Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Hum Mol Genet ; 31(19): 3325-3340, 2022 09 29.
Article in English | MEDLINE | ID: mdl-35604360

ABSTRACT

Intellectual disability (ID) is a neurodevelopmental disorder frequently caused by monogenic defects. In this study, we collected 14 SEMA6B heterozygous variants in 16 unrelated patients referred for ID to different centers. Whereas, until now, SEMA6B variants have mainly been reported in patients with progressive myoclonic epilepsy, our study indicates that the clinical spectrum is wider and also includes non-syndromic ID without epilepsy or myoclonus. To assess the pathogenicity of these variants, selected mutated forms of Sema6b were overexpressed in Human Embryonic Kidney 293T (HEK293T) cells and in primary neuronal cultures. shRNAs targeting Sema6b were also used in neuronal cultures to measure the impact of the decreased Sema6b expression on morphogenesis and synaptogenesis. The overexpression of some variants leads to a subcellular mislocalization of SEMA6B protein in HEK293T cells and to a reduced spine density owing to loss of mature spines in neuronal cultures. Sema6b knockdown also impairs spine density and spine maturation. In addition, we conducted in vivo rescue experiments in chicken embryos with the selected mutated forms of Sema6b expressed in commissural neurons after knockdown of endogenous SEMA6B. We observed that expression of these variants in commissural neurons fails to rescue the normal axon pathway. In conclusion, identification of SEMA6B variants in patients presenting with an overlapping phenotype with ID and functional studies highlight the important role of SEMA6B in neuronal development, notably in spine formation and maturation and in axon guidance. This study adds SEMA6B to the list of ID-related genes.


Subject(s)
Epilepsy , Intellectual Disability , Semaphorins , Animals , Axon Guidance , Chick Embryo , Dendritic Spines , Epilepsy/genetics , HEK293 Cells , Humans , Intellectual Disability/genetics , Semaphorins/genetics
2.
Am J Hum Genet ; 108(5): 951-961, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33894126

ABSTRACT

The collapsin response mediator protein (CRMP) family proteins are intracellular mediators of neurotrophic factors regulating neurite structure/spine formation and are essential for dendrite patterning and directional axonal pathfinding during brain developmental processes. Among this family, CRMP5/DPYSL5 plays a significant role in neuronal migration, axonal guidance, dendrite outgrowth, and synapse formation by interacting with microtubules. Here, we report the identification of missense mutations in DPYSL5 in nine individuals with brain malformations, including corpus callosum agenesis and/or posterior fossa abnormalities, associated with variable degrees of intellectual disability. A recurrent de novo p.Glu41Lys variant was found in eight unrelated patients, and a p.Gly47Arg variant was identified in one individual from the first family reported with Ritscher-Schinzel syndrome. Functional analyses of the two missense mutations revealed impaired dendritic outgrowth processes in young developing hippocampal primary neuronal cultures. We further demonstrated that these mutations, both located in the same loop on the surface of DPYSL5 monomers and oligomers, reduced the interaction of DPYSL5 with neuronal cytoskeleton-associated proteins MAP2 and ßIII-tubulin. Our findings collectively indicate that the p.Glu41Lys and p.Gly47Arg variants impair DPYSL5 function on dendritic outgrowth regulation by preventing the formation of the ternary complex with MAP2 and ßIII-tubulin, ultimately leading to abnormal brain development. This study adds DPYSL5 to the list of genes implicated in brain malformation and in neurodevelopmental disorders.


Subject(s)
Agenesis of Corpus Callosum/genetics , Cerebellum/abnormalities , Mutation, Missense/genetics , Neurodevelopmental Disorders/genetics , Adult , Agenesis of Corpus Callosum/diagnostic imaging , Cerebellum/diagnostic imaging , Child , Child, Preschool , Female , Humans , Hydrolases/chemistry , Hydrolases/genetics , Intellectual Disability/diagnostic imaging , Intellectual Disability/genetics , Male , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Models, Molecular , Neurodevelopmental Disorders/diagnostic imaging , Tubulin/metabolism , Young Adult
3.
Hum Mutat ; 42(7): 848-861, 2021 07.
Article in English | MEDLINE | ID: mdl-33856728

ABSTRACT

The X-linked PTCHD1 gene, encoding a synaptic membrane protein, has been involved in neurodevelopmental disorders with the description of deleterious genomic microdeletions or truncating coding mutations. Missense variants were also identified, however, without any functional evidence supporting their pathogenicity level. We investigated 13 missense variants of PTCHD1, including eight previously described (c.152G>A,p.(Ser51Asn); c.217C>T,p.(Leu73Phe); c.517A>G,p.(Ile173Val); c.542A>C,p.(Lys181Thr); c.583G>A,p.(Val195Ile); c.1076A>G,p.(His359Arg); c.1409C>A,p.(Ala470Asp); c.1436A>G,p.(Glu479Gly)), and five novel ones (c.95C>T,p.(Pro32Leu); c.95C>G,p.(Pro32Arg); c.638A>G,p.(Tyr213Cys); c.898G>C,p.(Gly300Arg); c.928G>C,p.(Ala310Pro)) identified in male patients with intellectual disability (ID) and/or autism spectrum disorder (ASD). Interestingly, several of these variants involve amino acids localized in structural domains such as transmembrane segments. To evaluate their potentially deleterious impact on PTCHD1 protein function, we performed in vitro overexpression experiments of the wild-type and mutated forms of PTCHD1-GFP in HEK 293T and in Neuro-2a cell lines as well as in mouse hippocampal primary neuronal cultures. We found that six variants impaired the expression level of the PTCHD1 protein, and were retained in the endoplasmic reticulum suggesting abnormal protein folding. Our functional analyses thus provided evidence of the pathogenic impact of missense variants in PTCHD1, which reinforces the involvement of the PTCHD1 gene in ID and in ASD.


Subject(s)
Autism Spectrum Disorder , Intellectual Disability , Membrane Proteins , Animals , Autism Spectrum Disorder/genetics , Cell Membrane/metabolism , Humans , Intellectual Disability/genetics , Male , Membrane Proteins/genetics , Mice , Mutation, Missense
4.
Hum Genet ; 140(6): 885-896, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33417013

ABSTRACT

The 22q11.2 deletion syndrome (22q11DS) is associated with a wide spectrum of cognitive and psychiatric symptoms. Despite the considerable work performed over the past 20 years, the genetic etiology of the neurodevelopmental phenotype remains speculative. Here, we report de novo heterozygous truncating variants in the HIRA (Histone cell cycle regulation defective, S. Cerevisiae, homolog of, A) gene associated with a neurodevelopmental disorder in two unrelated patients. HIRA is located within the commonly deleted region of the 22q11DS and encodes a histone chaperone that regulates neural progenitor proliferation and neurogenesis, and that belongs to the WD40 Repeat (WDR) protein family involved in brain development and neuronal connectivity. To address the specific impact of HIRA haploinsufficiency in the neurodevelopmental phenotype of 22q11DS, we combined Hira knock-down strategies in developing mouse primary hippocampal neurons, and the direct study of brains from heterozygous Hira+/- mice. Our in vitro analyses revealed that Hira gene is mostly expressed during neuritogenesis and early dendritogenesis stages in mouse total brain and in developing primary hippocampal neurons. Moreover, shRNA knock-down experiments showed that a twofold decrease of endogenous Hira expression level resulted in an impaired dendritic growth and branching in primary developing hippocampal neuronal cultures. In parallel, in vivo analyses demonstrated that Hira+/- mice displayed subtle neuroanatomical defects including a reduced size of the hippocampus, the fornix and the corpus callosum. Our results suggest that HIRA haploinsufficiency would likely contribute to the complex pathophysiology of the neurodevelopmental phenotype of 22q11DS by impairing key processes in neurogenesis and by causing neuroanatomical defects during cerebral development.


Subject(s)
Cell Cycle Proteins/genetics , DiGeorge Syndrome/genetics , Haploinsufficiency , Histone Chaperones/genetics , Neurodevelopmental Disorders/genetics , Neuronal Plasticity/genetics , Neurons/metabolism , Transcription Factors/genetics , Animals , Base Sequence , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/metabolism , Child , Child, Preschool , Corpus Callosum/metabolism , Corpus Callosum/pathology , DiGeorge Syndrome/metabolism , DiGeorge Syndrome/pathology , Female , Fornix, Brain/metabolism , Fornix, Brain/pathology , Gene Expression , Heterozygote , Hippocampus/metabolism , Hippocampus/pathology , Histone Chaperones/antagonists & inhibitors , Histone Chaperones/deficiency , Histone Chaperones/metabolism , Humans , Mice , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Neurogenesis/genetics , Neurons/pathology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/deficiency , Transcription Factors/metabolism
5.
Hum Mutat ; 41(9): 1615-1628, 2020 09.
Article in English | MEDLINE | ID: mdl-32579715

ABSTRACT

Serine biosynthesis disorders comprise a spectrum of very rare autosomal recessive inborn errors of metabolism with wide phenotypic variability. Neu-Laxova syndrome represents the most severe expression and is characterized by multiple congenital anomalies and pre- or perinatal lethality. Here, we present the mutation spectrum and a detailed phenotypic analysis in 15 unrelated families with severe types of serine biosynthesis disorders. We identified likely disease-causing variants in the PHGDH and PSAT1 genes, several of which have not been reported previously. Phenotype analysis and a comprehensive review of the literature corroborates the evidence that serine biosynthesis disorders represent a continuum with varying degrees of phenotypic expression and suggest that even gradual differences at the severe end of the spectrum may be correlated with particular genotypes. We postulate that the individual residual enzyme activity of mutant proteins is the major determinant of the phenotypic variability, but further functional studies are needed to explore effects at the enzyme protein level.


Subject(s)
Abnormalities, Multiple/genetics , Brain Diseases/genetics , Fetal Growth Retardation/genetics , Genetic Association Studies , Ichthyosis/genetics , Limb Deformities, Congenital/genetics , Microcephaly/genetics , Phosphoglycerate Dehydrogenase/genetics , Transaminases/genetics , Female , Fetus , Humans , Infant, Newborn , Male , Mutation , Serine/biosynthesis
6.
Hum Mutat ; 39(6): 790-805, 2018 06.
Article in English | MEDLINE | ID: mdl-29637653

ABSTRACT

Simpson-Golabi-Behmel syndrome (SGBS) is an X-linked multiple congenital anomalies and overgrowth syndrome caused by a defect in the glypican-3 gene (GPC3). Until now, GPC3 mutations have been reported in isolated cases or small series and the global genotypic spectrum of these mutations has never been delineated. In this study, we review the 57 previously described GPC3 mutations and significantly expand this mutational spectrum with the description of 29 novel mutations. Compiling our data and those of the literature, we provide an overview of 86 distinct GPC3 mutations identified in 120 unrelated families, ranging from single nucleotide variations to complex genomic rearrangements and dispersed throughout the entire coding region of GPC3. The vast majority of them are deletions or truncating mutations (frameshift, nonsense mutations) predicted to result in a loss-of-function. Missense mutations are rare and the two which were functionally characterized, impaired GPC3 function by preventing GPC3 cleavage and cell surface addressing respectively. This report by describing for the first time the wide mutational spectrum of GPC3 could help clinicians and geneticists in interpreting GPC3 variants identified incidentally by high-throughput sequencing technologies and also reinforces the need for functional validation of non-truncating mutations (missense, in frame mutations, duplications).


Subject(s)
Arrhythmias, Cardiac/genetics , Genes, X-Linked/genetics , Genetic Diseases, X-Linked/genetics , Gigantism/genetics , Glypicans/genetics , Heart Defects, Congenital/genetics , Intellectual Disability/genetics , Arrhythmias, Cardiac/pathology , Codon, Nonsense/genetics , Female , Frameshift Mutation/genetics , Genetic Diseases, X-Linked/pathology , Gigantism/pathology , Heart Defects, Congenital/pathology , Humans , Intellectual Disability/pathology , Male , Pedigree , Phenotype
7.
Prenat Diagn ; 38(13): 1111-1119, 2018 12.
Article in English | MEDLINE | ID: mdl-30328630

ABSTRACT

OBJECTIVE: The aim of this study is to evaluate the diagnostic utility of prenatal diagnosis using the chromosomal microarray analysis (CMA) for fetuses presenting with isolated or associated intrauterine growth restriction (IUGR). METHOD: We retrospectively included all fetuses with IUGR referred for prenatal testing and studied by rapid fluorescence in situ hybridization (FISH), karyotype, and CMA. RESULTS: Among the 162 IUGR fetuses (78 associated and 84 isolated IUGR) included, 15 had an abnormal FISH result: 10 associated and five isolated fetal IUGRs. Among the 143 fetuses studied by CMA, 10 (7%) presented pathogenic copy number variations (CNVs). All 10 were in the associated fetal IUGR group (10/65 or 15.4%; 95% confidence interval [CI]: 8.4%-26.2%) versus 0/78 in the isolated fetal IUGR group (95% CI: 0%-5.6%). Six fetuses (4.2%) carried variants of unknown significance (VOUS) (three associated and three isolated fetal IUGRs). CONCLUSION: Our study highlights the added value of CMA in the case of associated fetal IUGR with an incremental yield of 6.1% (4/65) over karyotyping. No pathogenic CNVs were reported in the isolated fetal IUGR group. More studies must be conducted to determine when and whether CMA would be wisely indicated in this population.


Subject(s)
Comparative Genomic Hybridization/methods , Fetal Growth Retardation/genetics , In Situ Hybridization, Fluorescence , Karyotyping , Microarray Analysis/methods , Adult , DNA Copy Number Variations , Female , Humans , Karyotype , Pregnancy , Prenatal Diagnosis , Retrospective Studies , Young Adult
8.
J Hum Genet ; 61(8): 693-9, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27193221

ABSTRACT

Otopalatodigital spectrum disorders (OPDSD) constitute a group of dominant X-linked osteochondrodysplasias including four syndromes: otopalatodigital syndromes type 1 and type 2 (OPD1 and OPD2), frontometaphyseal dysplasia, and Melnick-Needles syndrome. These syndromes variably associate specific facial and extremities features, hearing loss, cleft palate, skeletal dysplasia and several malformations, and show important clinical overlap over the different entities. FLNA gain-of-function mutations were identified in these conditions. FLNA encodes filamin A, a scaffolding actin-binding protein. Here, we report phenotypic descriptions and molecular results of FLNA analysis in a large series of 27 probands hypothesized to be affected by OPDSD. We identified 11 different missense mutations in 15 unrelated probands (n=15/27, 56%), of which seven were novel, including one of unknown significance. Segregation analyses within families made possible investigating 20 additional relatives carrying a mutation. This series allows refining the phenotypic and mutational spectrum of FLNA mutations causing OPDSD, and providing suggestions to avoid the overdiagnosis of OPD1.


Subject(s)
Craniofacial Abnormalities/diagnosis , Craniofacial Abnormalities/genetics , Genetic Association Studies , Hand Deformities, Congenital/diagnosis , Hand Deformities, Congenital/genetics , Mutation , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/genetics , Phenotype , Alleles , Amino Acid Substitution , Exons , Facies , Female , Filamins/genetics , Humans , Male , Pedigree , Sequence Analysis, DNA
10.
Am J Med Genet A ; 164A(8): 1965-75, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24782328

ABSTRACT

Syndromic obesity is defined by the association of obesity with one or more feature(s) including developmental delay, dysmorphic traits, and/or congenital malformations. Over 25 syndromic forms of obesity have been identified. However, most cases remain of unknown etiology. The aim of this study was to identify new candidate loci associated with syndromic obesity to find new candidate genes and to better understand molecular mechanisms involved in this pathology. We performed oligonucleotide microarray-based comparative genomic hybridization in a cohort of 100 children presenting with syndromic obesity of unknown etiology, after exhaustive clinical, biological, and molecular studies. Chromosomal copy number variations were detected in 42% of the children in our cohort, with 23% of patients with potentially pathogenic copy number variants. Our results support that chromosomal rearrangements are frequently associated with syndromic obesity with a variety of contributory genes having relevance to either obesity or developmental delay. A list of inherited or apparently de novo duplications and deletions including their enclosed genes and not previously linked to syndromic obesity was established. Proteins encoded by several of these genes are involved in lipid metabolism (ACOXL, MSMO1, MVD, and PDZK1) linked with nervous system function (BDH1 and LINGO2), neutral lipid storage (PLIN2), energy homeostasis and metabolic processes (CDH13, CNTNAP2, CPPED1, NDUFA4, PTGS2, and SOCS6).


Subject(s)
Obesity/diagnosis , Obesity/genetics , Phenotype , Quantitative Trait Loci , Child , Child, Preschool , Chromosome Aberrations , Chromosome Disorders/diagnosis , Chromosome Disorders/genetics , Comparative Genomic Hybridization , DNA Copy Number Variations , Female , Gene Expression , Genetic Association Studies , Genome-Wide Association Study , Genomics , Humans , Infant , Male , Syndrome
12.
Mol Genet Metab ; 110(1-2): 90-7, 2013.
Article in English | MEDLINE | ID: mdl-23920044

ABSTRACT

Congenital deletions at the 3q13.31 locus have been recently described as a novel microdeletion syndrome characterized by developmental delay, postnatal overgrowth, hypoplastic male genitalia and characteristic facial features. A common critical region of overlapping of 580kb was delineated including two strong candidate genes for developmental delay: DRD3 and ZBTB20. In this report, we describe a new case of 3q13.31 microdeletion identified by array-CGH in a 16year-old girl sharing clinical features commonly observed in the 3q13.31 microdeletion syndrome. This girl had a microdeletion of 7.39Mb spanning the common critical region of overlapping. More interestingly, we report for the first time the existence of a microduplication reciprocal to the microdeletion syndrome. This familial 2.76Mb microduplication identified by array-CGH was carried by two brothers and their father. The phenotype shared by the brothers resembled the phenotype related to the 3q13.31 microdeletion syndrome including especially severe intellectual disability, developmental delay, behavioral abnormalities and obesity. This microduplication involves three strong candidate genes for the developmental delay ZBTB20, LSAMP and GAP43. Further molecular characterization showed that DRD3, another strong candidate gene for developmental delay, was not included in the duplicated region. However, a dosage alteration of this gene cannot be completely excluded as the duplication was inverted at proximity of this gene, as revealed by FISH analysis. Finally, we hypothesized that the phenotype shared by the two brothers could be related to a gene dosage imbalance even if gene expression could not be measured in relevant tissues such as brain or adipocytes.


Subject(s)
Developmental Disabilities/genetics , Gene Deletion , Genes, Duplicate , Obesity/genetics , Adolescent , Cell Adhesion Molecules, Neuronal/genetics , Chromosome Deletion , Chromosomes, Human, Pair 3/genetics , Comparative Genomic Hybridization , Developmental Disabilities/diagnosis , Developmental Disabilities/pathology , Female , GAP-43 Protein/genetics , GPI-Linked Proteins/genetics , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Male , Nerve Tissue Proteins/genetics , Obesity/pathology , Receptors, Dopamine D3/genetics , Transcription Factors/genetics
13.
Eur J Hum Genet ; 28(3): 324-332, 2020 03.
Article in English | MEDLINE | ID: mdl-31591517

ABSTRACT

The HoxD cluster is critical for vertebrate limb development. Enhancers located in both the telomeric and centromeric gene deserts flanking the cluster regulate the transcription of HoxD genes. In rare patients, duplications, balanced translocations or inversions misregulating HOXD genes are responsible for mesomelic dysplasia of the upper and lower limbs. By aCGH, whole-genome mate-pair sequencing, long-range PCR and fiber fluorescent in situ hybridization, we studied patients from two families displaying mesomelic dysplasia limited to the upper limbs. We identified microduplications including the HOXD cluster and showed that microduplications were in an inverted orientation and inserted between the HOXD cluster and the telomeric enhancers. Our results highlight the existence of an autosomal dominant condition consisting of isolated ulnar dysplasia caused by microduplications inserted between the HOXD cluster and the telomeric enhancers. The duplications likely disconnect the HOXD9 to HOXD11 genes from their regulatory sequences. This presumptive loss-of-function may have contributed to the phenotype. In both cases, however, these rearrangements brought HOXD13 closer to telomeric enhancers, suggesting that the alterations derive from the dominant-negative effect of this digit-specific protein when ectopically expressed during the early development of forearms, through the disruption of topologically associating domain structure at the HOXD locus.


Subject(s)
Bone Diseases, Developmental/genetics , Gene Duplication , Homeodomain Proteins/genetics , Upper Extremity Deformities, Congenital/genetics , Bone Diseases, Developmental/pathology , Cells, Cultured , Female , Humans , Infant , Loss of Function Mutation , Male , Multigene Family , Phenotype , Upper Extremity Deformities, Congenital/pathology
14.
Eur J Hum Genet ; 27(4): 663-668, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30683921

ABSTRACT

NAME OF THE DISEASE (SYNONYMS): Simpson-Golabi-Behmel syndrome (SGBS). OMIM# OF THE DISEASE: 312870. NAME OF THE ANALYSED GENES OR DNA/CHROMOSOME SEGMENTS: GPC3. OMIM# OF THE GENE(S): 300037. Review of the analytical and clinical validity as well as of the clinical utility of DNA-based testing for mutations in the GPC3 gene(s) in ⊠ diagnostic, ☐ predictive and ⊠ prenatal settings and for ⊠ risk assessment in relatives.


Subject(s)
Abnormalities, Multiple/genetics , Arrhythmias, Cardiac/genetics , Genetic Diseases, X-Linked/genetics , Genetic Predisposition to Disease , Gigantism/genetics , Glypicans/genetics , Heart Defects, Congenital/genetics , Intellectual Disability/genetics , Abnormalities, Multiple/physiopathology , Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/physiopathology , Female , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/physiopathology , Genetic Testing , Gigantism/diagnosis , Gigantism/physiopathology , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/physiopathology , Humans , Intellectual Disability/diagnosis , Intellectual Disability/physiopathology , Male , Mutation/genetics , Phenotype
15.
Mol Genet Genomic Med ; 7(8): e786, 2019 08.
Article in English | MEDLINE | ID: mdl-31254375

ABSTRACT

BACKGROUND: There is a strong evidence for genetic factors as the main causes of Autism Spectrum Disorders (ASD). To date, hundreds of genes have been identified either by copy number variations (CNVs) and/or single nucleotide variations. However, despite all the findings, the genetics of these disorders have not been totally explored. METHODS: Thus, the aim of our work was to identify rare CNVs and genes present in these regions in ASD children, using a high-resolution comparative genomic hybridization technique and quantitative PCR (qPCR) approach. RESULTS: Our results have shown 60-70 chromosomal aberrations per patient. We have initially selected 66 CNVs that have been further assessed using qPCR. Finally, we have validated 22 CNVs including 11 deletions and 11 duplications. Ten CNVs are de novo, 11 are inherited and one of unknown origin of transmission. Among the CNVs detected, novel ASD candidate genes PJA2, SYNPO, APCS, and TAC1 have been identified in our group of Lebanese patients. In addition, previously described CNVs have been identified containing genes such as SHANK3, MBP, CHL1, and others. CONCLUSION: Our study broadens the population spectrum of studied ASD patients and adds new candidates at the list of genes contributing to these disorders.


Subject(s)
Autism Spectrum Disorder/genetics , Genetic Predisposition to Disease , Adult , Aged , Autism Spectrum Disorder/blood , Autism Spectrum Disorder/diagnosis , Child , Comparative Genomic Hybridization , DNA Copy Number Variations , Female , Humans , Lebanon , Male , Microfilament Proteins/genetics , Middle Aged , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Serum Amyloid P-Component/genetics , Tachykinins/genetics , Ubiquitin-Protein Ligases/genetics , Young Adult
17.
Ann Clin Biochem ; 55(5): 543-552, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29388433

ABSTRACT

Objectives Autism spectrum disorders and intellectual disability present a challenge for therapeutic and dietary management. We performed a re-analysis of plasma amino acid chromatography of children with autism spectrum disorders ( n = 22) or intellectual disability ( n = 29) to search for a metabolic signature that can distinguish individuals with these disorders from controls ( n = 30). Methods We performed univariate and multivariate analyses using different machine learning strategies, from the raw data of the amino acid chromatography. Finally, we analysed the metabolic pathways associated with discriminant biomarkers. Results Multivariate analysis revealed models to discriminate patients with autism spectrum disorders or intellectual disability and controls from plasma amino acid profiles ( P < 0.0003). Univariate analysis showed that autism spectrum disorder and intellectual disability patients shared similar differences relative to controls, including lower glutamate ( P < 0.0001 and P = 0.0002, respectively) and serine ( P = 0.002 for both) concentrations. The multivariate model ( P < 6.12.10-7) to discriminate between autism spectrum disorders and intellectual disability revealed the involvement of urea, 3-methyl-histidine and histidine metabolism. Biosigner analysis and univariate analysis confirmed the role of 3-methylhistidine ( P = 0.004), histidine ( P = 0.003), urea ( P = 0.0006) and lysine ( P = 0.002). Conclusions We revealed discriminant metabolic patterns between autism spectrum disorders, intellectual disability and controls. Amino acids known to play a role in neurotransmission were discriminant in the models comparing autism spectrum disorders or intellectual disability to controls, and histidine and b-alanine metabolism was specifically highlighted in the model.


Subject(s)
Amino Acids/blood , Autism Spectrum Disorder , Intellectual Disability , Metabolome , Adolescent , Child , Child, Preschool , Female , Humans , Male , Metabolome/physiology , Models, Biological , Multivariate Analysis , Reference Standards
18.
Eur J Hum Genet ; 26(1): 85-93, 2018 01.
Article in English | MEDLINE | ID: mdl-29184170

ABSTRACT

Syndromes caused by copy number variations are described as reciprocal when they result from deletions or duplications of the same chromosomal region. When comparing the phenotypes of these syndromes, various clinical features could be described as reversed, probably due to the opposite effect of these imbalances on the expression of genes located at this locus. The NFIX gene codes for a transcription factor implicated in neurogenesis and chondrocyte differentiation. Microdeletions and loss of function variants of NFIX are responsible for Sotos syndrome-2 (also described as Malan syndrome), a syndromic form of intellectual disability associated with overgrowth and macrocephaly. Here, we report a cohort of nine patients harboring microduplications encompassing NFIX. These patients exhibit variable intellectual disability, short stature and small head circumference, which can be described as a reversed Sotos syndrome-2 phenotype. Strikingly, such a reversed phenotype has already been described in patients harboring microduplications encompassing NSD1, the gene whose deletions and loss-of-function variants are responsible for classical Sotos syndrome. Even though the type/contre-type concept has been criticized, this model seems to give a plausible explanation for the pathogenicity of 19p13 microduplications, and the common phenotype observed in our cohort.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Duplication , Chromosomes, Human, Pair 19/genetics , Intellectual Disability/genetics , NFI Transcription Factors/genetics , Abnormalities, Multiple/pathology , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Intellectual Disability/pathology , Male , Syndrome
19.
Eur J Med Genet ; 60(11): 605-609, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28811189

ABSTRACT

Koolen-de Vries syndrome (MIM#610443) is a rare microdeletion syndrome involving the 17q21.31 region, which was first described by Koolen in 2006. Clinical and behavioral characteristics have been extensively reported from more than 100 postnatal cases including infants, children and young adults. The syndrome is highly clinically heterogeneous, but the main features associate characteristic cranio-facial dysmorphism, heart defects, limb, skeletal, genito-urinary anomalies, along with intellectual disability with early childhood epilepsy and behavioral disturbances. Central nervous system malformations usually consist in hydrocephalus and thin corpus callosum. We report herein an early fetal case with an apparently isolated abnormal corpus callosum diagnosed by ultrasonography, for which a medical termination of the pregnancy was achieved at 22 weeks of gestation. Postmortem examination displayed facial dysmorphism consisting of hypertelorism, short philtrum and flat and broad nose, cleft palate and left duplex ureter. Neuropathological examination revealed a mega corpus callosum that has never been reported so far in this syndrome. Array-CGH performed on thymic DNA tissue revealed a 17q21.31 microdeletion, which allowed for the confirmation of early occurring Koolen-de Vries syndrome.


Subject(s)
Abnormalities, Multiple/genetics , Intellectual Disability/genetics , Phenotype , Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/pathology , Adult , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Corpus Callosum/diagnostic imaging , Corpus Callosum/pathology , Female , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/pathology , Pregnancy , Ultrasonography, Prenatal
20.
NPJ Genom Med ; 2: 32, 2017.
Article in English | MEDLINE | ID: mdl-29263841

ABSTRACT

Phelan-McDermid syndrome (PMS) is characterized by a variety of clinical symptoms with heterogeneous degrees of severity, including intellectual disability (ID), absent or delayed speech, and autism spectrum disorders (ASD). It results from a deletion of the distal part of chromosome 22q13 that in most cases includes the SHANK3 gene. SHANK3 is considered a major gene for PMS, but the factors that modulate the severity of the syndrome remain largely unknown. In this study, we investigated 85 patients with different 22q13 rearrangements (78 deletions and 7 duplications). We first explored the clinical features associated with PMS, and provide evidence for frequent corpus callosum abnormalities in 28% of 35 patients with brain imaging data. We then mapped several candidate genomic regions at the 22q13 region associated with high risk of clinical features, and suggest a second locus at 22q13 associated with absence of speech. Finally, in some cases, we identified additional clinically relevant copy-number variants (CNVs) at loci associated with ASD, such as 16p11.2 and 15q11q13, which could modulate the severity of the syndrome. We also report an inherited SHANK3 deletion transmitted to five affected daughters by a mother without ID nor ASD, suggesting that some individuals could compensate for such mutations. In summary, we shed light on the genotype-phenotype relationship of patients with PMS, a step towards the identification of compensatory mechanisms for a better prognosis and possibly treatments of patients with neurodevelopmental disorders.

SELECTION OF CITATIONS
SEARCH DETAIL