Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Country/Region as subject
Publication year range
2.
Hum Genet ; 137(10): 831-846, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30291432

ABSTRACT

There is an increasing implication of non-coding regions in pathological processes of genetic origin. This is partly due to the emergence of sophisticated techniques that have transformed research into gene expression by allowing a more global understanding of the genome, both at the genomic, epigenomic and chromatin levels. Here, we implemented the analysis of PAX6, whose coding loss-of-function variants are mainly implied in aniridia, by studying its non-coding regions (untranslated regions, introns and cis-regulatory sequences). In particular, we have taken advantage of the development of high-throughput approaches to screen the upstream and downstream regulatory regions of PAX6 in 47 aniridia patients without identified mutation in the coding sequence. This was made possible through the use of custom targeted resequencing and/or CGH array to analyze the entire PAX6 locus on 11p13. We found candidate variants in 30 of the 47 patients. 9/30 correspond to the well-known described 3' deletions encompassing SIMO and other enhancer elements. In addition, we identified numerous different variants in various non-coding regions, in particular untranslated regions. Among these latter, most of them demonstrated an in vitro functional effect using a minigene strategy, and 12/21 are thus considered as causative mutations or very likely to explain the phenotypes. This new analysis strategy brings molecular diagnosis to more than 90% of our aniridia patients. This study revealed an outstanding mutation pattern in non-coding PAX6 regions confirming that PAX6 remains the major gene for aniridia.


Subject(s)
3' Untranslated Regions , Aniridia/genetics , Enhancer Elements, Genetic , Genetic Loci , Mutation , PAX6 Transcription Factor/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Male , Middle Aged
3.
Clin Genet ; 93(4): 837-845, 2018 04.
Article in English | MEDLINE | ID: mdl-29136273

ABSTRACT

Microphthalmia and anophthalmia (MA) are severe developmental eye anomalies, many of which are likely to have an underlying genetic cause. More than 30 genes have been described, each of which is responsible for a small percentage of these anomalies. Among these, is the FOXE3 gene, which was initially described in individuals with dominantly inherited anterior segment dysgenesis and, subsequently, associated with recessively inherited primary aphakia, sclerocornea and microphthalmia. In this work, we describe 8 individuals presenting with an MA phenotype. Among them, 7 are carrying biallelic recessive FOXE3 mutations and 2 of these have novel mutations: p.(Ala78Thr) and p.(Arg104Cys). The last of our patients is carrying in the heterozygous state the recessive p.(Arg90Leu) mutation in the FOXE3 gene. To further understand FOXE3 involvement in this wide spectrum of ocular anomalies with 2 different patterns of inheritance, we reviewed all individuals with ocular abnormalities described in the literature for which a FOXE3 mutation was identified. This review demonstrates that correlations exist between the mutation type, mode of inheritance and the phenotype severity. Furthermore, understanding the genetic basis of these conditions will contribute to overall understanding of eye development, improve the quality of care, genetic counseling and, in future, gene-based therapies.


Subject(s)
Aphakia/genetics , Forkhead Transcription Factors/genetics , Genetic Predisposition to Disease , Microphthalmos/genetics , Alleles , Aphakia/physiopathology , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Eye Abnormalities/genetics , Eye Abnormalities/physiopathology , Female , Humans , Male , Microphthalmos/physiopathology , Mutation
4.
Clin Genet ; 92(5): 477-486, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28105635

ABSTRACT

WNT10A gene encodes a canonical wingless pathway signaling molecule involved in cell fate specification as well as morphogenetic patterning of the developing ectoderm, nervous system, skeleton, and tooth. In patients, WNT10A mutations are responsible for ectodermal-derived pathologies including isolated hypo-oligodontia, tricho-odonto-onycho-dermal dysplasia and Schöpf-Schulz-Passarge syndrome (SSPS). Here we describe the dental, ectodermal, and extra-ectodermal phenotypic features of a cohort of 41 patients from 32 unrelated families. Correlations with WNT10A molecular status (heterozygous carrier, compound heterozygous, homozygous) and patient's phenotypes were performed. Mild to severe oligodontia was observed in all patients bearing biallelic WNT10A mutations. However, patients with compound heterozygous mutations presented no significant difference in phenotypes compared with homozygous individuals. Anomalies in tooth morphology were frequently observed with heterozygous patients displaying hypodontia. No signs of SSPS, especially eyelids cysts, were detected in our cohort. Interestingly, extra-ectodermal signs consisted of skeletal, neurological and vascular anomalies, the latter suggesting a wider phenotypic spectrum associated with WNT10A mutations. Indeed, the Wnt pathway plays a crucial role in skeletal development, lipid metabolism, and neurogenesis, potentially explaining patient's clinical manifestations.


Subject(s)
Genetic Association Studies , Mutation/genetics , Tooth/pathology , Wnt Proteins/genetics , Adolescent , Adult , Child , Cohort Studies , Cone-Beam Computed Tomography , Ectoderm/pathology , Heterozygote , Homozygote , Humans , Mandible/pathology , Middle Aged , Phenotype , Tooth/diagnostic imaging , Young Adult
5.
Clin Genet ; 90(1): 79-83, 2016 07.
Article in English | MEDLINE | ID: mdl-26586152

ABSTRACT

Olfactory bulb (OB) volume evaluation by magnetic resonance imaging (MRI) has been demonstrated to be related to olfactory dysfunction in many different diseases. Olfactory dysfunction is often overlooked in Bardet-Biedl syndrome (BBS) patients and is rarely objectively evaluated by MRI. We present a series of 20 BBS patients with olfactory dysfunction. The OB was evaluated separately and blindly by two radiologists (SR and SM) with 3 Tesla MRI imaging comparatively to 12 normal control subjects by global visual evaluation and by quantitative measurement of OB volume. In the 12 control cases OB visual evaluation was considered as normal in all cases for radiologist (SR) and in 10 cases for radiologist (SM). In the 20 BBS patients, OB visual evaluation was considered as abnormal in 18 cases for SR and in all cases for SM. OB volumetric evaluation for SR and SM in BBS patients was able to provide significant correlation between BBS and olfactory dysfunction. This study indicates that OB volume evaluation by MRI imaging like structural MRI scan for gray matter modifications demonstrates that olfactory dysfunction in BBS patients is a constant and cardinal symptom integrated in a genetical syndrome with peripheral and central olfactory structure alterations.


Subject(s)
Bardet-Biedl Syndrome/diagnostic imaging , Microtubule-Associated Proteins/genetics , Mutation , Olfaction Disorders/diagnostic imaging , Olfactory Bulb/diagnostic imaging , Adolescent , Adult , Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/pathology , Case-Control Studies , Female , Gene Expression , Humans , Magnetic Resonance Imaging , Male , Microtubule-Associated Proteins/metabolism , Middle Aged , Multigene Family , Olfaction Disorders/genetics , Olfaction Disorders/pathology , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Organ Size/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Smell/physiology
6.
Clin Genet ; 87(2): 161-6, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24400638

ABSTRACT

Bardet-Biedl syndrome (BBS), a ciliopathy disorder with pleiotropic effect manifests primarily as retinal degeneration along with renal insufficiency, polydactyly and obesity. In this study, we have performed homozygosity mapping using NspI 250K affymetrix gene chip followed by mutation screening of the candidate genes located in the homozygous blocks. These regions are prioritized based on the block length and candidature of the genes in BBS and other ciliopathies. Gene alterations in known BBS (22) and other ciliopathy genes such as ALMS1 (2) were seen in 24 of 30 families (80%). Mutations in BBS3 gene, inclusive of a novel recurrent mutation (p.I91T) accounted for 18% of the identified variations. Disease associated polymorphisms p.S70N (BBS2), rs1545 and rs1547 (BBS6) were also observed. This is the first study in Indian BBS patients and homozygosity mapping has proved to be an effective tool in prioritizing the candidate genes in consanguineous pedigrees. The study reveals a different mutation profile in the ciliopathy genes in Indian population and implication of novel loci/genes in 20% of the study group.


Subject(s)
ADP-Ribosylation Factors/genetics , Bardet-Biedl Syndrome/genetics , Group II Chaperonins/genetics , Neoplasm Proteins/genetics , Proteins/genetics , Bardet-Biedl Syndrome/physiopathology , Cell Cycle Proteins , Chaperonins , Chromosome Mapping , Cytoskeletal Proteins , Female , Genetic Association Studies , Homozygote , Humans , India , Male , Mutation , Polymorphism, Single Nucleotide , Renal Insufficiency/genetics , Renal Insufficiency/physiopathology , Retinal Degeneration/genetics , Retinal Degeneration/physiopathology
7.
Clin Genet ; 87(5): 430-9, 2015 May.
Article in English | MEDLINE | ID: mdl-24890733

ABSTRACT

WFS1 mutations are responsible for Wolfram syndrome (WS) characterized by juvenile-onset diabetes mellitus and optic atrophy, and for low-frequency sensorineural hearing loss (LFSNHL). Our aim was to analyze the French cohort of 96 patients with WFS1-related disorders in order (i) to update clinical and molecular data with 37 novel affected individuals, (ii) to describe uncommon phenotypes and, (iii) to precise the frequency of large-scale rearrangements in WFS1. We performed quantitative polymerase chain reaction (PCR) in 13 patients, carrying only one heterozygous variant, to identify large-scale rearrangements in WFS1. Among the 37 novel patients, 15 carried 15 novel deleterious putative mutations, including one large deletion of 17,444 base pairs. The analysis of the cohort revealed unexpected phenotypes including (i) late-onset symptoms in 13.8% of patients with a probable autosomal recessive transmission; (ii) two siblings with recessive optic atrophy without diabetes mellitus and, (iii) six patients from four families with dominantly-inherited deafness and optic atrophy. We highlight the expanding spectrum of WFS1-related disorders and we show that, even if large deletions are rare events, they have to be searched in patients with classical WS carrying only one WFS1 mutation after sequencing.


Subject(s)
Genetic Association Studies , Membrane Proteins/genetics , Mutation , Phenotype , Wolfram Syndrome/diagnosis , Wolfram Syndrome/genetics , Adolescent , Adult , Amino Acid Substitution , Child , Cohort Studies , Family , Female , France , Genes, Dominant , Genes, Recessive , Genotype , Humans , Male , Middle Aged , Young Adult
8.
Ann Dermatol Venereol ; 142(6-7): 399-409, 2015.
Article in French | MEDLINE | ID: mdl-25952794

ABSTRACT

BACKGROUND: Leucoderma is a frequent presenting complaint in children and it is sometimes difficult to make a definite diagnostic during the first consultation. The aim of this study is to analyse the diagnoses associated with leucoderma in children in order to propose a practical approach to their differential diagnosis. MATERIAL AND METHODS: We performed a review of the literature using the keywords "leucoderma children review", "leucoderma Ito" and "nevus depigmentosus" in the Medline database. All relevant articles were included. RESULTS: Four hundred and thirty-five articles were retrieved and 179 were analysed. A clinical approach was proposed in 6 articles and investigations in 15 articles. DISCUSSION: Causal diagnosis of leucoderma may frequently be made on clinical grounds by determining the age of onset and distribution of lesions. Nevertheless, some situations require investigation. The literature is limited regarding clinical approaches and examinations in leucoderma. Herein, we present a systematic clinical and laboratory approach to the differential diagnosis of these skin disorders.


Subject(s)
Hypopigmentation , Abnormalities, Multiple/genetics , Adolescent , Age of Onset , Albinism/epidemiology , Albinism/genetics , Child , Child, Preschool , Diagnosis, Differential , Genetic Counseling , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/epidemiology , Genetic Diseases, Inborn/genetics , Humans , Hypopigmentation/classification , Hypopigmentation/congenital , Hypopigmentation/diagnosis , Hypopigmentation/etiology , Hypopigmentation/genetics , Infant , Infant, Newborn , Intellectual Disability/genetics , Mutation , Sequence Deletion , Symptom Assessment , Syndrome
9.
Clin Genet ; 85(2): 172-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23432027

ABSTRACT

Bardet-Biedl syndrome (BBS, OMIM 209900) is a rare genetic disorder characterized by obesity, retinitis pigmentosa, post axial polydactyly, cognitive impairment, renal anomalies and hypogonadism. The aim of this study is to provide a comprehensive clinical and molecular analysis of a cohort of 11 Tunisian BBS consanguineous families in order to give insight into clinical and genetic spectrum and the genotype-phenotype correlations. Molecular analysis using combined sequence capture and high-throughput sequencing of 30 ciliopathies genes revealed 11 mutations in 11 studied families. Five mutations were novel and six were previously described. Novel mutations included c.1110G>A and c.39delA (p.G13fs*41) in BBS1, c.115+5G>A in BBS2, c.1272+1G>A in BBS6, c.1181_1182insGCATTTATACC in BBS10 (p.S396Lfs*6). Described mutations included c.436C>T (p.R146*) and c.1473+4A>G in BBS1, c.565C> (p.R189*) in BBS2, deletion of exons 4-6 in BBS4, c.149T>G (p.L50R) in BBS5, and c.459+1G>A in BBS8; most frequent mutations were described in BBS1 (4/11, 37%) and BBS2 (2/11, 18%) genes. No phenotype-genotype correlation was evidenced. This data expands the mutations profile of BBS genes in Tunisia and suggests a divergence of the genetic spectrum comparing Tunisian and other populations.


Subject(s)
Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/pathology , Group II Chaperonins/genetics , Microtubule-Associated Proteins/genetics , Phenotype , Proteins/genetics , Base Sequence , Chaperonins , Computational Biology , High-Throughput Nucleotide Sequencing , Humans , Molecular Sequence Data , Tunisia
10.
Clin Genet ; 86(6): 521-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24684473

ABSTRACT

Bardet-Biedl syndrome (BBS) is a well-recognized ciliopathy characterized by cardinal features namely: early onset retinitis pigmentosa, polydactyly, obesity, hypogonadism, renal and cognitive impairment. Recently, disorders of olfaction (anosmia, hyposmia) have been also described in BBS patients. Moreover, morphological brain anomalies have been reported and prompt for further investigations to determine whether they are primary or secondary to peripheral organ involvement (i.e. visual or olfactory neuronal tissue). The objective of this article is to evaluate olfactory disorders in BBS patients and to investigate putative correlation with morphological cerebral anomalies. To this end, 20 BBS patients were recruited and evaluated for olfaction using the University of Pennsylvania Smell Identification Test (UPSIT). All of them underwent a structural magnetic resonance imaging (MRI) scan. We first investigated brain morphological differences between BBS subjects and 14 healthy volunteers. Then, we showed objective olfaction disorders in BBS patients and highlight correlation between gray matter volume reduction and olfaction dysfunction in several brain areas.


Subject(s)
Bardet-Biedl Syndrome/physiopathology , Brain/pathology , Olfaction Disorders/etiology , Adolescent , Adult , Atrophy , Case-Control Studies , Female , Humans , Magnetic Resonance Imaging , Male , Retinitis Pigmentosa/etiology , Smell/physiology , Young Adult
11.
Clin Genet ; 85(5): 476-81, 2014 May.
Article in English | MEDLINE | ID: mdl-23692385

ABSTRACT

Ciliopathies are heterogeneous disorders sharing different clinical signs due to a defect at the level of the primary cilia/centrosome complex. Postaxial polydactyly is frequently reported in ciliopathies, especially in Bardet-Biedl syndrome (BBS). Clinical features and genetic results observed in a pair of dizygotic twins with BBS are reported. The following manifestations were present: retinitis pigmentosa, bilateral insertional polydactyly, cognitive impairment and renal dysfunction. X-rays of the hands confirmed the presence of a 4th mesoaxial extra-digit with Y-shaped metacarpal bones. The sequencing of LZTFL1 identified a missense mutation (NM_020347.2: p.Leu87Pro; c.260T>C) and a nonsense mutation (p.Glu260*; c.778G>T), establishing a compound heterozygous status for the twins. A major decrease of LZTFL1 transcript and protein was observed in the patient's fibroblasts. This is the second report of LZTFL1 mutations in BBS patients confirming LZTFL1 as a BBS gene. Interestingly, the only two families reported in literature thus far with LZTFL1 mutations have in common mesoaxial polydactyly, a very uncommon feature for BBS. This special subtype of polydactyly in BBS patients is easily identified on clinical examination and prompts for priority sequencing of LZTFL1 (BBS17).


Subject(s)
Bardet-Biedl Syndrome/genetics , Fingers/abnormalities , Mutation/genetics , Polydactyly/genetics , Toes/abnormalities , Transcription Factors/genetics , Adult , Bardet-Biedl Syndrome/physiopathology , DNA Mutational Analysis , Female , Fingers/physiopathology , Heterozygote , Humans , Pedigree , Polydactyly/physiopathology , Toes/physiopathology , Twins
12.
Clin Genet ; 86(4): 326-34, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24033328

ABSTRACT

Anophthalmia and microphthalmia (AM) are the most severe malformations of the eye, corresponding respectively to reduced size or absent ocular globe. Wide genetic heterogeneity has been reported and different genes have been demonstrated to be causative of syndromic and non-syndromic forms of AM. We screened seven AM genes [GDF6 (growth differentiation factor 6), FOXE3 (forkhead box E3), OTX2 (orthodenticle protein homolog 2), PAX6 (paired box 6), RAX (retina and anterior neural fold homeobox), SOX2 (SRY sex determining region Y-box 2), and VSX2 (visual system homeobox 2 gene)] in a cohort of 150 patients with isolated or syndromic AM. The causative genetic defect was identified in 21% of the patients (32/150). Point mutations were identified by direct sequencing of these genes in 25 patients (13 in SOX2, 4 in RAX, 3 in OTX2, 2 in FOXE3, 1 in VSX2, 1 in PAX6, and 1 in GDF6). In addition eight gene deletions (five SOX2, two OTX2 and one RAX) were identified using a semi-quantitative multiplex polymerase chain reaction (PCR) [quantitative multiplex PCR amplification of short fluorescent fragments (QMPSF)]. The causative genetic defect was identified in 21% of the patients. This result contributes to our knowledge of the molecular basis of AM, and will facilitate accurate genetic counselling.


Subject(s)
Anophthalmos/genetics , Genetic Heterogeneity , Microphthalmos/genetics , Point Mutation/genetics , Adolescent , Adult , Anophthalmos/diagnosis , Anophthalmos/pathology , Child , Child, Preschool , Eye Proteins/genetics , Female , Forkhead Transcription Factors/genetics , Growth Differentiation Factor 6/genetics , Homeodomain Proteins/genetics , Humans , Infant , Male , Microphthalmos/diagnosis , Microphthalmos/pathology , Otx Transcription Factors/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , SOXB1 Transcription Factors/genetics , Transcription Factors/genetics
13.
Dermatology ; 226(4): 353-7, 2013.
Article in English | MEDLINE | ID: mdl-23899764

ABSTRACT

The follow-up of a man from birth to adulthood, presenting with features both of RAPADILINO and Rothmund-Thomson syndrome (RTS), is described. Molecular studies confirmed the presence of two different mutations, c.2767_2768delTT and c.3061C>T, in the RECQL4 gene. This gene is known to be causative of a spectrum including Baller-Gerold syndrome, RAPADILINO syndrome and RTS. New and rare features such as oral leukoplakia and very prominent hyperkeratotic verrucous papules on both soles are shown. This patient has to date no cancer history despite bearing a truncating mutation at the age of 21 years, which is also unusual.


Subject(s)
Abnormalities, Multiple/genetics , Alopecia/genetics , Anal Canal/abnormalities , Anodontia/genetics , Dwarfism/genetics , Heart Septal Defects, Atrial/genetics , Limb Deformities, Congenital/genetics , Patella/abnormalities , Pigmentation Disorders/genetics , Radius/abnormalities , RecQ Helicases/genetics , Rothmund-Thomson Syndrome/genetics , Thumb/abnormalities , Adult , Child , Humans , Infant, Newborn , Keratosis/genetics , Male , Mutation , Pedigree , Time Factors , Young Adult
14.
Nat Genet ; 5(3): 308-11, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8275096

ABSTRACT

Stargardt's disease (fundus flavimaculatus) is one of the most frequent causes of macular degeneration in childhood and accounts for 7% of all retinal dystrophies. It is an autosomal recessive condition characterized by a bilateral loss of central vision occurring at age 7-12 years. Genetic linkage analysis of eight families has assigned the disease locus to chromosome 1p21-p13. Multipoint linkage analysis and haplotype analysis has allowed us to establish the best estimate for location of the gene over the locus D1S435 (maximum lod score of 12.66). Our results are consistent with the genetic homogeneity of this condition.


Subject(s)
Chromosomes, Human, Pair 1 , Retinal Diseases/genetics , Child , Chromosome Mapping , Female , Genes, Recessive , Genetic Linkage , Haplotypes , Humans , Male , Recombination, Genetic
15.
Nat Genet ; 14(4): 461-4, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8944027

ABSTRACT

Leber's congenital amaurosis (LCA, MIM 204,000), the earliest and most severe form of inherited retinopathy, accounts for at least 5% of all inherited retinal dystrophies. This autosomal recessive condition is usually recognized at birth or during the first months of life in an infant with total blindness or greatly impaired vision, normal fundus and extinguished electroretinogram (ERG). Nystagmus (pendular type) and characteristic eye poking are frequently observed in the first months of life (digito-ocular sign of Franceschetti). Hypermetropia and keratoconus frequently develop in the course of the disease. The observation by Waardenburg of normal children born to affected parents supports the genetic heterogeneity of LCA. Until now, however, little was known about the pathophysiology of the disease, but LCA is usually regarded as the consequence of either impaired development of photoreceptors or extremely early degeneration of cells that have developed normally. We have recently mapped a gene for LCA to chromosome 17p13.1 (LCA1) by homozygosity mapping in consanguineous families of North African origin and provided evidence of genetic heterogeneity in our sample, as LCA1 accounted for 8/15 LCA families in our series. Here, we report two missense mutations (F589S) and two frameshift mutations (nt 460 del C, nt 693 del C) of the retinal guanylate cyclase (RETGC, GDB symbol GUC2D) gene in four unrelated LCA1 probands of North African ancestry and ascribe LCA1 to an impaired production of cGMP in the retina, with permanent closure of cGMP-gated cation channels.


Subject(s)
Blindness/congenital , Guanylate Cyclase/genetics , Mutation , Optic Atrophies, Hereditary/enzymology , Retina/enzymology , Blindness/enzymology , Blindness/genetics , Chromosomes, Human, Pair 17 , Cyclic GMP/metabolism , Frameshift Mutation , Homozygote , Humans , Molecular Sequence Data , Optic Atrophies, Hereditary/genetics , Photoreceptor Cells/metabolism , Restriction Mapping
16.
Clin Genet ; 79(3): 225-35, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20950397

ABSTRACT

Since the first reports of polyglutamine-binding protein 1 (PQBP1) mutations in Renpenning syndrome and related disorders, the spectrum of PQBP1-linked clinical manifestations has been outlined from rare published case reports. The phenotypic description is often obtained from medical archives, and therefore, heterogeneous. Moreover, some aspects such as brain imaging or cognitive and behavioral functioning are rarely described. In this study, 13 PQBP1-mutated French patients were subjected to a standardized clinical, cognitive and behavioral assessment. Physical measurements of their relatives were also collected. We report on a recognizable clinical and radiological phenotype. All patients presented with microcephaly, leanness and mild short stature, relative to familial measurements. Three new clinical features are described: upper back progressive muscular atrophy, metacarpophalangeal ankylosis of the thumb and velar dysfunction. The specific facial dysmorphic features included at least four of the following signs: long triangular face, large ridged nose, half-depilated eyebrows, dysplastic or protruding ears and rough slightly sparse hair. An over-aged appearance was noticed in elderly patients. Cortical gyrification was normal based on available magnetic brain imaging of six patients. PQBP1-linked microcephaly (or Renpenning syndrome) is an X-linked mental retardation syndrome, which has clinically recognizable features.


Subject(s)
Carrier Proteins/genetics , Mutation , Nuclear Proteins/genetics , Phenotype , Adolescent , Adult , Brain/diagnostic imaging , Brain/pathology , Cerebral Palsy/complications , Cerebral Palsy/diagnostic imaging , Cerebral Palsy/genetics , Cerebral Palsy/pathology , Child , Child, Preschool , Cognition Disorders/etiology , DNA-Binding Proteins , Female , France , Genotype , Humans , Male , Mental Retardation, X-Linked/complications , Mental Retardation, X-Linked/diagnostic imaging , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Pregnancy , Radiography , Young Adult
17.
Clin Genet ; 80(2): 177-83, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20825432

ABSTRACT

Raine syndrome is an autosomal recessive disorder caused by mutations in the FAM20C gene. FAM20C codes for the human homolog of DMP4, a dentin matrix protein highly expressed in odontoblasts and moderately in bone. DMP4 is probably playing a role in the mineralization process. Since the first case reported in 1989 by Raine et al. 21 cases have been published delineating a phenotype which associates dysmorphic features, cerebral calcifications, choanal atresia or stenosis and thoracic/pulmonary hypoplasia. Kan and Kozlowski suggested the name of Raine syndrome to describe this new lethal osteosclerotic bone dysplasia. All the cases described were lethal during the neonatal period except for the last two reported patients aged 8 and 11 years who presented severe mental retardation. Here we describe two sisters, with an attenuated phenotype of Raine syndrome, who present an unexpectedly normal psychomotor development at ages 4 and 1, respectively. Identification of a homozygous mutation in the FAM20C gene confirmed the Raine syndrome diagnosis, thus contributing to the expansion of the Raine syndrome phenotype. This case report also prompted us to revisit the FAM20 gene classification and allowed us to highlight the ancestral status of Fam20C.


Subject(s)
Abnormalities, Multiple/genetics , Cleft Palate/genetics , Exophthalmos/genetics , Extracellular Matrix Proteins/genetics , Microcephaly/genetics , Mutation , Osteosclerosis/genetics , Abnormalities, Multiple/diagnosis , Amino Acid Sequence , Base Sequence , Bone and Bones/pathology , Casein Kinase I , Child , Child, Preschool , Choanal Atresia/genetics , Choanal Atresia/metabolism , Cleft Palate/diagnosis , Exophthalmos/diagnosis , Female , Humans , Male , Microcephaly/diagnosis , Molecular Sequence Data , Osteosclerosis/diagnosis , Phenotype
18.
Clin Genet ; 80(6): 523-31, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21517826

ABSTRACT

The Bardet-Biedl syndrome (BBS) is a rare ciliopathy clinically defined by the association of retinitis pigmentosa, polydactyly, obesity, kidney disease and cognitive impairment. The cognitive functioning, behavioral phenotype, prevalence of psychiatric diseases and memory performances of a cohort of 34 patients with BBS were evaluated and a systemic brain magnetic resonance imaging (MRI) was performed. The patients' cognitive functioning was of marked variable efficiency ranging from normal to disabling performances. Neuropsychological disorders such as slow thought process, attention difficulties and obsessive-compulsive traits were observed. Our main finding was hippocampal dysgenesis, diagnosed by MRI, found in 42.31% of the patients in this cohort. Moreover, we show that BBS proteins are expressed in the human hippocampus and in the human brain in the normal subject. Recent literature in the murine model shows that hippocampal neurogenesis, in particular in the adult mouse, requires an intact primary cilia. These results encourage us to further investigate the possible role of BBS proteins in the hippocampus and related central nervous system structures.


Subject(s)
Bardet-Biedl Syndrome/pathology , Cilia/pathology , Hippocampus/pathology , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Adolescent , Adult , Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/metabolism , Chaperonins , Cilia/genetics , Cognition Disorders/diagnosis , Cognition Disorders/pathology , Cohort Studies , Female , Gene Expression , Group II Chaperonins/genetics , Group II Chaperonins/metabolism , Hippocampus/metabolism , Humans , Magnetic Resonance Imaging , Male , Memory , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Middle Aged , Neurogenesis , Phenotype , RNA, Messenger/analysis , RNA, Messenger/genetics , Young Adult
19.
J Med Genet ; 47(6): 377-84, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20522426

ABSTRACT

BACKGROUND Genome-wide screening of large patient cohorts with mental retardation using microarray-based comparative genomic hybridisation (array-CGH) has recently led to identification several novel microdeletion and microduplication syndromes. METHODS Owing to the national array-CGH network funded by the French Ministry of Health, shared information about patients with rare disease helped to define critical intervals and evaluate their gene content, and finally determine the phenotypic consequences of genomic array findings. RESULTS In this study, nine unrelated patients with overlapping de novo interstitial microdeletions involving 4q21 are reported. Several major features are common to all patients, including neonatal muscular hypotonia, severe psychomotor retardation, marked progressive growth restriction, distinctive facial features and absent or severely delayed speech. The boundaries and the sizes of the nine deletions are different, but an overlapping region of 1.37 Mb is defined; this region contains five RefSeq genes: PRKG2, RASGEF1B, HNRNPD, HNRPDL and ENOPH1. DISCUSSION Adding new individuals with similar clinical features and 4q21 deletion allowed us to reduce the critical genomic region encompassing two genes, PRKG2 and RASGEF1B. PRKG2 encodes cGMP-dependent protein kinase type II, which is expressed in brain and in cartilage. Information from genetically modified animal models is pertinent to the clinical phenotype. RASGEF1B is a guanine nucleotide exchange factor for Ras family proteins, and several members have been reported as key regulators of actin and microtubule dynamics during both dendrite and spine structural plasticity. CONCLUSION Clinical and molecular delineation of 4q21 deletion supports a novel microdeletion syndrome and suggests a major contribution of PRKG2 and RASGEF1B haploinsufficiency to the core phenotype.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Deletion , Chromosomes, Human, Pair 4/genetics , Growth Disorders/pathology , Intellectual Disability/pathology , Language Development Disorders/pathology , Abnormalities, Multiple/pathology , Adolescent , Child , Child, Preschool , Chromosome Disorders/genetics , Chromosome Disorders/pathology , Comparative Genomic Hybridization , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Syndrome , Young Adult
20.
Hum Mutat ; 31(2): 113-26, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19894250

ABSTRACT

Cockayne syndrome is an autosomal recessive multisystem disorder characterized principally by neurological and sensory impairment, cachectic dwarfism, and photosensitivity. This rare disease is linked to mutations in the CSB/ERCC6 and CSA/ERCC8 genes encoding proteins involved in the transcription-coupled DNA repair pathway. The clinical spectrum of Cockayne syndrome encompasses a wide range of severity from severe prenatal forms to mild and late-onset presentations. We have reviewed the 45 published mutations in CSA and CSB to date and we report 43 new mutations in these genes together with the corresponding clinical data. Among the 84 reported kindreds, 52 (62%) have mutations in the CSB gene. Many types of mutations are scattered along the whole coding sequence of both genes, but clusters of missense mutations can be recognized and highlight the role of particular motifs in the proteins. Genotype-phenotype correlation hypotheses are considered with regard to these new molecular and clinical data. Additional cases of molecular prenatal diagnosis are reported and the strategy for prenatal testing is discussed. Two web-based locus-specific databases have been created to list all identified variants and to allow the inclusion of future reports (www.umd.be/CSA/ and www.umd.be/CSB/).


Subject(s)
Cockayne Syndrome/genetics , DNA Helicases/genetics , DNA Repair Enzymes/genetics , Mutation/genetics , Transcription Factors/genetics , Amino Acid Sequence , Cockayne Syndrome/diagnosis , DNA Helicases/chemistry , DNA Repair Enzymes/chemistry , Databases, Genetic , Genetic Association Studies , Humans , Molecular Sequence Data , Poly-ADP-Ribose Binding Proteins , Polymorphism, Genetic , Sequence Alignment , Structure-Activity Relationship , Transcription Factors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL