data further define and expand the clinical spectrum associated with duplications of 17q12 and provide support for the role of genomic modifiers contributing to phenotypic variability.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Duplication , Adolescent , Child , Child, Preschool , DNA Copy Number Variations , Developmental Disabilities/genetics , Face/abnormalities , Female , Humans , Infant , Male , Microcephaly/genetics , Phenotype , Young Adult
11.
Nat Genet ; 38(9): 999-1001, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16906164

ABSTRACT

Submicroscopic genomic copy number changes have been identified only recently as an important cause of mental retardation. We describe the detection of three interstitial, overlapping 17q21.31 microdeletions in a cohort of 1,200 mentally retarded individuals associated with a clearly recognizable clinical phenotype of mental retardation, hypotonia and a characteristic face. The deletions encompass the MAPT and CRHR1 genes and are associated with a common inversion polymorphism.


Subject(s)
Chromosome Deletion , Chromosome Inversion , Chromosomes, Human, Pair 17 , Polymorphism, Genetic , Adolescent , Adult , Brain/abnormalities , Brain/diagnostic imaging , Child, Preschool , Cohort Studies , Face/pathology , Female , Gene Dosage , Gene Frequency , Haplotypes , Humans , Intellectual Disability/epidemiology , Intellectual Disability/genetics , Intellectual Disability/pathology , Magnetic Resonance Imaging , Male , Muscle Hypotonia/genetics , Muscle Hypotonia/physiopathology , Physical Chromosome Mapping , Prevalence , Radiography , Receptors, Corticotropin-Releasing Hormone/genetics , Syndrome , tau Proteins/genetics
12.
Am J Hum Genet ; 89(4): 551-63, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21981781

ABSTRACT

Persons with neurodevelopmental disorders or autism spectrum disorder (ASD) often harbor chromosomal microdeletions, yet the individual genetic contributors within these regions have not been systematically evaluated. We established a consortium of clinical diagnostic and research laboratories to accumulate a large cohort with genetic alterations of chromosomal region 2q23.1 and acquired 65 subjects with microdeletion or translocation. We sequenced translocation breakpoints; aligned microdeletions to determine the critical region; assessed effects on mRNA expression; and examined medical records, photos, and clinical evaluations. We identified a single gene, methyl-CpG-binding domain 5 (MBD5), as the only locus that defined the critical region. Partial or complete deletion of MBD5 was associated with haploinsufficiency of mRNA expression, intellectual disability, epilepsy, and autistic features. Fourteen alterations, including partial deletions of noncoding regions not typically captured or considered pathogenic by current diagnostic screening, disrupted MBD5 alone. Expression profiles and clinical characteristics were largely indistinguishable between MBD5-specific alteration and deletion of the entire 2q23.1 interval. No copy-number alterations of MBD5 were observed in 7878 controls, suggesting MBD5 alterations are highly penetrant. We surveyed MBD5 coding variations among 747 ASD subjects compared to 2043 non-ASD subjects analyzed by whole-exome sequencing and detected an association with a highly conserved methyl-CpG-binding domain missense variant, p.79Gly>Glu (c.236G>A) (p = 0.012). These results suggest that genetic alterations of MBD5 cause features of 2q23.1 microdeletion syndrome and that this epigenetic regulator significantly contributes to ASD risk, warranting further consideration in research and clinical diagnostic screening and highlighting the importance of chromatin remodeling in the etiology of these complex disorders.


Subject(s)
Child Development Disorders, Pervasive/genetics , Chromosomes, Human, Pair 2 , DNA-Binding Proteins/genetics , Epilepsy/genetics , Gene Deletion , Intellectual Disability/genetics , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , CpG Islands , Epigenesis, Genetic , Female , Humans , Male , Phenotype , Syndrome
13.
Am J Med Genet A ; 164A(5): 1262-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24478251

ABSTRACT

Klippel-Trenaunay syndrome comprises congenital vascular malformations of the capillary (nevus flammeus), venous (varicosities) or lymphatic systems and disturbed (usually over-) growth of one or more extremities and adjacent parts of the trunk. In some individuals the affected body area may show reduced rather than increased growth. Such patients have been described inverse Klippel-Trenaunay syndrome and included within the spectrum of the syndrome. We report on a 3-year-old boy with vascular malformation of the nevus flammeus type extending from the right buttock to the sole of the right foot with clinical and radiological evidence of leg varicosities and underlying deficiency of the soft tissues and bone. In addition, he had macrodactyly of the first, second, and third toes with small nails, and cutaneous syndactyly of the second and third toes of the ipsilateral foot. Cranial magnetic resonance imaging showed high signal lesions in the peritrigonal areas with normal spinal images. This mosaic phenotype demonstrates that decreased and increased growth can coexist in the same body area of an individual with Klippel-Trenaunay syndrome.


Subject(s)
Klippel-Trenaunay-Weber Syndrome/diagnosis , Phenotype , Child, Preschool , Humans , Magnetic Resonance Imaging , Male
14.
Am J Med Genet A ; 164A(3): 828-33, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24458984

ABSTRACT

Interstitial deletion of 2q24.2 is a rarely described cytogenetic aberration in patients with intellectual disability (ID). Previously reported genotype-phenotype correlation identified a minimum deleted region of 2.65 Mb including 15 genes. Recently, a patient with a de novo 2q24.2 microdeletion of 0.4 Mb encompassing only three genes was described. However, the precise relationship between most deleted genes and the clinical features remains unclear. Here we describe a 12-year-old male patient diagnosed with growth retardation and ID. He also showed microcephaly, right palpebral ptosis, scapular winging, and pectus excavatum. Single nucleotide polymorphisms (SNP) array analysis showed a de novo interstitial deletion of 0.122 Mb at 2q24.2 region harboring only TBR1 (T-box, brain, 1; OMIM: 604616), which encodes a T-box family transcription factor expressed in post-mitotic projection neurons and functionally significant in embryologic corticogenesis. This is the first case of a deletion at 2q24.2 involving only TBR1. This finding narrows the smallest region of overlap (SRO) for deletions in this region and strengthens the previously suggested hypothesis that this gene is a strong candidate for the ID phenotype. The identification of TBR1 as candidate for ID encourages further molecular studies to identify novel mutations to understand the pathogenic effects of its haploinsufficiency. Finally, this report provides a review on 10 2q24.2 microdeletion patients.


Subject(s)
Chromosomes, Human, Pair 2 , Intellectual Disability/diagnosis , Intellectual Disability/genetics , T-Box Domain Proteins/genetics , Child, Preschool , Comparative Genomic Hybridization , Genetic Association Studies , Humans , Male , Polymorphism, Single Nucleotide
15.
Am J Med Genet A ; 164A(11): 2843-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25251717

ABSTRACT

Microdeletions of the 5q11.2 region are rare; in literature only two patients with a deletion in this region have been reported so far. In this study, we describe four additional patients and further define this new 5q11.2 microdeletion syndrome. A comparison of the features observed in all six patients with overlapping 5q11.2 deletions showed a phenotypic spectrum that overlaps with CHARGE syndrome and 22q11.2 deletion syndrome including choanal atresia, developmental delay, heart defects, external ear abnormalities, and short stature. No colobomas or abnormalities of semicircular canals and olfactory nerves were reported. Two male patients had genital abnormalities. We estimated a 2.0 Mb (53.0-55.0 Mb) Shortest Region of Overlap (SRO) for the main clinical characteristics of the syndrome. This region contains nine genes and two non-coding microRNAs. In this region DHX29 serves as the candidate gene as it encodes an ATP-dependent RNA-helicase that is involved in the initiation of RNA translation. Screening a small cohort of 14 patients who presented the main features, however, did not reveal any pathogenic abnormalities of DHX29.


Subject(s)
22q11 Deletion Syndrome/diagnosis , 22q11 Deletion Syndrome/genetics , CHARGE Syndrome/diagnosis , CHARGE Syndrome/genetics , Chromosome Disorders/diagnosis , Chromosome Disorders/genetics , Phenotype , Adolescent , Adult , Child , Child, Preschool , Chromosome Deletion , Chromosome Mapping , Chromosomes, Human, Pair 5 , Comparative Genomic Hybridization , Diagnosis, Differential , Facies , Female , Genetic Association Studies , Humans , Infant , Male , Young Adult
16.
Am J Med Genet A ; 164A(8): 1923-30, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24733578

ABSTRACT

Typical Xq25 duplications are large and associated with heterogeneous phenotypes. Recently, small duplications involving this genomic region and encompassing the GRIA3 and STAG2 genes have been reported. These Xq25 microduplications are associated with a recognizable syndrome including intellectual disability and distinctive facial appearance. We report on Xq25 microduplications in two unrelated families identified by array comparative genomic hybridization. In both families, the genomic imbalances segregated with the disease in male individuals, while the phenotypes of the heterozygous females appeared to be modulated by their X-inactivation pattern. These rearrangements of about 600 kb involved only three genes: THOC2, XIAP, and STAG2. Further characterization by FISH analyses showed tandem duplication in the Xq25 locus of these genes. These data refine the Xq25 candidate region, identifying a minimal duplicated region of about 270 kb encompassing the XIAP and STAG2 genes. We discuss the function of the genes in the rearrangements and their involvement in the pathogenesis of this disorder.


Subject(s)
Antigens, Nuclear/genetics , Chromosome Duplication , Trisomy/diagnosis , Trisomy/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics , Adolescent , Adult , Aged , Brain/pathology , Cell Cycle Proteins , Child , Child, Preschool , Chromosome Breakpoints , Chromosome Mapping , Chromosomes, Human, X/genetics , Comparative Genomic Hybridization , Exons , Facies , Female , Genetic Association Studies , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Pedigree , Phenotype , Sex Chromosome Aberrations , Syndrome , Young Adult
17.
Am J Med Genet A ; 164A(7): 1666-76, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24700646

ABSTRACT

Phelan-McDermid syndrome (22q13.3 deletion syndrome) is a contiguous gene disorder resulting from the deletion of the distal long arm of chromosome 22. SHANK3, a gene within the minimal critical region, is a candidate gene for the major neurological features of this syndrome. We report clinical and molecular data from a study of nine patients with overlapping interstitial deletions in 22q13 not involving SHANK3. All of these deletions overlap with the largest, but not with the smallest deletion associated with Phelan-McDermid syndrome. The deletion sizes and breakpoints varied considerably among our patients, with the largest deletion spanning 6.9 Mb and the smallest deletion spanning 2.7 Mb. Eight out of nine patients had a de novo deletion, while in one patient the origin of deletion was unknown. These patients shared clinical features common to Phelan-McDermid syndrome: developmental delay (11/12), speech delay (11/12), hypotonia (9/12), and feeding difficulties (7/12). Moreover, the majority of patients (8/12) exhibited macrocephaly. In the minimal deleted region, we identified two candidate genes, SULT4A1 and PARVB (associated with the PTEN pathway), which could be associated in our cohort with neurological features and macrocephaly/hypotonia, respectively. This study suggests that the haploinsufficiency of genes in the 22q13 region beside SHANK3 contributes to cognitive and speech development, and that these genes are involved in the phenotype associated with the larger Phelan-McDermid syndrome 22q13 deletions. Moreover, because the deletions in our patients do not involve the SHANK3 gene, we posit the existence of a new contiguous gene syndrome proximal to the smallest terminal deletions in the 22q13 region.


Subject(s)
Chromosome Deletion , Chromosome Disorders/diagnosis , Chromosome Disorders/genetics , Chromosomes, Human, Pair 22 , Nerve Tissue Proteins/genetics , Child , Child, Preschool , Chromosomes, Human, Pair 22/genetics , Comparative Genomic Hybridization , Diagnosis, Differential , Facies , Female , Humans , Infant , Male , Phenotype , Syndrome
18.
J Med Genet ; 50(12): 802-11, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24123876

ABSTRACT

BACKGROUND: Intellectual disability (ID) is a common neurodevelopmental disorder affecting 1-3% of the general population. Mutations in more than 10% of all human genes are considered to be involved in this disorder, although the majority of these genes are still unknown. OBJECTIVES: We investigated 19 small non-consanguineous families with two to five affected siblings in order to identify pathogenic gene variants in known, novel and potential ID candidate genes. Non-consanguineous families have been largely ignored in gene identification studies as small family size precludes prior mapping of the genetic defect. METHODS AND RESULTS: Using exome sequencing, we identified pathogenic mutations in three genes, DDHD2, SLC6A8, and SLC9A6, of which the latter two have previously been implicated in X-linked ID phenotypes. In addition, we identified potentially pathogenic mutations in BCORL1 on the X-chromosome and in MCM3AP, PTPRT, SYNE1, and ZNF528 on autosomes. CONCLUSIONS: We show that potentially pathogenic gene variants can be identified in small, non-consanguineous families with as few as two affected siblings, thus emphasising their value in the identification of syndromic and non-syndromic ID genes.


Subject(s)
Exome/genetics , Intellectual Disability/genetics , Mutation/genetics , DNA Mutational Analysis , Family , Female , Humans , Male , Pedigree
19.
PLoS Genet ; 7(11): e1002334, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22102821

ABSTRACT

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).


Subject(s)
Autistic Disorder/genetics , DNA Copy Number Variations/genetics , Dyslexia/genetics , Intellectual Disability/genetics , Neurogenesis/immunology , Adolescent , Autistic Disorder/diagnosis , Autistic Disorder/pathology , Child , Comparative Genomic Hybridization/methods , Cytoskeletal Proteins , Dyslexia/diagnosis , Dyslexia/pathology , Endopeptidases/genetics , Female , Forkhead Transcription Factors/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Intellectual Disability/diagnosis , Intellectual Disability/pathology , Male , Nervous System Malformations/genetics , Nervous System Malformations/pathology , Neurogenesis/genetics , Phenotype , Proteins/genetics , Repressor Proteins/genetics , Sequence Deletion/genetics , Transcription Factors
20.
PLoS Genet ; 7(7): e1002173, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21779178

ABSTRACT

In this study, we used deletions at 22q13, which represent a substantial source of human pathology (Phelan/McDermid syndrome), as a model for investigating the molecular mechanisms of terminal deletions that are currently poorly understood. We characterized at the molecular level the genomic rearrangement in 44 unrelated patients with 22q13 monosomy resulting from simple terminal deletions (72%), ring chromosomes (14%), and unbalanced translocations (7%). We also discovered interstitial deletions between 17-74 kb in 9% of the patients. Haploinsufficiency of the SHANK3 gene, confirmed in all rearrangements, is very likely the cause of the major neurological features associated with PMS. SHANK3 mutations can also result in language and/or social interaction disabilities. We determined the breakpoint junctions in 29 cases, providing a realistic snapshot of the variety of mechanisms driving non-recurrent deletion and repair at chromosome ends. De novo telomere synthesis and telomere capture are used to repair terminal deletions; non-homologous end-joining or microhomology-mediated break-induced replication is probably involved in ring 22 formation and translocations; non-homologous end-joining and fork stalling and template switching prevail in cases with interstitial 22q13.3. For the first time, we also demonstrated that distinct stabilizing events of the same terminal deletion can occur in different early embryonic cells, proving that terminal deletions can be repaired by multistep healing events and supporting the recent hypothesis that rare pathogenic germline rearrangements may have mitotic origin. Finally, the progressive clinical deterioration observed throughout the longitudinal medical history of three subjects over forty years supports the hypothesis of a role for SHANK3 haploinsufficiency in neurological deterioration, in addition to its involvement in the neurobehavioral phenotype of PMS.


Subject(s)
Chromosome Deletion , Chromosome Disorders/genetics , Chromosomes, Human, Pair 22/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Base Sequence , Child , Child, Preschool , Comparative Genomic Hybridization , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Molecular Sequence Data , Parents , Ring Chromosomes , Sequence Deletion/genetics , Translocation, Genetic , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL