Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nucleic Acids Res ; 51(W1): W39-W45, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37216590

ABSTRACT

Much of the human genetics variant repertoire is composed of single nucleotide variants (SNV) and small insertion/deletions (indel) but structural variants (SV) remain a major part of our modified DNA. SV detection has often been a complex question to answer either because of the necessity to use different technologies (array CGH, SNP array, Karyotype, Optical Genome Mapping…) to detect each category of SV or to get an appropriate resolution (Whole Genome Sequencing). Thanks to the deluge of pangenomic analysis, Human geneticists are accumulating SV and their interpretation remains time consuming and challenging. The AnnotSV webserver (https://www.lbgi.fr/AnnotSV/) aims at being an efficient tool to (i) annotate and interpret SV potential pathogenicity in the context of human diseases, (ii) recognize potential false positive variants from all the SV identified and (iii) visualize the patient variants repertoire. The most recent developments in the AnnotSV webserver are: (i) updated annotations sources and ranking, (ii) three novel output formats to allow diverse utilization (analysis, pipelines), as well as (iii) two novel user interfaces including an interactive circos view.


Subject(s)
INDEL Mutation , Polymorphism, Single Nucleotide , Software , Humans , Genome, Human , High-Throughput Nucleotide Sequencing , Restriction Mapping , Sequence Analysis, DNA , Whole Genome Sequencing , Disease/genetics
2.
Nucleic Acids Res ; 49(W1): W21-W28, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34023905

ABSTRACT

With the dramatic increase of pangenomic analysis, Human geneticists have generated large amount of genomic data including millions of small variants (SNV/indel) but also thousands of structural variations (SV) mainly from next-generation sequencing and array-based techniques. While the identification of the complete SV repertoire of a patient is getting possible, the interpretation of each SV remains challenging. To help identifying human pathogenic SV, we have developed a web server dedicated to their annotation and ranking (AnnotSV) as well as their visualization and interpretation (knotAnnotSV) freely available at the following address: https://www.lbgi.fr/AnnotSV/. A large amount of annotations from >20 sources is integrated in our web server including among others genes, haploinsufficiency, triplosensitivity, regulatory elements, known pathogenic or benign genomic regions, phenotypic data. An ACMG/ClinGen compliant prioritization module allows the scoring and the ranking of SV into 5 SV classes from pathogenic to benign. Finally, the visualization interface displays the annotated SV in an interactive way including popups, search fields, filtering options, advanced colouring to highlight pathogenic SV and hyperlinks to the UCSC genome browser or other public databases. This web server is designed for diagnostic and research analysis by providing important resources to the user.


Subject(s)
Genomic Structural Variation , Software , Genome, Human , Genomics , Humans , Internet , Molecular Sequence Annotation , Phenotype , Polymorphism, Single Nucleotide
3.
Int J Mol Sci ; 24(10)2023 May 13.
Article in English | MEDLINE | ID: mdl-37240074

ABSTRACT

Bardet-Biedl syndrome (BBS) is an autosomal recessive ciliopathy that affects multiple organs, leading to retinitis pigmentosa, polydactyly, obesity, renal anomalies, cognitive impairment, and hypogonadism. Until now, biallelic pathogenic variants have been identified in at least 24 genes delineating the genetic heterogeneity of BBS. Among those, BBS5 is a minor contributor to the mutation load and is one of the eight subunits forming the BBSome, a protein complex implied in protein trafficking within the cilia. This study reports on a European BBS5 patient with a severe BBS phenotype. Genetic analysis was performed using multiple next-generation sequencing (NGS) tests (targeted exome, TES and whole exome, WES), and biallelic pathogenic variants could only be identified using whole-genome sequencing (WGS), including a previously missed large deletion of the first exons. Despite the absence of family samples, the biallelic status of the variants was confirmed. The BBS5 protein's impact was confirmed on the patient's cells (presence/absence and size of the cilium) and ciliary function (Sonic Hedgehog pathway). This study highlights the importance of WGS and the challenge of reliable structural variant detection in patients' genetic explorations as well as functional tests to assess a variant's pathogenicity.


Subject(s)
Bardet-Biedl Syndrome , Polydactyly , Humans , Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/pathology , Cytoskeletal Proteins/genetics , Hedgehog Proteins/genetics , Mutation , Phenotype , Phosphate-Binding Proteins/genetics , Protein Transport , Male , Child, Preschool
4.
Am J Hum Genet ; 105(3): 509-525, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31422817

ABSTRACT

The human RNA helicase DDX6 is an essential component of membrane-less organelles called processing bodies (PBs). PBs are involved in mRNA metabolic processes including translational repression via coordinated storage of mRNAs. Previous studies in human cell lines have implicated altered DDX6 in molecular and cellular dysfunction, but clinical consequences and pathogenesis in humans have yet to be described. Here, we report the identification of five rare de novo missense variants in DDX6 in probands presenting with intellectual disability, developmental delay, and similar dysmorphic features including telecanthus, epicanthus, arched eyebrows, and low-set ears. All five missense variants (p.His372Arg, p.Arg373Gln, p.Cys390Arg, p.Thr391Ile, and p.Thr391Pro) are located in two conserved motifs of the RecA-2 domain of DDX6 involved in RNA binding, helicase activity, and protein-partner binding. We use functional studies to demonstrate that the first variants identified (p.Arg373Gln and p.Cys390Arg) cause significant defects in PB assembly in primary fibroblast and model human cell lines. These variants' interactions with several protein partners were also disrupted in immunoprecipitation assays. Further investigation via complementation assays included the additional variants p.Thr391Ile and p.Thr391Pro, both of which, similarly to p.Arg373Gln and p.Cys390Arg, demonstrated significant defects in P-body assembly. Complementing these molecular findings, modeling of the variants on solved protein structures showed distinct spatial clustering near known protein binding regions. Collectively, our clinical and molecular data describe a neurodevelopmental syndrome associated with pathogenic missense variants in DDX6. Additionally, we suggest DDX6 join the DExD/H-box genes DDX3X and DHX30 in an emerging class of neurodevelopmental disorders involving RNA helicases.


Subject(s)
DEAD-box RNA Helicases/genetics , Intellectual Disability/genetics , Mutation, Missense , Proto-Oncogene Proteins/genetics , RNA/genetics , Humans
5.
Clin Genet ; 100(2): 206-212, 2021 08.
Article in English | MEDLINE | ID: mdl-33890303

ABSTRACT

Periodontal Ehlers-Danlos syndrome (pEDS) is a rare condition caused by pathogenic variants in the C1R and C1S genes, encoding subunits C1r and C1s of the first component of the classical complement pathway. It is characterized by early-onset periodontitis with premature tooth loss, pretibial hyperpigmentation and skin fragility. Rare arterial complications have been reported, but venous insufficiency is rarely described. Here we report 13 novel patients carrying heterozygous pathogenic variants in C1R and C1S including three novel C1S variants (c.962G > C, c.961 T > G and c.961 T > A). In addition to the pEDS phenotype, three patients and one relative displayed widespread venous insufficiency leading to persistent varicose leg ulcers. One patient suffered an intracranial aneurysm with familial vascular complications including thoracic and abdominal aortic aneurysm and dissection and intracranial aneurysm rupture. This work confirms that vascular complications can occur, although they are not frequent, which leads us to propose to carry out a first complete non-invasive vascular evaluation at the time of the diagnosis in pEDS patients. However, larger case series are needed to improve our understanding of the link between complement pathway activation and connective tissue alterations observed in these patients, and to better assess the frequency, type and consequences of the vascular complications.


Subject(s)
Ehlers-Danlos Syndrome/etiology , Mutation , Adolescent , Adult , Aged , Aortic Aneurysm, Abdominal/genetics , Child, Preschool , Complement C1r/genetics , Complement C1s/genetics , Ehlers-Danlos Syndrome/genetics , Female , Heterozygote , Humans , Male , Middle Aged , Varicose Ulcer/etiology , Varicose Ulcer/genetics , Young Adult
6.
Clin Genet ; 99(2): 318-324, 2021 02.
Article in English | MEDLINE | ID: mdl-33169370

ABSTRACT

Bardet-Biedl syndrome (BBS) is a ciliopathy characterized by retinitis pigmentosa, obesity, polydactyly, cognitive impairment and renal failure. Pathogenic variants in 24 genes account for the molecular basis of >80% of cases. Toward saturated discovery of the mutational basis of the disorder, we carefully explored our cohorts and identified a hominid-specific SINE-R/VNTR/Alu type F (SVA-F) insertion in exon 13 of BBS1 in eight families. In six families, the repeat insertion was found in trans with c.1169 T > G, p.Met390Arg and in two families the insertion was found in addition to other recessive BBS loci. Whole genome sequencing, de novo assembly and SNP array analysis were performed to characterize the genomic event. This insertion is extremely rare in the general population (found in 8 alleles of 8 BBS cases but not in >10 800 control individuals from gnomAD-SV) and due to a founder effect. Its 2435 bp sequence contains hallmarks of LINE1 mediated retrotransposition. Functional studies with patient-derived cell lines confirmed that the BBS1 SVA-F is deleterious as evidenced by a significant depletion of both mRNA and protein levels. Such findings highlight the importance of dedicated bioinformatics pipelines to identify all types of variation.


Subject(s)
Bardet-Biedl Syndrome/genetics , Microtubule-Associated Proteins/genetics , Retroelements , Cohort Studies , Female , Founder Effect , Gene Frequency , Humans , Male , Mutagenesis, Insertional , Pedigree , Whole Genome Sequencing
7.
Hum Mutat ; 41(1): 240-254, 2020 01.
Article in English | MEDLINE | ID: mdl-31549751

ABSTRACT

Polydactyly is one of the most frequent inherited defects of the limbs characterized by supernumerary digits and high-genetic heterogeneity. Among the many genes involved, either in isolated or syndromic forms, eight have been implicated in postaxial polydactyly (PAP). Among those, IQCE has been recently identified in a single consanguineous family. Using whole-exome sequencing in patients with uncharacterized ciliopathies, including PAP, we identified three families with biallelic pathogenic variations in IQCE. Interestingly, the c.895_904del (p.Val301Serfs*8) was found in all families without sharing a common haplotype, suggesting a recurrent mechanism. Moreover, in two families, the systemic phenotype could be explained by additional pathogenic variants in known genes (TULP1, ATP6V1B1). RNA expression analysis on patients' fibroblasts confirms that the dysfunction of IQCE leads to the dysregulation of genes associated with the hedgehog-signaling pathway, and zebrafish experiments demonstrate a full spectrum of phenotypes linked to defective cilia: Body curvature, kidney cysts, left-right asymmetry, misdirected cilia in the pronephric duct, and retinal defects. In conclusion, we identified three additional families confirming IQCE as a nonsyndromic PAP gene. Our data emphasize the importance of taking into account the complete set of variations of each individual, as each clinical presentation could finally be explained by multiple genes.


Subject(s)
Ciliopathies/diagnosis , Ciliopathies/genetics , Fingers/abnormalities , Genetic Predisposition to Disease , Genetic Variation , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Phenotype , Polydactyly/diagnosis , Polydactyly/genetics , Toes/abnormalities , Animals , Consanguinity , Fluorescent Antibody Technique , Gene Expression Profiling , Genetic Association Studies/methods , Homozygote , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Pedigree , Signal Transduction , Transcriptome , Exome Sequencing , Zebrafish
8.
Hum Mutat ; 40(10): 1826-1840, 2019 10.
Article in English | MEDLINE | ID: mdl-31116475

ABSTRACT

Mutations in genes encoding aminoacyl-tRNA synthetases have been reported in several neurological disorders. KARS is a dual localized lysyl-tRNA synthetase and its cytosolic isoform belongs to the multiple aminoacyl-tRNA synthetase complex (MSC). Biallelic mutations in the KARS gene were described in a wide phenotypic spectrum ranging from nonsyndromic deafness to complex impairments. Here, we report on a patient with severe neurological and neurosensory disease investigated by whole-exome sequencing and found to carry biallelic mutations c.683C>T (p.Pro228Leu) and c.871T>G (p.Phe291Val), the second one being novel, in the KARS gene. The patient presented with an atypical clinical presentation with an optic neuropathy not previously reported. At the cellular level, we show that cytoplasmic KARS was expressed at a lower level in patient cells and displayed decreased interaction with MSC. In vitro, these two KARS variants have a decreased aminoacylation activity compared with wild-type KARS, the p.Pro228Leu being the most affected. Our data suggest that dysfunction of cytoplasmic KARS resulted in a decreased level of translation of the nuclear-encoded lysine-rich proteins belonging to the respiratory chain complex, thus impairing mitochondria functions.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Lysine-tRNA Ligase/genetics , Mutation , Nervous System Diseases/complications , Nervous System Diseases/genetics , Optic Nerve Diseases/complications , Sensation Disorders/complications , Sensation Disorders/genetics , Alleles , Amino Acid Sequence , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/metabolism , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex IV/metabolism , Fibroblasts/metabolism , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Lysine-tRNA Ligase/chemistry , Lysine-tRNA Ligase/metabolism , Magnetic Resonance Imaging , Models, Molecular , Nervous System Diseases/diagnosis , Optic Nerve Diseases/diagnosis , Pedigree , Protein Binding , Protein Conformation , Sensation Disorders/diagnosis , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Bioinformatics ; 34(20): 3572-3574, 2018 10 15.
Article in English | MEDLINE | ID: mdl-29669011

ABSTRACT

Summary: Structural Variations (SV) are a major source of variability in the human genome that shaped its actual structure during evolution. Moreover, many human diseases are caused by SV, highlighting the need to accurately detect those genomic events but also to annotate them and assist their biological interpretation. Therefore, we developed AnnotSV that compiles functionally, regulatory and clinically relevant information and aims at providing annotations useful to (i) interpret SV potential pathogenicity and (ii) filter out SV potential false positive. In particular, AnnotSV reports heterozygous and homozygous counts of single nucleotide variations (SNVs) and small insertions/deletions called within each SV for the analyzed patients, this genomic information being extremely useful to support or question the existence of an SV. We also report the computed allelic frequency relative to overlapping variants from DGV (MacDonald et al., 2014), that is especially powerful to filter out common SV. To delineate the strength of AnnotSV, we annotated the 4751 SV from one sample of the 1000 Genomes Project, integrating the sample information of four million of SNV/indel, in less than 60 s. Availability and implementation: AnnotSV is implemented in Tcl and runs in command line on all platforms. The source code is available under the GNU GPL license. Source code, README and Supplementary data are available at http://lbgi.fr/AnnotSV/. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Software , Gene Frequency , Genome, Human , Genomics , Humans , Molecular Sequence Annotation
10.
Hum Mutat ; 39(7): 983-992, 2018 07.
Article in English | MEDLINE | ID: mdl-29688594

ABSTRACT

Ciliopathies represent a wide spectrum of rare diseases with overlapping phenotypes and a high genetic heterogeneity. Among those, IFT140 is implicated in a variety of phenotypes ranging from isolated retinis pigmentosa to more syndromic cases. Using whole-genome sequencing in patients with uncharacterized ciliopathies, we identified a novel recurrent tandem duplication of exon 27-30 (6.7 kb) in IFT140, c.3454-488_4182+2588dup p.(Tyr1152_Thr1394dup), missed by whole-exome sequencing. Pathogenicity of the mutation was assessed on the patients' skin fibroblasts. Several hundreds of patients with a ciliopathy phenotype were screened and biallelic mutations were identified in 11 families representing 12 pathogenic variants of which seven are novel. Among those unrelated families especially with a Mainzer-Saldino syndrome, eight carried the same tandem duplication (two at the homozygous state and six at the heterozygous state). In conclusion, we demonstrated the implication of structural variations in IFT140-related diseases expanding its mutation spectrum. We also provide evidences for a unique genomic event mediated by an Alu-Alu recombination occurring on a shared haplotype. We confirm that whole-genome sequencing can be instrumental in the ability to detect structural variants for genomic disorders.


Subject(s)
Carrier Proteins/genetics , Cerebellar Ataxia/genetics , Ciliopathies/genetics , Retinitis Pigmentosa/genetics , Whole Genome Sequencing , Alu Elements/genetics , Cerebellar Ataxia/pathology , Ciliopathies/pathology , Databases, Genetic , Exons/genetics , Female , Heterozygote , Homozygote , Humans , Male , Mutation/genetics , Pedigree , Phenotype , Retinitis Pigmentosa/pathology
11.
Am J Hum Genet ; 96(4): 666-74, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25817018

ABSTRACT

We have identified TUBGCP4 variants in individuals with autosomal-recessive microcephaly and chorioretinopathy. Whole-exome sequencing performed on one family with two affected siblings and independently on another family with one affected child revealed compound-heterozygous mutations in TUBGCP4. Subsequent Sanger sequencing was performed on a panel of individuals from 12 French families affected by microcephaly and ophthalmic manifestations, and one other individual was identified with compound-heterozygous mutations in TUBGCP4. One synonymous variant was common to all three families and was shown to induce exon skipping; the other mutations were frameshift mutations and a deletion. TUBGCP4 encodes γ-tubulin complex protein 4, a component belonging to the γ-tubulin ring complex (γ-TuRC) and known to regulate the nucleation and organization of microtubules. Functional analysis of individual fibroblasts disclosed reduced levels of the γ-TuRC, altered nucleation and organization of microtubules, abnormal nuclear shape, and aneuploidy. Moreover, zebrafish treated with morpholinos against tubgcp4 were found to have reduced head volume and eye developmental anomalies with chorioretinal dysplasia. In summary, the identification of TUBGCP4 mutations in individuals with microcephaly and a spectrum of anomalies in eye development, particularly photoreceptor anomalies, provides evidence of an important role for the γ-TuRC in brain and eye development.


Subject(s)
Choroid Diseases/genetics , Eye Diseases, Hereditary/genetics , Microcephaly/genetics , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Retinal Diseases/genetics , Tubulin/metabolism , Base Sequence , Exome/genetics , Frameshift Mutation/genetics , France , Gene Components , Humans , Microtubules/metabolism , Molecular Sequence Data , Pedigree , Sequence Analysis, DNA
12.
Hum Mol Genet ; 24(11): 3038-49, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25669657

ABSTRACT

Inherited dental malformations constitute a clinically and genetically heterogeneous group of disorders. Here, we report on four families, three of them consanguineous, with an identical phenotype, characterized by significant short stature with brachyolmia and hypoplastic amelogenesis imperfecta (AI) with almost absent enamel. This phenotype was first described in 1996 by Verloes et al. as an autosomal recessive form of brachyolmia associated with AI. Whole-exome sequencing resulted in the identification of recessive hypomorphic mutations including deletion, nonsense and splice mutations, in the LTBP3 gene, which is involved in the TGF-beta signaling pathway. We further investigated gene expression during mouse development and tooth formation. Differentiated ameloblasts synthesizing enamel matrix proteins and odontoblasts expressed the gene. Study of an available knockout mouse model showed that the mutant mice displayed very thin to absent enamel in both incisors and molars, hereby recapitulating the AI phenotype in the human disorder.


Subject(s)
Amelogenesis Imperfecta/genetics , Latent TGF-beta Binding Proteins/genetics , Osteochondrodysplasias/genetics , Adolescent , Amelogenesis Imperfecta/diagnostic imaging , Animals , Base Sequence , Child , Consanguinity , DNA Mutational Analysis , Female , Frameshift Mutation , Genetic Association Studies , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation, Missense , Osteochondrodysplasias/diagnostic imaging , Pedigree , Radiography , Sequence Deletion
13.
J Med Genet ; 53(2): 98-110, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26502894

ABSTRACT

BACKGROUND: Orodental diseases include several clinically and genetically heterogeneous disorders that can present in isolation or as part of a genetic syndrome. Due to the vast number of genes implicated in these disorders, establishing a molecular diagnosis can be challenging. We aimed to develop a targeted next-generation sequencing (NGS) assay to diagnose mutations and potentially identify novel genes mutated in this group of disorders. METHODS: We designed an NGS gene panel that targets 585 known and candidate genes in orodental disease. We screened a cohort of 101 unrelated patients without a molecular diagnosis referred to the Reference Centre for Oro-Dental Manifestations of Rare Diseases, Strasbourg, France, for a variety of orodental disorders including isolated and syndromic amelogenesis imperfecta (AI), isolated and syndromic selective tooth agenesis (STHAG), isolated and syndromic dentinogenesis imperfecta, isolated dentin dysplasia, otodental dysplasia and primary failure of tooth eruption. RESULTS: We discovered 21 novel pathogenic variants and identified the causative mutation in 39 unrelated patients in known genes (overall diagnostic rate: 39%). Among the largest subcohorts of patients with isolated AI (50 unrelated patients) and isolated STHAG (21 unrelated patients), we had a definitive diagnosis in 14 (27%) and 15 cases (71%), respectively. Surprisingly, COL17A1 mutations accounted for the majority of autosomal-dominant AI cases. CONCLUSIONS: We have developed a novel targeted NGS assay for the efficient molecular diagnosis of a wide variety of orodental diseases. Furthermore, our panel will contribute to better understanding the contribution of these genes to orodental disease. TRIAL REGISTRATION NUMBERS: NCT01746121 and NCT02397824.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Mutation , Tooth Abnormalities/genetics , Amelogenesis Imperfecta/genetics , Autoantigens/genetics , Chromosome Deletion , Chromosome Disorders/genetics , Chromosomes, Human, Pair 11/genetics , Cohort Studies , Coloboma/genetics , Dentin Dysplasia/genetics , France , Hearing Loss, Sensorineural/genetics , Humans , Non-Fibrillar Collagens/genetics , Reproducibility of Results , Collagen Type XVII
14.
J Hum Genet ; 61(5): 447-50, 2016 May.
Article in English | MEDLINE | ID: mdl-26763875

ABSTRACT

Bardet-Biedl syndrome (BBS; MIM 209900) is a recessive heterogeneous ciliopathy characterized by retinitis pigmentosa (RP), postaxial polydactyly, obesity, hypogonadism, cognitive impairment and kidney dysfunction. So far, 20 BBS genes have been identified, with the last reported ones being found in one or very few families. Whole-exome sequencing was performed in a consanguineous family in which two affected children presented typical BBS features (retinitis pigmentosa, postaxial polydactyly, obesity, hypogonadism and cognitive impairment) without any mutation identified in known BBS genes at the time of the study. We identified a homozygous splice-site mutation (NM_015662.2: c.4428+3A>G) in both affected siblings in the last reported BBS gene, namely, Intraflagellar Transport 172 Homolog (IFT172). Familial mutation segregation was consistent with autosomal recessive inheritance. IFT172 mutations were initially reported in Jeune and Mainzer-Saldino syndromes. Recently, mutations have also been found in isolated RP and Bardet-Biedl-like ciliopathy. This is the second report of IFT172 mutations in BBS patients validating IFT172 as the twentieth BBS gene (BBS20). Moreover, another IFT gene, IFT27, was already associated with BBS, confirming the implication of IFT genes in the pathogenesis of BBS.


Subject(s)
Bardet-Biedl Syndrome/diagnosis , Bardet-Biedl Syndrome/genetics , Carrier Proteins/genetics , Mutation , Adaptor Proteins, Signal Transducing , Child , Child, Preschool , Computational Biology/methods , Cytoskeletal Proteins , Exome , High-Throughput Nucleotide Sequencing , Homozygote , Humans , Male , Pedigree , Phenotype , Skeleton/diagnostic imaging , Skeleton/pathology
15.
J Med Genet ; 51(2): 132-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24026985

ABSTRACT

BACKGROUND: Bardet-Biedl syndrome (BBS) is a recessive and genetically heterogeneous ciliopathy characterised by retinitis pigmentosa, obesity, kidney dysfunction, postaxial polydactyly, behavioural dysfunction and hypogonadism. 7 of the 17 BBS gene products identified to date assemble together with the protein BBIP1/BBIP10 into the BBSome, a protein complex that ferries signalling receptors to and from cilia. METHODS AND RESULTS: Exome sequencing performed on a sporadic BBS case revealed for the first time a homozygous stop mutation (NM_001195306: c.173T>G, p.Leu58*) in the BBIP1 gene. This mutation is pathogenic since no BBIP1 protein could be detected in fibroblasts from the patient, and BBIP1[Leu58*] is unable to associate with the BBSome subunit BBS4. CONCLUSIONS: These findings identify BBIP1 as the 18th BBS gene (BBS18) and suggest that BBSome assembly may represent a unifying pathomechanism for BBS.


Subject(s)
Bardet-Biedl Syndrome/genetics , Carrier Proteins/genetics , Codon, Nonsense , Exome , Animals , Bardet-Biedl Syndrome/metabolism , Base Sequence , Consanguinity , DNA Mutational Analysis , Fibroblasts/metabolism , Genetic Association Studies , Genetic Linkage , HEK293 Cells , Humans , Male , Middle Aged , Molecular Sequence Annotation , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Zebrafish
16.
J Med Genet ; 51(11): 724-36, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25167861

ABSTRACT

BACKGROUND: Intellectual disability (ID) is characterised by an extreme genetic heterogeneity. Several hundred genes have been associated to monogenic forms of ID, considerably complicating molecular diagnostics. Trio-exome sequencing was recently proposed as a diagnostic approach, yet remains costly for a general implementation. METHODS: We report the alternative strategy of targeted high-throughput sequencing of 217 genes in which mutations had been reported in patients with ID or autism as the major clinical concern. We analysed 106 patients with ID of unknown aetiology following array-CGH analysis and other genetic investigations. Ninety per cent of these patients were males, and 75% sporadic cases. RESULTS: We identified 26 causative mutations: 16 in X-linked genes (ATRX, CUL4B, DMD, FMR1, HCFC1, IL1RAPL1, IQSEC2, KDM5C, MAOA, MECP2, SLC9A6, SLC16A2, PHF8) and 10 de novo in autosomal-dominant genes (DYRK1A, GRIN1, MED13L, TCF4, RAI1, SHANK3, SLC2A1, SYNGAP1). We also detected four possibly causative mutations (eg, in NLGN3) requiring further investigations. We present detailed reasoning for assigning causality for each mutation, and associated patients' clinical information. Some genes were hit more than once in our cohort, suggesting they correspond to more frequent ID-associated conditions (KDM5C, MECP2, DYRK1A, TCF4). We highlight some unexpected genotype to phenotype correlations, with causative mutations being identified in genes associated to defined syndromes in patients deviating from the classic phenotype (DMD, TCF4, MECP2). We also bring additional supportive (HCFC1, MED13L) or unsupportive (SHROOM4, SRPX2) evidences for the implication of previous candidate genes or mutations in cognitive disorders. CONCLUSIONS: With a diagnostic yield of 25% targeted sequencing appears relevant as a first intention test for the diagnosis of ID, but importantly will also contribute to a better understanding regarding the specific contribution of the many genes implicated in ID and autism.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Molecular Diagnostic Techniques/methods , Adolescent , Adult , Child , Child, Preschool , DNA Mutational Analysis/methods , Female , Humans , Infant , Infant, Newborn , Male , Sequence Analysis, DNA/methods , Young Adult
17.
Am J Hum Genet ; 89(6): 773-81, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22152679

ABSTRACT

Inherited dental malformations constitute a clinically and genetically heterogeneous group of disorders. Here, we report on a severe developmental dental defect that results in a dentin dysplasia phenotype with major microdontia, oligodontia, and shape abnormalities in a highly consanguineous family. Homozygosity mapping revealed a unique zone on 6q27-ter. The two affected children were found to carry a homozygous mutation in SMOC2. Knockdown of smoc2 in zebrafish showed pharyngeal teeth that had abnormalities reminiscent of the human phenotype. Moreover, smoc2 depletion in zebrafish affected the expression of three major odontogenesis genes: dlx2, bmp2, and pitx2.


Subject(s)
Calcium-Binding Proteins/genetics , Chromosome Mapping , Dentin Dysplasia/genetics , Exome , Homozygote , Sequence Analysis, DNA , Tooth/growth & development , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Child , Child, Preschool , Chromosomes, Human, Pair 6 , Consanguinity , Dentin Dysplasia/diagnosis , Female , Gene Expression , Gene Expression Regulation, Developmental , Genetic Association Studies , Humans , Mice , Molecular Sequence Data , Neoplasm Proteins/genetics , Pedigree , Zebrafish/genetics , Zebrafish/growth & development
18.
J Med Genet ; 49(8): 502-12, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22773737

ABSTRACT

BACKGROUND: Bardet-Biedl syndrome (BBS) is a pleiotropic recessive disorder that belongs to the rapidly growing family of ciliopathies. It shares phenotypic traits with other ciliopathies, such as Alström syndrome (ALMS), nephronophthisis (NPHP) or Joubert syndrome. BBS mutations have been detected in 16 different genes (BBS1-BBS16) without clear genotype-to-phenotype correlation. This extensive genetic heterogeneity is a major concern for molecular diagnosis and genetic counselling. While various strategies have been recently proposed to optimise mutation detection, they either fail to detect mutations in a majority of patients or are time consuming and costly. METHOD: We tested a targeted exon-capture strategy coupled with multiplexing and high-throughput sequencing on 52 patients: 14 with known mutations as proof-of-principle and 38 with no previously detected mutation. Thirty genes were targeted in total including the 16 BBS genes, the 12 known NPHP genes, the single ALMS gene ALMS1 and the proposed modifier CCDC28B. RESULTS: This strategy allowed the reliable detection of causative mutations (including homozygous/heterozygous exon deletions) in 68% of BBS patients without previous molecular diagnosis and in all proof-of-principle samples. Three probands carried homozygous truncating mutations in ALMS1 confirming the major phenotypic overlap between both disorders. The efficiency of detecting mutations in patients was positively correlated with their compliance with the classical BBS phenotype (mutations were identified in 81% of 'classical' BBS patients) suggesting that only a few true BBS genes remain to be identified. We illustrate some interpretation problems encountered due to the multiplicity of identified variants. CONCLUSION: This strategy is highly efficient and cost effective for diseases with high genetic heterogeneity, and guarantees a quality of coverage in coding sequences of target genes suited for diagnosis purposes.


Subject(s)
Alstrom Syndrome/diagnosis , Bardet-Biedl Syndrome/diagnosis , DNA Mutational Analysis/methods , High-Throughput Nucleotide Sequencing/methods , Sequence Deletion , Alstrom Syndrome/genetics , Bardet-Biedl Syndrome/genetics , Cell Cycle Proteins/genetics , Cohort Studies , Cytoskeletal Proteins , Exons , Genetic Association Studies , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Genetic Heterogeneity , Genetic Testing/methods , Genome, Human , Heterozygote , Homozygote , Humans , Proteins/genetics , Reproducibility of Results
19.
EMBO Mol Med ; 12(7): e11861, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32500975

ABSTRACT

The ubiquitin-proteasome system degrades ubiquitin-modified proteins to maintain protein homeostasis and to control signalling. Whole-genome sequencing of patients with severe deafness and early-onset cataracts as part of a neurological, sensorial and cutaneous novel syndrome identified a unique deep intronic homozygous variant in the PSMC3 gene, encoding the proteasome ATPase subunit Rpt5, which lead to the transcription of a cryptic exon. The proteasome content and activity in patient's fibroblasts was however unaffected. Nevertheless, patient's cells exhibited impaired protein homeostasis characterized by accumulation of ubiquitinated proteins suggesting severe proteotoxic stress. Indeed, the TCF11/Nrf1 transcriptional pathway allowing proteasome recovery after proteasome inhibition is permanently activated in the patient's fibroblasts. Upon chemical proteasome inhibition, this pathway was however impaired in patient's cells, which were unable to compensate for proteotoxic stress although a higher proteasome content and activity. Zebrafish modelling for knockout in PSMC3 remarkably reproduced the human phenotype with inner ear development anomalies as well as cataracts, suggesting that Rpt5 plays a major role in inner ear, lens and central nervous system development.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Cataract/genetics , Deafness/genetics , Mutation , Proteasome Endopeptidase Complex/genetics , Proteolysis , Stress, Physiological , Zebrafish Proteins/genetics , Adolescent , Animals , Cataract/pathology , Child , Child, Preschool , Consanguinity , Deafness/physiopathology , Female , Humans , Infant , Male , Nuclear Respiratory Factor 1/genetics , Pedigree , Phenotype , Proteasome Inhibitors/pharmacology , Proteolysis/drug effects , Stress, Physiological/drug effects , Stress, Physiological/genetics , Syndrome , Ubiquitin/metabolism , Zebrafish/genetics
20.
Front Genet ; 10: 21, 2019.
Article in English | MEDLINE | ID: mdl-30761183

ABSTRACT

Bardet-Biedl syndrome (BBS; MIM 209900) is a rare ciliopathy characterized by retinitis pigmentosa, postaxial polydactyly, obesity, hypogonadism, cognitive impairment and kidney dysfunction. Mutations in 22 BBS genes have been identified to cause the disease. We report a family with typical BBS features (retinitis pigmentosa, postaxial polydactyly, obesity, cognitive impairment, and atrioventricular septal defect) mutated in IFT27/BBS19. IFT27 is part of the Intraflagellar transport (IFT), a bidirectional mechanism allowing the protein motility within the cilia. Using whole exome sequencing, two compound heterozygous mutations were found in the proband (NM_006860.4:c.[104A > G];[349+1G > T], p.[Tyr35Cys];[?]) consistent with the expected autosomal recessive inheritance mode. These two mutations have already been reported but independently in other families and lacking either familial segregation or functional validation. This is the third report of IFT27 mutations in BBS patients confirming IFT27 as a BBS gene (BBS19). Mutations in IFT genes (IFT27, IFT172 and IFT74) confirm the IFT-pathway as a pathomechanism for BBS.

SELECTION OF CITATIONS
SEARCH DETAIL