Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nat Commun ; 14(1): 853, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36792598

ABSTRACT

Following the diagnosis of a paediatric disorder caused by an apparently de novo mutation, a recurrence risk of 1-2% is frequently quoted due to the possibility of parental germline mosaicism; but for any specific couple, this figure is usually incorrect. We present a systematic approach to providing individualized recurrence risk. By combining locus-specific sequencing of multiple tissues to detect occult mosaicism with long-read sequencing to determine the parent-of-origin of the mutation, we show that we can stratify the majority of couples into one of seven discrete categories associated with substantially different risks to future offspring. Among 58 families with a single affected offspring (representing 59 de novo mutations in 49 genes), the recurrence risk for 35 (59%) was decreased below 0.1%, but increased owing to parental mixed mosaicism for 5 (9%)-that could be quantified in semen for paternal cases (recurrence risks of 5.6-12.1%). Implementation of this strategy offers the prospect of driving a major transformation in the practice of genetic counselling.


Subject(s)
Fathers , Parturition , Male , Pregnancy , Female , Humans , Child , Mutation , Risk Assessment , Germ Cells , Mosaicism , Pedigree , Germ-Line Mutation
2.
Genet Med ; 24(6): 1261-1273, 2022 06.
Article in English | MEDLINE | ID: mdl-35341651

ABSTRACT

PURPOSE: This study aimed to undertake a multidisciplinary characterization of the phenotype associated with SOX11 variants. METHODS: Individuals with protein altering variants in SOX11 were identified through exome and genome sequencing and international data sharing. Deep clinical phenotyping was undertaken by referring clinicians. Blood DNA methylation was assessed using Infinium MethylationEPIC array. The expression pattern of SOX11 in developing human brain was defined using RNAscope. RESULTS: We reported 38 new patients with SOX11 variants. Idiopathic hypogonadotropic hypogonadism was confirmed as a feature of SOX11 syndrome. A distinctive pattern of blood DNA methylation was identified in SOX11 syndrome, separating SOX11 syndrome from other BAFopathies. CONCLUSION: SOX11 syndrome is a distinct clinical entity with characteristic clinical features and episignature differentiating it from BAFopathies.


Subject(s)
DNA Methylation , Hypogonadism , Klinefelter Syndrome , Neurodevelopmental Disorders , SOXC Transcription Factors , DNA Methylation/genetics , Humans , Hypogonadism/genetics , Klinefelter Syndrome/genetics , Neurodevelopmental Disorders/genetics , Phenotype , SOXC Transcription Factors/genetics , Exome Sequencing
3.
Eur J Hum Genet ; 30(3): 291-297, 2022 03.
Article in English | MEDLINE | ID: mdl-34897289

ABSTRACT

Individuals with the three base pair deletion NM_000267.3(NF1):c.2970_2972del p.(Met992del) have been recognised to present with a milder neurofibromatosis type 1 (NF1) phenotype characterised by café-au-lait macules (CALs) and intertriginous freckling, as well as a lack of cutaneous, subcutaneous and plexiform neurofibromas and other NF1-associated complications. Examining large cohorts of patients over time with this specific genotype is important to confirm the presentation and associated risks of this variant across the lifespan. Forty-one individuals with the in-frame NF1 deletion p.Met992del were identified from 31 families. Clinicians completed a standardised clinical questionnaire for each patient and the resulting data were collated and compared to published cohorts. Thirteen patients have been previously reported, and updated clinical information has been obtained for these individuals. Both CALs and intertriginous freckling were present in the majority of individuals (26/41, 63%) and the only confirmed features in 11 (27%). 34/41 (83%) of the cohort met NIH diagnostic criteria. There was a notable absence of all NF1-associated tumour types (neurofibroma and glioma). Neurofibroma were observed in only one individual-a subcutaneous lesion (confirmed histologically). Nineteen individuals were described as having a learning disability (46%). This study confirms that individuals with p.Met992del display a mild tumoural phenotype compared to those with 'classical', clinically diagnosed NF1, and this appears to be the case longitudinally through time as well as at presentation. Learning difficulties, however, appear to affect a significant proportion of NF1 subjects with this phenotype. Knowledge of this genotype-phenotype association is fundamental to accurate prognostication for families and caregivers.


Subject(s)
Neurofibroma , Neurofibromatosis 1 , Cafe-au-Lait Spots/genetics , Genetic Association Studies , Humans , Longitudinal Studies , Neurofibroma/genetics , Neurofibromatosis 1/diagnosis , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology
4.
J Med Genet ; 57(7): 466-474, 2020 07.
Article in English | MEDLINE | ID: mdl-32277047

ABSTRACT

PURPOSE: Marfanoid habitus (MH) combined with intellectual disability (ID) (MHID) is a clinically and genetically heterogeneous presentation. The combination of array CGH and targeted sequencing of genes responsible for Marfan or Lujan-Fryns syndrome explain no more than 20% of subjects. METHODS: To further decipher the genetic basis of MHID, we performed exome sequencing on a combination of trio-based (33 subjects) or single probands (31 subjects), of which 61 were sporadic. RESULTS: We identified eight genes with de novo variants (DNVs) in at least two unrelated individuals (ARID1B, ATP1A1, DLG4, EHMT1, NFIX, NSD1, NUP205 and ZEB2). Using simulation models, we showed that five genes (DLG4, NFIX, EHMT1, ZEB2 and ATP1A1) met conservative Bonferroni genomewide significance for an excess of the observed de novo point variants. Overall, at least one pathogenic or likely pathogenic variant was identified in 54.7% of subjects (35/64). These variants fell within 27 genes previously associated with Mendelian disorders, including NSD1 and NFIX, which are known to be mutated in overgrowth syndromes. CONCLUSION: We demonstrated that DNVs were enriched in chromatin remodelling (p=2×10-4) and genes regulated by the fragile X mental retardation protein (p=3×10-8), highlighting overlapping genetic mechanisms between MHID and related neurodevelopmental disorders.


Subject(s)
Craniofacial Abnormalities/genetics , Histone-Lysine N-Methyltransferase/genetics , Intellectual Disability/genetics , Marfan Syndrome/genetics , Mental Retardation, X-Linked/genetics , NFI Transcription Factors/genetics , Adolescent , Adult , Child , Chromatin Assembly and Disassembly , Craniofacial Abnormalities/pathology , Exome/genetics , Female , Genetic Predisposition to Disease , Humans , Intellectual Disability/pathology , Male , Marfan Syndrome/pathology , Mental Retardation, X-Linked/pathology , Middle Aged , Mutation/genetics , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Exome Sequencing , Young Adult
5.
Hum Mutat ; 39(12): 1980-1994, 2018 12.
Article in English | MEDLINE | ID: mdl-30168660

ABSTRACT

SH3 and cysteine-rich domain-containing protein 3 (STAC3) is an essential component of the skeletal muscle excitation-contraction coupling (ECC) machinery, though its role and function are not yet completely understood. Here, we report 18 patients carrying a homozygous p.(Trp284Ser) STAC3 variant in addition to a patient compound heterozygous for the p.(Trp284Ser) and a novel splice site change (c.997-1G > T). Clinical severity ranged from prenatal onset with severe features at birth, to a milder and slowly progressive congenital myopathy phenotype. A malignant hyperthermia (MH)-like reaction had occurred in several patients. The functional analysis demonstrated impaired ECC. In particular, KCl-induced membrane depolarization resulted in significantly reduced sarcoplasmic reticulum Ca2+ release. Co-immunoprecipitation of STAC3 with CaV 1.1 in patients and control muscle samples showed that the protein interaction between STAC3 and CaV 1.1 was not significantly affected by the STAC3 variants. This study demonstrates that STAC3 gene analysis should be included in the diagnostic work up of patients of any ethnicity presenting with congenital myopathy, in particular if a history of MH-like episodes is reported. While the precise pathomechanism remains to be elucidated, our functional characterization of STAC3 variants revealed that defective ECC is not a result of CaV 1.1 sarcolemma mislocalization or impaired STAC3-CaV 1.1 interaction.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Substitution , Malignant Hyperthermia/genetics , Myotonia Congenita/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adolescent , Calcium/metabolism , Child , Child, Preschool , Excitation Contraction Coupling , Female , Genetic Predisposition to Disease , Humans , Infant , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Male , Malignant Hyperthermia/etiology , Malignant Hyperthermia/metabolism , Myotonia Congenita/complications , Myotonia Congenita/metabolism , Pedigree , Phenotype , Protein Binding , Protein Transport , Sarcoplasmic Reticulum/metabolism , Severity of Illness Index , Exome Sequencing , Young Adult
6.
Prenat Diagn ; 38(1): 33-43, 2018 01.
Article in English | MEDLINE | ID: mdl-29096039

ABSTRACT

OBJECTIVE: Rare genetic disorders resulting in prenatal or neonatal death are genetically heterogeneous, but testing is often limited by the availability of fetal DNA, leaving couples without a potential prenatal test for future pregnancies. We describe our novel strategy of exome sequencing parental DNA samples to diagnose recessive monogenic disorders in an audit of the first 50 couples referred. METHOD: Exome sequencing was carried out in a consecutive series of 50 couples who had 1 or more pregnancies affected with a lethal or prenatal-onset disorder. In all cases, there was insufficient DNA for exome sequencing of the affected fetus. Heterozygous rare variants (MAF < 0.001) in the same gene in both parents were selected for analysis. Likely, disease-causing variants were tested in fetal DNA to confirm co-segregation. RESULTS: Parental exome analysis identified heterozygous pathogenic (or likely pathogenic) variants in 24 different genes in 26/50 couples (52%). Where 2 or more fetuses were affected, a genetic diagnosis was obtained in 18/29 cases (62%). In most cases, the clinical features were typical of the disorder, but in others, they result from a hypomorphic variant or represent the most severe form of a variable phenotypic spectrum. CONCLUSION: We conclude that exome sequencing of parental samples is a powerful strategy with high clinical utility for the genetic diagnosis of lethal or prenatal-onset recessive disorders. © 2017 The Authors Prenatal Diagnosis published by John Wiley & Sons Ltd.


Subject(s)
Congenital Abnormalities/genetics , Exome Sequencing , Genetic Diseases, Inborn/diagnosis , Parents , Prenatal Diagnosis/methods , Female , Genes, Recessive , Humans , Male , Pregnancy
7.
J Med Genet ; 53(8): 536-47, 2016 08.
Article in English | MEDLINE | ID: mdl-27073233

ABSTRACT

BACKGROUND: The pseudoautosomal short stature homeobox-containing (SHOX) gene encodes a homeodomain transcription factor involved in cell-cycle and growth regulation. SHOX/SHOX enhancers deletions cause short stature and skeletal abnormalities in a female-dominant fashion; duplications appear to be rare. Neurodevelopmental disorders (NDDs), such as autism spectrum disorders (ASDs), are complex disorders with high heritability and skewed sex ratio; several rare (<1% frequency) CNVs have been implicated in risk. METHODS: We analysed data from a discovery series of 90 adult ASD cases, who underwent clinical genetic testing by array-comparative genomic hybridisation (CGH). Twenty-seven individuals harboured CNV abnormalities, including two unrelated females with microduplications affecting SHOX. To determine the prevalence of SHOX duplications and delineate their associated phenotypic spectrum, we subsequently examined array-CGH data from a follow-up sample of 26 574 patients, including 18 857 with NDD (3541 with ASD). RESULTS: We found a significant enrichment of SHOX microduplications in the NDD cases (p=0.00036; OR 2.21) and, particularly, in those with ASD (p=9.18×10(-7); OR 3.63) compared with 12 594 population-based controls. SHOX duplications affecting the upstream or downstream enhancers were enriched only in females with NDD (p=0.0043; OR 2.69/p=0.00020; OR 7.20), but not in males (p=0.404; OR 1.38/p=0.096; OR 2.21). CONCLUSIONS: Microduplications at the SHOX locus are a low penetrance risk factor for ASD/NDD, with increased risk in both sexes. However, a concomitant duplication of SHOX enhancers may be required to trigger a NDD in females. Since specific SHOX isoforms are exclusively expressed in the developing foetal brain, this may reflect the pathogenic effect of altered SHOX protein dosage on neurodevelopment.


Subject(s)
Autism Spectrum Disorder/genetics , DNA Copy Number Variations/genetics , Gene Duplication/genetics , Homeodomain Proteins/genetics , Neurodevelopmental Disorders/genetics , Pseudoautosomal Regions/genetics , Adolescent , Adult , Child , Child, Preschool , Comparative Genomic Hybridization/methods , Female , Genetic Testing/methods , Growth Disorders/genetics , Humans , Male , Middle Aged , Sequence Deletion/genetics , Short Stature Homeobox Protein , Transcription Factors/genetics , Young Adult
8.
Pediatr Dev Pathol ; 19(4): 334-7, 2016.
Article in English | MEDLINE | ID: mdl-26529397

ABSTRACT

We present a case of pure erythroleukemia, diagnosed at autopsy, in a dysmorphic premature infant who died of multiorgan failure within 24 hours of birth. Dysmorphic features included facial and limb abnormalities with long philtrum, microagnathia, downturned mouth, short neck as well as abnormal and missing nails, missing distal phalanx from the second toe, and overlapping toes. Internal findings included gross hepatomegaly and patchy hemorrhages in the liver, splenomegaly, and cardiomegaly; and subdural, intracerebral, and intraventricular hemorrhages. Histology revealed infiltration of bone marrow, kidney, heart, liver, adrenal, lung, spleen, pancreas, thyroid, testis, thymus, and placenta by pure erythroleukemia. Only 6 cases of congenital erythroleukemia have been previously reported with autopsy findings similar to those of this case. The dysmorphic features, although not fitting any specific syndrome, make this case unique. Congenital erythroleukemia and possible syndromes suggested by the dysmorphic features are discussed.


Subject(s)
Abnormalities, Multiple , Infant, Newborn, Diseases , Leukemia, Erythroblastic, Acute/congenital , Humans , Infant, Newborn , Infant, Premature , Male
9.
Eur J Hum Genet ; 23(1): 92-102, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24736735

ABSTRACT

The phenotypic spectrum of GLI3 mutations includes autosomal dominant Greig cephalopolysyndactyly syndrome (GCPS) and Pallister-Hall syndrome (PHS). PHS was first described as a lethal condition associating hypothalamic hamartoma, postaxial or central polydactyly, anal atresia and bifid epiglottis. Typical GCPS combines polysyndactyly of hands and feet and craniofacial features. Genotype-phenotype correlations have been found both for the location and the nature of GLI3 mutations, highlighting the bifunctional nature of GLI3 during development. Here we report on the molecular and clinical study of 76 cases from 55 families with either a GLI3 mutation (49 GCPS and 21 PHS), or a large deletion encompassing the GLI3 gene (6 GCPS cases). Most of mutations are novel and consistent with the previously reported genotype-phenotype correlation. Our results also show a correlation between the location of the mutation and abnormal corpus callosum observed in some patients with GCPS. Fetal PHS observations emphasize on the possible lethality of GLI3 mutations and extend the phenotypic spectrum of malformations such as agnathia and reductional limbs defects. GLI3 expression studied by in situ hybridization during human development confirms its early expression in target tissues.


Subject(s)
Genetic Association Studies , Kruppel-Like Transcription Factors/genetics , Mutation , Nerve Tissue Proteins/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Acrocephalosyndactylia/diagnosis , Acrocephalosyndactylia/genetics , Cohort Studies , DNA Mutational Analysis , Family , Gene Expression , Gene Rearrangement , Haploinsufficiency , Humans , In Situ Hybridization, Fluorescence , Phenotype , Zinc Finger Protein Gli3
10.
EMBO Mol Med ; 6(7): 984-92, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24940003

ABSTRACT

Protocadherin-15 (Pcdh15) is a component of the tip-links, the extracellular filaments that gate hair cell mechano-electrical transduction channels in the inner ear. There are three Pcdh15 splice isoforms (CD1, CD2 and CD3), which only differ by their cytoplasmic domains; they are thought to function redundantly in mechano-electrical transduction during hair-bundle development, but whether any of these isoforms composes the tip-link in mature hair cells remains unknown. By immunolabelling and both morphological and electrophysiological analyses of post-natal hair cell-specific conditional knockout mice (Pcdh15ex38-fl/ex38-fl Myo15-cre+/-) that lose only this isoform after normal hair-bundle development, we show that Pcdh15-CD2 is an essential component of tip-links in mature auditory hair cells. The finding, in the homozygous or compound heterozygous state, of a PCDH15 frameshift mutation (p.P1515Tfs*4) that affects only Pcdh15-CD2, in profoundly deaf children from two unrelated families, extends this conclusion to humans. These results provide key information for identification of new components of the mature auditory mechano-electrical transduction machinery. This will also serve as a basis for the development of gene therapy for deafness caused by PCDH15 defects.


Subject(s)
Cadherins/genetics , Deafness/genetics , Hair Cells, Auditory/cytology , Protein Precursors/genetics , Animals , Cadherin Related Proteins , Cadherins/analysis , Child , Female , Frameshift Mutation , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/ultrastructure , Humans , Male , Mechanotransduction, Cellular , Mice , Mice, Knockout , Mutation , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Precursors/analysis
11.
Am J Med Genet A ; 164A(6): 1537-44, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24668847

ABSTRACT

The 15q13.3 heterozygous microdeletion is a fairly common microdeletion syndrome with marked clinical variability and incomplete penetrance. The average size of the deletion, which comprises six genes including CHRNA7, is 1.5 Mb. CHRNA7 has been identified as the gene responsible for the neurological phenotype in this microdeletion syndrome. Only seven patients with a homozygous microdeletion that includes at least CHRNA7, and is inherited from both parents have been described in the literature. The aim of this study was to further describe the distinctive eye manifestations from the analysis in the three French patients diagnosed with the classical 1.5 Mb homozygous microdeletion. Patients' ages ranged from 30 months to 9 years, and included one sib pair. They all displayed a remarkably severe identifiable clinical phenotype that included congenital blindness and convulsive encephalopathy with inconstant abnormal movements. The ophthalmological examination revealed a lack of eye tracking, optic nerve pallor, an immature response with increased latencies with no response to the checkerboard stimulations at the visual evoked potential examination, and a distinctive retina dystrophy with a negative electroretinogram in which the "b" wave was smaller than the "a" wave after a dark adapted pupil and bright flash in all patients. Clear genotype-phenotype correlations emerged, showing that this eye phenotype was secondary to homozygous deletion of TRPM1, the gene responsible for autosomal recessive congenital stationary night blindness. The main differential diagnosis is ceroid lipofuscinosis.


Subject(s)
Blindness/genetics , Chromosome Disorders/genetics , Intellectual Disability/genetics , Neuronal Ceroid-Lipofuscinoses/genetics , Seizures/genetics , TRPM Cation Channels/genetics , alpha7 Nicotinic Acetylcholine Receptor/genetics , Child , Child, Preschool , Chromosome Deletion , Chromosome Disorders/pathology , Chromosomes, Human, Pair 15/genetics , Electroretinography , Eye/pathology , Eye Abnormalities/genetics , Eye Diseases, Hereditary/genetics , Female , Genetic Association Studies , Genetic Diseases, X-Linked/genetics , Humans , Intellectual Disability/pathology , Male , Myopia/genetics , Neuronal Ceroid-Lipofuscinoses/pathology , Night Blindness/genetics , Optic Nerve/abnormalities , Retinal Dystrophies/genetics , Seizures/pathology
12.
Eur J Hum Genet ; 22(2): 289-92, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23674175

ABSTRACT

Intellectual disability (ID) is frequent in the general population, with 1 in 50 individuals directly affected worldwide. The multiple etiologies include X-linked ID (XLID). Among syndromic XLID, few syndromes present severe ID associated with postnatal microcephaly and midline stereotypic hand movements. We report on three male patients with ID, midline stereotypic hand movements, hypotonia, hyperkinesia, strabismus, as well as seizures (2/3), and non-inherited and postnatal onset microcephaly (2/3). Using array CGH and exome sequencing we characterised two truncating mutations in IQSEC2, namely two de novo intragenic duplication mapped to the Xp11.22 region and a nonsense mutation in exon 7. We propose that truncating mutations in IQSEC2 are responsible for syndromic severe ID in male patients and should be screened in patients without mutations in MECP2, FOXG1, CDKL5 and MEF2C.


Subject(s)
Abnormalities, Multiple/diagnosis , Guanine Nucleotide Exchange Factors/genetics , Intellectual Disability/diagnosis , Abnormalities, Multiple/genetics , Adult , Child, Preschool , Codon, Nonsense , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Intellectual Disability/classification , Intellectual Disability/genetics , Male , Phenotype
13.
Hum Genet ; 133(3): 367-77, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24178751

ABSTRACT

Oral-facial-digital syndrome type VI (OFD VI) is a recessive ciliopathy defined by two diagnostic criteria: molar tooth sign (MTS) and one or more of the following: (1) tongue hamartoma (s) and/or additional frenula and/or upper lip notch; (2) mesoaxial polydactyly of one or more hands or feet; (3) hypothalamic hamartoma. Because of the MTS, OFD VI belongs to the "Joubert syndrome related disorders". Its genetic aetiology remains largely unknown although mutations in the TMEM216 gene, responsible for Joubert (JBS2) and Meckel-Gruber (MKS2) syndromes, have been reported in two OFD VI patients. To explore the molecular cause(s) of OFD VI syndrome, we used an exome sequencing strategy in six unrelated families followed by Sanger sequencing. We identified a total of 14 novel mutations in the C5orf42 gene in 9/11 families with positive OFD VI diagnostic criteria including a severe fetal case with microphthalmia, cerebellar hypoplasia, corpus callosum agenesis, polydactyly and skeletal dysplasia. C5orf42 mutations have already been reported in Joubert syndrome confirming that OFD VI and JBS are allelic disorders, thus enhancing our knowledge of the complex, highly heterogeneous nature of ciliopathies.


Subject(s)
Membrane Proteins/genetics , Orofaciodigital Syndromes/diagnosis , Orofaciodigital Syndromes/genetics , Abnormalities, Multiple , Adolescent , Adult , Alleles , Cerebellar Diseases/diagnosis , Cerebellar Diseases/genetics , Cerebellum/abnormalities , Child , Developmental Disabilities/diagnosis , Developmental Disabilities/genetics , Exome , Eye Abnormalities/diagnosis , Eye Abnormalities/genetics , Female , Hamartoma/diagnosis , Hamartoma/genetics , Humans , Hypothalamic Diseases/diagnosis , Hypothalamic Diseases/genetics , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/genetics , Male , Mutation , Nervous System Malformations/diagnosis , Nervous System Malformations/genetics , Phenotype , Polydactyly/diagnosis , Polydactyly/genetics , Retina/abnormalities , Sequence Analysis, DNA , Young Adult
14.
Eur J Med Genet ; 56(10): 556-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23933090

ABSTRACT

Kabuki syndrome (KS) is a rare syndrome associating malformations with intellectual deficiency and numerous visceral, orthopedic, endocrinological, immune and autoimmune complications. The early establishment of a diagnostic of KS leads to better care of the patients and therefore prevents complications such as perception deafness, severe complications of auto-immune diseases or obesity. However, the diagnosis of KS remains difficult because based on the appreciation of facial features combined with other highly variable features. We describe a novel sign, namely the attenuation and/or congenital absence of the IPD crease of the third and fourth fingers associated with limitation of flexion of the corresponding joints, which seems to be specific of KS and could help the clinician to diagnose KS.


Subject(s)
Abnormalities, Multiple/diagnosis , Face/abnormalities , Fingers/abnormalities , Hematologic Diseases/diagnosis , Vestibular Diseases/diagnosis , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Male
15.
J Pediatr Ophthalmol Strabismus ; 50 Online: e33-6, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23819954

ABSTRACT

The authors report a 5-year follow-up examination of two sisters diagnosed as having a juvenile form of type II sialidosis. Diagnosis occurred during a routine ophthalmic examination when the girls were 5 and 3 years old after bilateral macular cherry-red spots were revealed. Main clinical findings were hypotonia, hepatosplenomegaly, hearing loss, dysostosis, and respiratory distress. Ophthalmic symptoms were low visual acuity and nystagmus. Spectral-domain optical coherence tomography examination showed increased reflectivity of the retinal ganglion cells. Sialidosis may present as a mild form with slow progression. The cherry-red spots may be the first clue for proper diagnosis of storage disease. Spectral-domain optical coherence tomography examination unveiled the accumulation of sialic acid in the retinal ganglion cells and could potentially be used to monitor the progression of storage diseases.


Subject(s)
Eye Diseases/diagnosis , Mucolipidoses/diagnosis , Child, Preschool , Diagnosis, Differential , Female , Follow-Up Studies , Humans , Infant , Retinal Ganglion Cells/pathology , Tomography, Optical Coherence
16.
Blood ; 120(1): 86-9, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22611161

ABSTRACT

Fanconi anemia (FA) is a rare bone marrow failure disorder with defective DNA interstrand crosslink repair. Still, there are FA patients without mutations in any of the 15 genes individually underlying the disease. A candidate protein for those patients, FA nuclease 1 (FAN1), whose gene is located at chromosome 15q13.3, is recruited to stalled replication forks by binding to monoubiquitinated FANCD2 and is required for interstrand crosslink repair, suggesting that mutation of FAN1 may cause FA. Here we studied clinical, cellular, and genetic features in 4 patients carrying a homozygous 15q13.3 micro-deletion, including FAN1 and 6 additional genes. Biallelic deletion of the entire FAN1 gene was confirmed by failure of 3'- and 5'-PCR amplification. Western blot analysis failed to show FAN1 protein in the patients' cell lines. Chromosome fragility was normal in all 4 FAN1-deficient patients, although their cells showed mild sensitivity to mitomycin C in terms of cell survival and G(2) phase arrest, dissimilar in degree to FA cells. Clinically, there were no symptoms pointing the way to FA. Our results suggest that FAN1 has a minor role in interstrand crosslink repair compared with true FA genes and exclude FAN1 as a novel FA gene.


Subject(s)
DNA Repair/physiology , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/physiology , Fanconi Anemia/genetics , Fanconi Anemia/physiopathology , Cell Survival/physiology , Cells, Cultured , Child , Child, Preschool , Chromosomes, Human, Pair 15 , DNA Replication/physiology , Endodeoxyribonucleases , Fanconi Anemia/pathology , Gene Deletion , Homozygote , Humans , Infant , Multifunctional Enzymes
17.
Am J Hum Genet ; 90(5): 907-14, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22560091

ABSTRACT

Auriculocondylar syndrome (ACS) is a rare, autosomal-dominant craniofacial malformation syndrome characterized by variable micrognathia, temporomandibular joint ankylosis, cleft palate, and a characteristic "question-mark" ear malformation. Careful phenotypic characterization of severely affected probands in our cohort suggested the presence of a mandibular patterning defect resulting in a maxillary phenotype (i.e., homeotic transformation). We used exome sequencing of five probands and identified two novel (exclusive to the patient and/or family studied) missense mutations in PLCB4 and a shared mutation in GNAI3 in two unrelated probands. In confirmatory studies, three additional novel PLCB4 mutations were found in multigenerational ACS pedigrees. All mutations were confirmed by Sanger sequencing, were not present in more than 10,000 control chromosomes, and resulted in amino-acid substitutions located in highly conserved protein domains. Additionally, protein-structure modeling demonstrated that all ACS substitutions disrupt the catalytic sites of PLCB4 and GNAI3. We suggest that PLCB4 and GNAI3 are core signaling molecules of the endothelin-1-distal-less homeobox 5 and 6 (EDN1-DLX5/DLX6) pathway. Functional studies demonstrated a significant reduction in downstream DLX5 and DLX6 expression in ACS cases in assays using cultured osteoblasts from probands and controls. These results support the role of the previously implicated EDN1-DLX5/6 pathway in regulating mandibular specification in other species, which, when disrupted, results in a maxillary phenotype. This work defines the molecular basis of ACS as a homeotic transformation (mandible to maxilla) in humans.


Subject(s)
Ear Diseases/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Mutation , Phospholipase C beta/genetics , Amino Acid Sequence , Cohort Studies , Ear/abnormalities , Ear/physiopathology , Ear Diseases/physiopathology , Endothelin-1/genetics , Endothelin-1/metabolism , Exome , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Molecular Sequence Data , Pedigree , Phenotype , Phospholipase C beta/metabolism , Protein Conformation , Sequence Analysis, RNA
18.
Am J Hematol ; 87(2): 208-10, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22139979

ABSTRACT

We present a rapid strategy based on Restriction Fragment Length Polymorphism (RFLP) analysis to characterize the more frequent glucose 6-phosphate dehydrogenase (G6PD) variants observed in a population with high gene flow. During a study involving more than 600 patients, we observed mainly G6PD A(-) (c.202G>A, c.376A>G; p.Val68Met, p.Asn126Asp), G6PD Mediterranean (Med) (c.563C>T, p.Ser188Phe), and G6PD Betica (c.376A>G, 542A>T; p.126Asn>Asp, 181Asp>Val) with addition of a few rare ones. A number of 10 abnormalities amounted to 92% of all the molecular defects. In addition, seven new mutations were found: three presented with acute hemolytic anemia following oxidative stress [G6PD Nice (c.1380G>C, p.Glu460Asp), G6PD Roubaix (c.811G>C, p.Val271Leu), and G6PD Toledo (c.496C>T, p.Arg166Cys)], three with different degrees of chronic hemolytic anemia [G6PD Lille (c.821A>T, p.Glu274Val), G6PD Villeurbanne (c.1000_1002delACC, p.Thr334del), and G6PD Amiens (c.1367A>T, p.Asp456Val)] and one found fortuitously G6PD Montpellier (c.1132G>A, p.Gly378Ser).


Subject(s)
Ethnicity , Glucosephosphate Dehydrogenase Deficiency/ethnology , Glucosephosphate Dehydrogenase Deficiency/genetics , Glucosephosphate Dehydrogenase/genetics , Mutation , Exons , France/epidemiology , Glucosephosphate Dehydrogenase Deficiency/pathology , Humans , Isoenzymes/genetics , Male , Models, Molecular , Polymorphism, Restriction Fragment Length , Protein Multimerization , Protein Structure, Quaternary
19.
Pediatrics ; 128(3): e703-6, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21807695

ABSTRACT

This article focuses on the case of a newborn infant boy with bilateral choanal atresia, tracheoesophageal fistula, and bilateral fifth-finger clinodactyly. This infant had been exposed to carbimazole in utero during the treatment of maternal Graves disease. Teratogenic defects caused by carbimazole were recently recognized, and their phenotypes have been defined. Choanal atresia, esophageal atresia, athelia or hypothelia, developmental delay, hearing loss, and dysmorphic facial features have all been reported. To our knowledge, this is the first documented case of tracheoesophageal fistula without esophageal atresia (H type). Knowledge of the teratogenic potential of carbimazole is important when managing Graves disease in women of childbearing age.


Subject(s)
Antithyroid Agents/adverse effects , Carbimazole/adverse effects , Choanal Atresia/chemically induced , Prenatal Exposure Delayed Effects/chemically induced , Tracheoesophageal Fistula/chemically induced , Antithyroid Agents/therapeutic use , Carbimazole/therapeutic use , Choanal Atresia/complications , Choanal Atresia/diagnosis , Endoscopy , Female , Graves Disease/drug therapy , Humans , Infant, Newborn , Male , Pregnancy , Tracheoesophageal Fistula/complications
20.
Clin J Am Soc Nephrol ; 6(1): 22-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20876674

ABSTRACT

BACKGROUND AND OBJECTIVES: Bardet-Biedl Syndrome (BBS) is a rare autosomal recessive ciliopathy with a wide spectrum of clinical features including obesity, retinitis pigmentosa, polydactyly, mental retardation, hypogonadism, and renal abnormalities. The molecular genetic profile of BBS is currently being investigated after the recent identification of 14 BBS genes involved in primary cilia-linked disease. This study aims to characterize the renal and cardiovascular presentations and to analyze possible relationships between genotypes and clinical phenotypes. DESIGN, SETTING, PARTICIPANTS & MEASUREMENTS: This clinical study was performed in a national cohort of 33 BBS patients, 22 men and 11 women, all aged >16 years (mean age 26.3 years). RESULTS: Renal abnormalities, including impairment of renal function and signs of chronic interstitial nephropathy of dysplastic nature, were documented in 82% of the patients. Cardiovascular evaluations revealed that this group of young patients had significant cardiovascular risk factors. Hypertension was found in >30% of the patients and hyperlipidemia in >60%, and almost 50% had other metabolic abnormalities. Overt diabetes was present in only 6%. With regard to genotype-phenotype correlation, patients with a mutation in the BBS6, BBS10, or BBS12 gene (10 of 33 patients) had more severe renal disease. CONCLUSIONS: Our study results confirm the frequent occurrence of renal involvement in patients with BBS, underscore the high risk of cardiovascular disease in these patients, and provide new information on a possible genotype-phenotype correlation.


Subject(s)
Bardet-Biedl Syndrome/complications , Cardiovascular Diseases/etiology , Kidney Diseases/etiology , Adult , Bardet-Biedl Syndrome/physiopathology , Cohort Studies , Creatinine/urine , Female , Genotype , Glucose/metabolism , Humans , Kidney/diagnostic imaging , Kidney/physiopathology , Male , Microtubule-Associated Proteins , Phenotype , Proteins/genetics , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL
...