Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Epilepsia ; 64(8): e164-e169, 2023 08.
Article in English | MEDLINE | ID: mdl-36810721

ABSTRACT

The progressive myoclonus epilepsies (PMEs) are a heterogeneous group of neurodegenerative disorders, typically presenting in late childhood. An etiologic diagnosis is achieved in about 80% of patients with PME, and genome-wide molecular studies on remaining, well-selected, undiagnosed cases can further dissect the underlying genetic heterogeneity. Through whole-exome sequencing (WES), we identified pathogenic truncating variants in the IRF2BPL gene in two, unrelated patients presenting with PME. IRF2BPL belongs to the transcriptional regulators family and it is expressed in multiple human tissues, including the brain. Recently missense and nonsense mutations in IRF2BPL were found in patients presenting with developmental delay and epileptic encephalopathy, ataxia, and movement disorders, but none with clear PME. We identified 13 other patients in the literature with myoclonic seizures and IRF2BPL variants. There was no clear genotype-phenotype correlation. With the description of these cases, the IRF2BPL gene should be considered in the list of genes to be tested in the presence of PME, in addition to patients with neurodevelopmental or movement disorders.


Subject(s)
Epilepsies, Myoclonic , Movement Disorders , Myoclonic Epilepsies, Progressive , Humans , Child , Myoclonic Epilepsies, Progressive/genetics , Seizures/genetics , Genotype , Carrier Proteins/genetics , Nuclear Proteins/genetics
2.
Am J Med Genet A ; 185(4): 1204-1210, 2021 04.
Article in English | MEDLINE | ID: mdl-33442900

ABSTRACT

Feingold Syndrome type 1 (FS1) is an autosomal dominant disorder due to a loss of function mutations in the MYCN gene. FS1 is generally clinically characterized by mild learning disability, microcephaly, short palpebral fissures, short stature, brachymesophalangy, hypoplastic thumbs, as well as syndactyly of toes, variably associated with organ abnormalities, the most common being gastrointestinal atresia. In current literature, more than 120 FS1 patients have been described, but diagnostic criteria are not well agreed upon, likewise the genotype-phenotype correlations are not well understood. Here, we describe 11 FS1 patients, belonging to six distinct families, where we have identified three novel MYCN mutations along with three pathogenetic variants, the latter which have already been reported. Several patients presented a mild phenotype of the condition and they have been diagnosed as being affected only after segregation analyses of the MYCN mutation identified in the propositus. We also describe here the first ever FS1 patient with severe intellectual disability having a maternally inherited MYCN variant together with an additional GNAO1 mutation inherited paternally. Mutations in the GNAO1 gene are associated with a specific form of intellectual disability and epilepsy, thus the finding of two different rare diseases in the same patient could explain his severe phenotype. Therein, a thorough investigation is merited into the possibility that additional variants in patients with a MYCN mutation and severe phenotype do exist. Finally, in order to guarantee a more reliable diagnosis of FS1, we suggest using both major and minor clinical-molecular diagnostic criteria.


Subject(s)
Eyelids/abnormalities , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Genetic Predisposition to Disease , Intellectual Disability/genetics , Limb Deformities, Congenital/genetics , Microcephaly/genetics , N-Myc Proto-Oncogene Protein/genetics , Tracheoesophageal Fistula/genetics , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Adolescent , Child , Child, Preschool , Eyelids/pathology , Female , Genetic Association Studies , Genetic Testing , Genotype , Humans , Infant , Infant, Newborn , Intellectual Disability/complications , Intellectual Disability/pathology , Limb Deformities, Congenital/complications , Limb Deformities, Congenital/pathology , Male , Microcephaly/complications , Microcephaly/pathology , Phenotype , Syndactyly/complications , Syndactyly/genetics , Syndactyly/pathology , Tracheoesophageal Fistula/complications , Tracheoesophageal Fistula/pathology
3.
Int J Mol Sci ; 22(16)2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34445317

ABSTRACT

The 22q11.2 deletion syndrome (22q11.2DS) is the most common genomic disorder in humans and is the result of a recurrent 1.5 to 2.5 Mb deletion, encompassing approximately 20-40 genes, respectively. The clinical presentation of the typical deletion includes: Velocardiofacial, Di George, Opitz G/BBB and Conotruncalanomaly face syndromes. Atypical deletions (proximal, distal or nested) are rare and characterized mainly by normal phenotype or mild intellectual disability and variable clinical features. The pathogenetic mechanisms underlying this disorder are not completely understood. Because the 22q11.2 region harbours genes coding for transcriptional factors and chromatin remodelers, in this study, we performed analysis of genome-wide DNA methylation of peripheral blood from 49 patients with 22q11.2DS using the Illumina Infinium Methylation EPIC bead chip arrays. This cohort comprises 43 typical, 2 proximal and 4 distal deletions. We demonstrated the evidence of a unique and highly specific episignature in all typical and proximal 22q11.2DS. The sensitivity and specificity of this signature was further confirmed by comparing it to over 1500 patients with other neurodevelopmental disorders with known episignatures. Mapping the 22q11.2DS DNA methylation episignature provides both novel insights into the molecular pathogenesis of this disorder and an effective tool in the molecular diagnosis of 22q11.2DS.


Subject(s)
DNA Methylation , DiGeorge Syndrome/genetics , Epigenome , Female , Humans , Infant , Male
4.
Hum Mutat ; 40(2): 201-206, 2019 02.
Article in English | MEDLINE | ID: mdl-30427563

ABSTRACT

Retinoblastoma (RB), which represents the most common childhood eye cancer, is caused by biallelic inactivation of RB1 gene. Promoter hypermethylation is quite frequent in RB tissues but conclusive evidence of soma-wide predisposing epimutations is currently scant. Here, 50 patients who tested negative for RB1 germline sequence alterations were screened for aberrant promoter methylation using methylation-specific MLPA. The assay, performed on blood, identified a sporadic patient with methylation of CpG106, absent in parents' DNA. Bisulfite pyrosequencing accurately quantified CpG methylation in blood DNA (mean ∼49%) and also confirmed the aberration in DNA isolated from oral mucosa although at lower levels (mean ∼34%). Using a tag-SNP, methylation was demonstrated to affect the maternal allele. Real-time qPCR demonstrated RB1 transcriptional silencing. In conclusion, we documented that promoter methylation can act as the first "hit" in Knudson's model. This mosaic epimutation mimics the effect of an inactivating mutation and phenocopies RB onset.


Subject(s)
DNA Methylation/genetics , Genetic Predisposition to Disease , Retinoblastoma Binding Proteins/genetics , Retinoblastoma/genetics , Ubiquitin-Protein Ligases/genetics , Alleles , Epigenesis, Genetic , Female , Gene Silencing , Humans , Infant , Male , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic , Retinoblastoma/pathology
5.
Exp Cell Res ; 368(2): 225-235, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29730163

ABSTRACT

Mutations in MECP2 gene have been identified in more than 95% of patients with classic Rett syndrome, one of the most common neurodevelopmental disorders in females. Taking advantage of the breakthrough technology of genetic reprogramming, we investigated transcriptome changes in neurons differentiated from induced Pluripotent Stem Cells (iPSCs) derived from patients with different mutations. Profiling by RNA-seq in terminally differentiated neurons revealed a prominent GABAergic circuit disruption along with a perturbation of cytoskeleton dynamics. In particular, in mutated neurons we identified a significant decrease of acetylated α-tubulin which can be reverted by treatment with selective inhibitors of HDAC6, the main α-tubulin deacetylase. These findings contribute to shed light on Rett pathogenic mechanisms and provide hints for the treatment of Rett-associated epileptic behavior as well as for the definition of new therapeutic strategies for Rett syndrome.


Subject(s)
GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Histone Deacetylase 6/metabolism , Induced Pluripotent Stem Cells/physiology , Rett Syndrome/metabolism , Rett Syndrome/physiopathology , Tubulin/metabolism , Acetylation , Cell Differentiation/physiology , Female , Humans , Male
6.
Hum Mutat ; 39(2): 302-314, 2018 02.
Article in English | MEDLINE | ID: mdl-29098738

ABSTRACT

Alport Syndrome (ATS) is a rare genetic disorder caused by collagen IV genes mutations, leading to glomerular basement membrane damage up to end-stage renal disease. Podocytes, the main component of the glomerular structure, are the only cells able to produce all the three collagens IV alpha chains associated with ATS and thus, they are key players in ATS pathogenesis. However, podocytes-targeted therapeutic strategies have been hampered by the difficulty of non-invasively isolating them and transcripts-based diagnostic approaches are complicated by the inaccessibility of other COL4 chains-expressing cells. We firstly isolated podocyte-lineage cells from ATS patients' urine samples, in a non-invasive way. RT-PCR analysis revealed COL4A3, COL4A4, and COL4A5 expression. Transcripts analysis on RNA extracted from patient's urine derived podocyte-lineage cells allowed defining the pathogenic role of intronic variants, namely one mutation in COL4A3 (c.3882+5G>A), three mutations in COL4A4 (c.1623+2T>A, c.3699_3706+1del, c.2545+143T>A), and one mutation in COL4A5 (c.3454+2T>C). Therefore, our cellular model represents a novel tool, essential to unequivocally prove the effect of spliceogenic intronic variants on transcripts expressed exclusively at a glomerular level. This process is a key step for providing the patient with a definite molecular diagnosis and with a proper recurrence risk. The established system also opens up the possibility of testing personalized therapeutic approaches on disease-relevant cells.


Subject(s)
Nephritis, Hereditary/therapy , Podocytes/cytology , Precision Medicine/methods , Adolescent , Adult , Aged , Autoantigens/genetics , Child , Collagen Type IV/genetics , Female , Humans , Male , Middle Aged , Mutation/genetics , Pathology, Molecular/methods , Pedigree , Young Adult
7.
J Hum Genet ; 63(5): 563-568, 2018 May.
Article in English | MEDLINE | ID: mdl-29531337

ABSTRACT

Biallelic mutations in NDUFAF6 have been identified as responsible for cases of autosomal recessive Leigh syndrome associated with mitochondrial complex I deficiency. Here we report two siblings and two unrelated subjects with Leigh syndrome, in which we found the same compound heterozygous missense (c.532G>C:p.A178P) and deep intronic (c.420+784C>T) variants in NDUFAF6. We demonstrated that the identified intronic variant creates an alternative splice site, leading to the production of an aberrant transcript. A detailed analysis of whole-exome sequencing data together with the functional validation based on mRNA analysis may reveal pathogenic variants even in non-exonic regions.


Subject(s)
Exome Sequencing , Heterozygote , Introns , Leigh Disease/diagnosis , Leigh Disease/genetics , Mutation, Missense , RNA, Messenger/genetics , Alleles , Child , Child, Preschool , Female , Fibroblasts/metabolism , Gene Expression , Haplotypes , Humans , Infant , Lymphocytes/metabolism , Magnetic Resonance Imaging/methods , Male , Mitochondrial Proteins , Pedigree , Phenotype
8.
Mol Genet Metab ; 119(3): 214-222, 2016 11.
Article in English | MEDLINE | ID: mdl-27623250

ABSTRACT

We report here the case of a young male who started to show verbal fluency disturbance, clumsiness and gait anomalies at the age of 3.5years and presented bilateral striatal necrosis. Clinically, the diagnosis was compatible with Leigh syndrome but the underlying molecular defect remained elusive even after exome analysis using autosomal/X-linked recessive or de novo models. Dosage of respiratory chain activity on fibroblasts, but not in muscle, underlined a deficit in complex I. Re-analysis of heterozygous probably pathogenic variants, inherited from one healthy parent, identified the p.Ala178Pro in NDUFAF6, a complex I assembly factor. RNA analysis showed an almost mono-allelic expression of the mutated allele in blood and fibroblasts and puromycin treatment on cultured fibroblasts did not lead to the rescue of the maternal allele expression, not supporting the involvement of nonsense-mediated RNA decay mechanism. Complementation assay underlined a recovery of complex I activity after transduction of the wild-type gene. Since the second mutation was not detected and promoter methylation analysis resulted normal, we hypothesized a non-exonic event in the maternal allele affecting a regulatory element that, in conjunction with the paternal mutation, leads to the autosomal recessive disorder and the different allele expression in various tissues. This paper confirms NDUFAF6 as a genuine morbid gene and proposes the coupling of exome sequencing with mRNA analysis as a method useful for enhancing the exome sequencing detection rate when the simple application of classical inheritance models fails.


Subject(s)
Exome/genetics , Leigh Disease/genetics , Mitochondrial Proteins/genetics , Speech Disorders/genetics , Alleles , Child, Preschool , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Leigh Disease/physiopathology , Male , Mutation , Pedigree , Phenotype , RNA, Messenger/genetics , Speech Disorders/physiopathology , Striatonigral Degeneration/congenital , Striatonigral Degeneration/genetics , Striatonigral Degeneration/physiopathology
9.
Int J Mol Sci ; 17(3): 306, 2016 Feb 27.
Article in English | MEDLINE | ID: mdl-26927095

ABSTRACT

We highlight the importance of exome sequencing in solving a clinical case of a child who died at 14 months after a series of respiratory crises. He was the half-brother of a girl diagnosed at 7 years with the early-onset seizure variant of Rett syndrome due to CDKL5 mutation. We performed a test for CDKL5 in the boy, which came back negative. Driven by the mother's compelling need for a diagnosis, we moved forward performing whole exome sequencing analysis. Surprisingly, two missense mutations in compound heterozygosity were identified in the RAPSN gene encoding a receptor-associated protein with a key role in clustering and anchoring nicotinic acetylcholine receptors at synaptic sites. This gene is responsible for a congenital form of myasthenic syndrome, a disease potentially treatable with cholinesterase inhibitors. Therefore, an earlier diagnosis in this boy would have led to a better clinical management and prognosis. Our study supports the key role of exome sequencing in achieving a definite diagnosis in severe perinatal diseases, an essential step especially when a specific therapy is available.


Subject(s)
Exome , Muscle Proteins/genetics , Myasthenic Syndromes, Congenital/genetics , Respiration Disorders/genetics , Autopsy , Child , Female , Humans , Infant , Male , Mutation, Missense , Myasthenic Syndromes, Congenital/pathology , Respiration Disorders/pathology
10.
BMC Med Genomics ; 15(1): 181, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35978409

ABSTRACT

BACKGROUND: PCDH19-related epilepsy is a rare X-linked type of epilepsy caused by genomic variants of the Protocadherin 19 (PCDH19) gene. The clinical characteristics of PCDH19-related epilepsy are epileptic and non-epileptic symptoms with highly variable severity among patients. CASE PRESENTATION: We present a case of a 4-year old female with PCDH19-related epilepsycaused by new variants in the PCDH19 gene. Our patient was admitted for the first time at the age of 12 months for seizure clusters arising under condition of apyrexia. The electroencephalography (EEG) showed frontal paroxysmal activity. The genetic analysis identified the two variants c.1006G > A (p.Val336Met) and c.1014C > A (p.Asp338Glu) in the gene PCDH19. The patient was treated with Carbamazepine and Clonazepam achieving the disappearance of seizures. During the follow-up, the neurological examination was persistently normal with neither cognitive impairment nor behavior disturbances. From 2 years of age EEG controls were persistently normal. CONCLUSION: This patient presents two novel variants of the PCDH19 gene associated with a mild form of epilepsy with normal cognitive development with an apparently better prognosis. According to our experience, the dual therapy with Carbamazepine and Clonazepam has led to a good control of seizures.


Subject(s)
Epilepsy , Protocadherins , Cadherins/genetics , Carbamazepine/therapeutic use , Child, Preschool , Clonazepam/therapeutic use , Epilepsy/genetics , Female , Humans , Infant , Mutation , Seizures/genetics
11.
Genes (Basel) ; 12(9)2021 09 12.
Article in English | MEDLINE | ID: mdl-34573388

ABSTRACT

In recent years, a rare form of autosomal recessive brachyolmia associated with amelogenesis imperfecta (AI) has been described as a novel nosologic entity. This disorder is characterized by skeletal dysplasia (e.g., platyspondyly, short trunk, scoliosis, broad ilia, elongated femoral necks with coxa valga) and severe enamel and dental anomalies. Pathogenic variants in the latent transforming growth factor-ß binding protein 3 (LTBP3) gene have been found implicated in the pathogenesis of this disorder. So far, biallelic pathogenic LTBP3 variants have been identified in less than 10 families. We here report a young boy born from consanguineous parents with a complex phenotype including skeletal dysplasia associated with aortic stenosis, hypertrophic cardiomyopathy, hypodontia and amelogenesis imperfecta caused by a previously unreported homozygous LTBP3 splice site variant. We also compare the genotypes and phenotypes of patients reported to date. This work provides further evidence that brachyolmia with amelogenesis imperfecta is a distinct nosologic entity and that variations in LTBP3 are involved in its pathogenesis.


Subject(s)
Amelogenesis Imperfecta/genetics , Latent TGF-beta Binding Proteins/genetics , Osteochondrodysplasias/genetics , Adolescent , Amelogenesis Imperfecta/complications , Amelogenesis Imperfecta/diagnosis , Consanguinity , Humans , Male , Osteochondrodysplasias/complications , Osteochondrodysplasias/diagnosis , Pedigree , Peru , Phenotype , Rare Diseases , Exome Sequencing
12.
Eur J Hum Genet ; 27(8): 1260-1266, 2019 08.
Article in English | MEDLINE | ID: mdl-30936464

ABSTRACT

Schilbach-Rott syndrome (SRS, OMIM%164220) is a disorder of unknown aetiology that is characterised by hypotelorism, epichantal folds, cleft palate, dysmorphic face, hypospadia in males and mild mental retardation in some patients. To date, 5 families and 17 patients have exhibited this phenotype, and recurrence in two of these families suggests an autosomal dominant inheritance. SRS overlaps with a mild form of holoprosencephaly (HPE), but array-CGH analysis and sequencing of some HPE-related genes (SEPT9, SHH and TWIST) did not reveal any variants in at least one family. Herein, we investigated by array-CGH analysis a 11-year-old female patient and her father, both exhibiting the typical SRS phenotype, disclosing in the daughter-father couple the same microduplication of chromosome 9q22.32q22.33 [arr[hg19]9q22.32(98,049,611_98,049,636)x3,9q22.33 (99,301,483_99,301,508)x3], involving eight genes, including PTCH1. The duplication segregated with the disease, since it was not found in the healthy paternal grandparents of the proband. The gain-of-function variants of the PTCH1 gene are responsible for a mild form of HPE. This is the first genetic variant found in SRS. This finding reinforces the hypothesis that SRS belongs to the HPE clinical spectrum and suggests to perform array-CGH in patients with SRS phenotype and, if negative, to consider a potential benefit from sequencing of HPE-related genes.


Subject(s)
Chromosome Duplication , Chromosomes, Human, Pair 9/genetics , Cleft Palate/genetics , Craniofacial Abnormalities/genetics , Gene Duplication , Holoprosencephaly/genetics , Hypospadias/genetics , Patched-1 Receptor/genetics , Child , Cleft Palate/diagnosis , Comparative Genomic Hybridization , Craniofacial Abnormalities/diagnosis , Fathers , Female , Holoprosencephaly/diagnosis , Humans , Hypospadias/diagnosis , Intellectual Disability/genetics , Male , Phenotype
13.
Cancers (Basel) ; 11(3)2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30832263

ABSTRACT

Highly penetrant variants of BRCA1/2 genes are involved in hereditary predisposition to breast and ovarian cancer. The detection of pathogenic BRCA variants has a considerable clinical impact, allowing appropriate cancer-risk management. However, a major drawback is represented by the identification of variants of uncertain significance (VUS). Many VUS potentially affect mRNA splicing, making transcript analysis an essential step for the definition of their pathogenicity. Here, we characterize the impact on splicing of ten BRCA1/2 variants. Aberrant splicing patterns were demonstrated for eight variants whose alternative transcripts were fully characterized. Different events were observed, including exon skipping, intron retention, and usage of de novo and cryptic splice sites. Transcripts with premature stop codons or in-frame loss of functionally important residues were generated. Partial/complete splicing effect and quantitative contribution of different isoforms were assessed, leading to variant classification according to Evidence-based Network for the Interpretation of Mutant Alleles (ENIGMA) consortium guidelines. Two variants could be classified as pathogenic and two as likely benign, while due to a partial splicing effect, six variants remained of uncertain significance. The association with an undefined tumor risk justifies caution in recommending aggressive risk-reduction treatments, but prevents the possibility of receiving personalized therapies with potential beneficial effect. This indicates the need for applying additional approaches for the analysis of variants resistant to classification by gene transcript analyses.

14.
Eur J Hum Genet ; 26(7): 1026-1037, 2018 07.
Article in English | MEDLINE | ID: mdl-29662154

ABSTRACT

Retinoblastoma is the most common eye cancer in children. Numerous families have been described displaying reduced penetrance and expressivity. An extensive molecular characterization of seven families led us to characterize the two main mechanisms impacting on phenotypic expression, as follows: (i) mosaicism of amorphic pathogenic variants; and (ii) parent-of-origin-effect of hypomorphic pathogenic variants. Somatic mosaicism for RB1 splicing variants (c.1960+5G>C and c.2106+2T>C), leading to a complete loss of function was demonstrated by high-depth NGS in two families. In both cases, the healthy carrier parent (one with retinoma) showed a variant frequency lower than that expected for a heterozygous individual, indicating a 56-60% mosaicism level. Previous evidences of a ~3-fold excess of RB1 maternal canonical transcript led us to hypothesize that this differential allelic expression could influence phenotypic outcome in families at risk for RB onset. Accordingly, in five families, we identified a higher tumor risk associated with paternally inherited hypomorphic pathogenic variants, namely a deletion resulting in the loss of 37 amino acids at the N-terminus (c.608-16_608del), an exonic substitution with a "leaky" splicing effect (c.1331A>G), a partially deleterious substitution (c.1981C>T) and a truncating C-terminal variant (c.2663+2T>C). The identification of these mechanisms changes the genetic/prenatal counseling and the clinical management of families, indicating a higher recurrence risk when the hypomorphic pathogenic variant is inherited from the father, and suggesting the need for second tumor surveillance in unaffected carriers at risk of developing adult-onset cancer such as osteosarcoma or leiomyosarcoma.


Subject(s)
Genetic Counseling , RNA Splicing/genetics , Retinoblastoma Binding Proteins/genetics , Retinoblastoma/genetics , Ubiquitin-Protein Ligases/genetics , Adolescent , Adult , Child , Child, Preschool , Exons/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Infant , Male , Mosaicism , Mutation , Pedigree , Pregnancy , Retinoblastoma/epidemiology , Retinoblastoma/pathology , Risk Factors , Young Adult
15.
Clin Dysmorphol ; 26(1): 18-25, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27749392

ABSTRACT

Orofacial clefts are the most common congenital craniofacial anomalies and can occur as an isolated defect or be associated with other anomalies such as posterior fossa anomalies as a part of several genetic syndromes. We report two consecutive voluntary pregnancy interruptions in a nonconsanguineous couple following the fetal ultrasound finding of cleft lip and palate and posterior fossa anomalies confirmed by means of post-termination examination on the second fetus. The quantitative fluorescent PCR, the karyotype, and the comparative genomic hybridization-array analysis after amniocentesis were normal. Exome sequencing on abortive material from both fetuses detected a missense mutation in MID1, resulting in a clinical diagnosis of Opitz G/BBB syndrome. The same mutation was found in the mother and in her brother, who both revealed cerebellar anomalies at an MRI examination. Our study supports the efficacy of exome sequencing in the presence of both a family history suggestive of an inherited disorder and well-documented ultrasound findings. It reveals the importance of a synergistic effort between gynecologists and geneticists aimed at the integration of the most sophisticated ultrasound techniques with the next-generation sequencing tools to provide a definite diagnosis essential to orient the final decision and to estimate a proper recurrence risk.


Subject(s)
Esophagus/abnormalities , Exome , High-Throughput Nucleotide Sequencing , Hypertelorism/diagnosis , Hypertelorism/genetics , Hypospadias/diagnosis , Hypospadias/genetics , Ultrasonography, Prenatal , Abortion, Induced , Comparative Genomic Hybridization , Female , Fetus , Genetic Association Studies , Humans , Karyotyping , Magnetic Resonance Imaging , Male , Mutation , Pedigree , Phenotype , Pregnancy
16.
Brain Dev ; 38(6): 590-6, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26754451

ABSTRACT

BACKGROUND: Neurodevelopmental disorders include a broad spectrum of conditions, which are characterized by delayed motor and/or cognitive milestones and by a variable range of intellectual disability with or without an autistic behavior. Several genetic factors have been implicated in intellectual disability onset and exome sequencing studies have recently identified new inherited or de novo mutations in patients with neurodevelopmental disorders. CASE: We report the case of two monozygotic twins who came for the first time to our attention at the age of 20months for a global neurodevelopmental delay associated with an autism spectrum disorder, hypotonia, postnatal microcephaly, stereotypic movements and circadian rhythm alterations in association with late-onset epilepsy. MECP2 sequence was normal. A CGH-array analysis revealed in both twins two maternally inherited duplications on chromosomes 8p22 and 16p13.11. The latter has been previously associated with neurodevelopmental disorders. We performed an exome sequencing analysis on one twin and her parents and identified a CHD2 mutation, previously described in association with a phenotypic spectrum overlapping our patients' phenotype. CONCLUSIONS: This work underlines the importance to consider a CHD2 involvement in children with intellectual disability and autism spectrum disorder even in the absence of epilepsy at an early age. It also highlights the necessity to re-evaluate inherited copy number variants with low penetrance and/or high phenotypic variability because an underlying de novo molecular event can be the major cause of the phenotype. This is essential in order to reach a correct diagnosis and provide the couple with a proper recurrence risk.


Subject(s)
DNA-Binding Proteins/genetics , Intellectual Disability/genetics , Twins, Monozygotic/genetics , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/physiopathology , DNA Mutational Analysis/methods , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Developmental Disabilities/physiopathology , Diseases in Twins , Exome , Face/abnormalities , Genotyping Techniques , Humans , Infant , Intellectual Disability/pathology , Intellectual Disability/physiopathology , Methyl-CpG-Binding Protein 2/genetics , Pedigree , Phenotype
17.
Eur J Hum Genet ; 23(11): 1523-30, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25712084

ABSTRACT

In about 50% of sporadic cases of retinoblastoma, no constitutive RB1 mutations are detected by conventional methods. However, recent research suggests that, at least in some of these cases, there is somatic mosaicism with respect to RB1 normal and mutant alleles. The increased availability of next generation sequencing improves our ability to detect the exact percentage of patients with mosaicism. Using this technology, we re-tested a series of 40 patients with sporadic retinoblastoma: 10 of them had been previously classified as constitutional heterozygotes, whereas in 30 no RB1 mutations had been found in lymphocytes. In 3 of these 30 patients, we have now identified low-level mosaic variants, varying in frequency between 8 and 24%. In 7 out of the 10 cases previously classified as heterozygous from testing blood cells, we were able to test additional tissues (ocular tissues, urine and/or oral mucosa): in three of them, next generation sequencing has revealed mosaicism. Present results thus confirm that a significant fraction (6/40; 15%) of sporadic retinoblastoma cases are due to postzygotic events and that deep sequencing is an efficient method to unambiguously distinguish mosaics. Re-testing of retinoblastoma patients through next generation sequencing can thus provide new information that may have important implications with respect to genetic counseling and family care.


Subject(s)
High-Throughput Nucleotide Sequencing , Mosaicism , Retinoblastoma Protein/genetics , Retinoblastoma/genetics , Adolescent , Adult , Alleles , Child , Child, Preschool , Female , Germ-Line Mutation , Heterozygote , Humans , Male , Retinoblastoma/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL