Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Am J Hum Genet ; 110(2): 215-227, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36586412

ABSTRACT

Neurodevelopmental disorders (NDDs) result from highly penetrant variation in hundreds of different genes, some of which have not yet been identified. Using the MatchMaker Exchange, we assembled a cohort of 27 individuals with rare, protein-altering variation in the transcriptional coregulator ZMYM3, located on the X chromosome. Most (n = 24) individuals were males, 17 of which have a maternally inherited variant; six individuals (4 male, 2 female) harbor de novo variants. Overlapping features included developmental delay, intellectual disability, behavioral abnormalities, and a specific facial gestalt in a subset of males. Variants in almost all individuals (n = 26) are missense, including six that recurrently affect two residues. Four unrelated probands were identified with inherited variation affecting Arg441, a site at which variation has been previously seen in NDD-affected siblings, and two individuals have de novo variation resulting in p.Arg1294Cys (c.3880C>T). All variants affect evolutionarily conserved sites, and most are predicted to damage protein structure or function. ZMYM3 is relatively intolerant to variation in the general population, is widely expressed across human tissues, and encodes a component of the KDM1A-RCOR1 chromatin-modifying complex. ChIP-seq experiments on one variant, p.Arg1274Trp, indicate dramatically reduced genomic occupancy, supporting a hypomorphic effect. While we are unable to perform statistical evaluations to definitively support a causative role for variation in ZMYM3, the totality of the evidence, including 27 affected individuals, recurrent variation at two codons, overlapping phenotypic features, protein-modeling data, evolutionary constraint, and experimentally confirmed functional effects strongly support ZMYM3 as an NDD-associated gene.


Subject(s)
Intellectual Disability , Nervous System Malformations , Neurodevelopmental Disorders , Humans , Male , Female , Neurodevelopmental Disorders/genetics , Intellectual Disability/genetics , Phenotype , Gene Expression Regulation , Face , Nuclear Proteins/genetics , Histone Demethylases/genetics
2.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928247

ABSTRACT

The purpose of this study was to assess the added diagnostic value of whole genome sequencing (WGS) for patients with inherited retinal diseases (IRDs) who remained undiagnosed after whole exome sequencing (WES). WGS was performed for index patients in 66 families. The datasets were analyzed according to GATK's guidelines. Additionally, DeepVariant was complemented by GATK's workflow, and a novel structural variant pipeline was developed. Overall, a molecular diagnosis was established in 19/66 (28.8%) index patients. Pathogenic deletions and one deep-intronic variant contributed to the diagnostic yield in 4/19 and 1/19 index patients, respectively. The remaining diagnoses (14/19) were attributed to exonic variants that were missed during WES analysis due to bioinformatic limitations, newly described loci, or unclear pathogenicity. The added diagnostic value of WGS equals 5/66 (9.6%) for our cohort, which is comparable to previous studies. This figure would decrease further to 1/66 (1.5%) with a standardized and reliable copy number variant workflow during WES analysis. Given the higher costs and limited added value, the implementation of WGS as a first-tier assay for inherited eye disorders in a diagnostic laboratory remains untimely. Instead, progress in bioinformatic tools and communication between diagnostic and clinical teams have the potential to ameliorate diagnostic yields.


Subject(s)
Genetic Testing , Retinal Diseases , Whole Genome Sequencing , Humans , Retinal Diseases/genetics , Retinal Diseases/diagnosis , Genetic Testing/methods , Whole Genome Sequencing/methods , Male , Female , Switzerland , Cohort Studies , Adult , DNA Copy Number Variations , Exome Sequencing/methods , Computational Biology/methods , Middle Aged , Child , Adolescent , Pedigree
3.
J Cell Sci ; 134(14)2021 07 15.
Article in English | MEDLINE | ID: mdl-34155518

ABSTRACT

Mutations in CEP290 (also known as NPHP6), a large multidomain coiled coil protein, are associated with multiple cilia-associated syndromes. Over 130 CEP290 mutations have been linked to a wide spectrum of human ciliopathies, raising the question of how mutations in a single gene cause different disease syndromes. In zebrafish, the expressivity of cep290 deficiencies were linked to the type of genetic ablation: acute cep290 morpholino knockdown caused severe cilia-related phenotypes, whereas deficiencies in a CRISPR/Cas9 genetic mutant were restricted to photoreceptor defects. Here, we show that milder phenotypes in genetic mutants were associated with the upregulation of genes encoding the cilia-associated small GTPases arl3, arl13b and unc119b. Upregulation of UNC119b was also observed in urine-derived renal epithelial cells from human Joubert syndrome CEP290 patients. Ectopic expression of arl3, arl13b and unc119b in cep290 morphant zebrafish embryos rescued Kupffer's vesicle cilia and partially rescued photoreceptor outer segment defects. The results suggest that genetic compensation by upregulation of genes involved in a common subcellular process, lipidated protein trafficking to cilia, may be a conserved mechanism contributing to genotype-phenotype variations observed in CEP290 deficiencies. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Antigens, Neoplasm , Cell Cycle Proteins , Cilia , Cytoskeletal Proteins , Monomeric GTP-Binding Proteins , Adaptor Proteins, Signal Transducing , Animals , Antigens, Neoplasm/genetics , Cell Cycle Proteins/genetics , Cilia/genetics , Cilia/metabolism , Cytoskeletal Proteins/genetics , Humans , Microtubule-Associated Proteins , Mutation/genetics , Up-Regulation/genetics , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
4.
Hum Mol Genet ; 29(7): 1132-1143, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32129449

ABSTRACT

The molecular cause of the majority of rare autosomal recessive disorders remains unknown. Consanguinity due to extensive homozygosity unravels many recessive phenotypes and facilitates the detection of novel gene-disease links. Here, we report two siblings with phenotypic signs, including intellectual disability (ID), developmental delay and microcephaly from a Pakistani consanguineous family in which we have identified homozygosity for p(Tyr103His) in the PSMB1 gene (Genbank NM_002793) that segregated with the disease phenotype. PSMB1 encodes a ß-type proteasome subunit (i.e. ß6). Modeling of the p(Tyr103His) variant indicates that this variant weakens the interactions between PSMB1/ß6 and PSMA5/α5 proteasome subunits and thus destabilizes the 20S proteasome complex. Biochemical experiments in human SHSY5Y cells revealed that the p(Tyr103His) variant affects both the processing of PSMB1/ß6 and its incorporation into proteasome, thus impairing proteasome activity. CRISPR/Cas9 mutagenesis or morpholino knock-down of the single psmb1 zebrafish orthologue resulted in microcephaly, microphthalmia and reduced brain size. Genetic evidence in the family and functional experiments in human cells and zebrafish indicates that PSMB1/ß6 pathogenic variants are the cause of a recessive disease with ID, microcephaly and developmental delay due to abnormal proteasome assembly.


Subject(s)
Dwarfism/genetics , Microcephaly/genetics , Proteasome Endopeptidase Complex/genetics , Alleles , Animals , Child , Consanguinity , Developmental Disabilities/complications , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Dwarfism/complications , Female , Homozygote , Humans , Intellectual Disability/complications , Intellectual Disability/genetics , Intellectual Disability/pathology , Male , Microcephaly/complications , Microcephaly/pathology , Models, Molecular , Pedigree , Phenotype , Zebrafish/genetics
5.
Am J Hum Genet ; 104(4): 701-708, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30879638

ABSTRACT

Developmental delay and intellectual disability (DD and ID) are heterogeneous phenotypes that arise in many rare monogenic disorders. Because of this rarity, developing cohorts with enough individuals to robustly identify disease-associated genes is challenging. Social-media platforms that facilitate data sharing among sequencing labs can help to address this challenge. Through one such tool, GeneMatcher, we identified nine DD- and/or ID-affected probands with a rare, heterozygous variant in the gene encoding the serine/threonine-protein kinase BRSK2. All probands have a speech delay, and most present with intellectual disability, motor delay, behavioral issues, and autism. Six of the nine variants are predicted to result in loss of function, and computational modeling predicts that the remaining three missense variants are damaging to BRSK2 structure and function. All nine variants are absent from large variant databases, and BRSK2 is, in general, relatively intolerant to protein-altering variation among humans. In all six probands for whom parents were available, the mutations were found to have arisen de novo. Five of these de novo variants were from cohorts with at least 400 sequenced probands; collectively, the cohorts span 3,429 probands, and the observed rate of de novo variation in these cohorts is significantly higher than the estimated background-mutation rate (p = 2.46 × 10-6). We also find that exome sequencing provides lower coverage and appears less sensitive to rare variation in BRSK2 than does genome sequencing; this fact most likely reduces BRSK2's visibility in many clinical and research sequencing efforts. Altogether, our results implicate damaging variation in BRSK2 as a source of neurodevelopmental disease.


Subject(s)
Developmental Disabilities/genetics , Gene Deletion , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , Protein Serine-Threonine Kinases/genetics , Adolescent , Autistic Disorder/genetics , Child , Child Behavior Disorders/genetics , Child, Preschool , Exome , Female , Genetic Predisposition to Disease , Genetic Variation , Heterozygote , Humans , Male , Motor Skills Disorders/genetics , Mutation , Phenotype , Exome Sequencing , Young Adult
6.
Am J Hum Genet ; 101(1): 23-36, 2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28625504

ABSTRACT

Joubert syndrome (JS) is a recessive neurodevelopmental disorder characterized by hypotonia, ataxia, abnormal eye movements, and variable cognitive impairment. It is defined by a distinctive brain malformation known as the "molar tooth sign" on axial MRI. Subsets of affected individuals have malformations such as coloboma, polydactyly, and encephalocele, as well as progressive retinal dystrophy, fibrocystic kidney disease, and liver fibrosis. More than 35 genes have been associated with JS, but in a subset of families the genetic cause remains unknown. All of the gene products localize in and around the primary cilium, making JS a canonical ciliopathy. Ciliopathies are unified by their overlapping clinical features and underlying mechanisms involving ciliary dysfunction. In this work, we identify biallelic rare, predicted-deleterious ARMC9 variants (stop-gain, missense, splice-site, and single-exon deletion) in 11 individuals with JS from 8 families, accounting for approximately 1% of the disorder. The associated phenotypes range from isolated neurological involvement to JS with retinal dystrophy, additional brain abnormalities (e.g., heterotopia, Dandy-Walker malformation), pituitary insufficiency, and/or synpolydactyly. We show that ARMC9 localizes to the basal body of the cilium and is upregulated during ciliogenesis. Typical ciliopathy phenotypes (curved body shape, retinal dystrophy, coloboma, and decreased cilia) in a CRISPR/Cas9-engineered zebrafish mutant model provide additional support for ARMC9 as a ciliopathy-associated gene. Identifying ARMC9 mutations as a cause of JS takes us one step closer to a full genetic understanding of this important disorder and enables future functional work to define the central biological mechanisms underlying JS and other ciliopathies.


Subject(s)
Abnormalities, Multiple/genetics , Armadillo Domain Proteins/genetics , Basal Bodies/metabolism , Cerebellum/abnormalities , Ciliopathies/genetics , Eye Abnormalities/genetics , Kidney Diseases, Cystic/genetics , Mutation/genetics , Retina/abnormalities , Zebrafish Proteins/genetics , Zebrafish/genetics , Abnormalities, Multiple/pathology , Animals , Armadillo Domain Proteins/metabolism , Base Sequence , Brain/pathology , Cerebellum/pathology , Cilia/metabolism , Ciliopathies/pathology , Diagnostic Imaging , Exome/genetics , Eye Abnormalities/pathology , Genetic Predisposition to Disease , Humans , Kidney Diseases, Cystic/pathology , Phenotype , Retina/pathology , Sequence Analysis, DNA , Up-Regulation/genetics , Zebrafish Proteins/metabolism
7.
Am J Med Genet A ; 182(1): 229-249, 2020 01.
Article in English | MEDLINE | ID: mdl-31710777

ABSTRACT

Joubert syndrome (JS) is a recessive neurodevelopmental disorder defined by a characteristic cerebellar and brainstem malformation recognizable on axial brain magnetic resonance imaging as the "Molar Tooth Sign". Although defined by the neurological features, JS is associated with clinical features affecting many other organ systems, particularly progressive involvement of the retina, kidney, and liver. JS is a rare condition; therefore, many affected individuals may not have easy access to subspecialty providers familiar with JS (e.g., geneticists, neurologists, developmental pediatricians, ophthalmologists, nephrologists, hepatologists, psychiatrists, therapists, and educators). Expert recommendations can enable practitioners of all types to provide quality care to individuals with JS and know when to refer for subspecialty care. This need will only increase as precision treatments targeting specific genetic causes of JS emerge. The goal of these recommendations is to provide a resource for general practitioners, subspecialists, and families to maximize the health of individuals with JS throughout the lifespan.


Subject(s)
Abnormalities, Multiple/epidemiology , Cerebellum/abnormalities , Eye Abnormalities/epidemiology , Health Personnel , Kidney Diseases, Cystic/epidemiology , Neurodevelopmental Disorders/epidemiology , Retina/abnormalities , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Abnormalities, Multiple/therapy , Brain Stem/pathology , Cerebellum/pathology , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Eye Abnormalities/therapy , Health Planning Guidelines , Humans , Kidney/pathology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Kidney Diseases, Cystic/therapy , Liver/pathology , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Neurodevelopmental Disorders/therapy , Retina/pathology
8.
J Pathol ; 248(4): 393-395, 2019 08.
Article in English | MEDLINE | ID: mdl-31095728

ABSTRACT

Recent recognition of the key role of primary cilia in orchestrating human development and of the dire consequences of their dysfunction on human health has placed this small organelle in the spotlight. While the causal link between mutations in ciliary genes and central nervous system malformations and dysfunction is well established, the mechanisms by which primary cilia dysfunction acts on development and function of the CNS remain partly unknown. The recent article by Bashford and Subramanian in The Journal of Pathology describes a new mouse model for the neurodevelopmental ciliopathy Joubert syndrome, supporting a role for ciliary-mediated Hedgehog signaling on proliferation, survival, and differentiation of cerebellar granule cell progenitors. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Abnormalities, Multiple , Ciliopathies , Eye Abnormalities , Kidney Diseases, Cystic , Animals , Cell Cycle Proteins , Cerebellum , Cilia , Hedgehog Proteins , Humans , Mice , Retina , United Kingdom
9.
PLoS Genet ; 13(12): e1007150, 2017 12.
Article in English | MEDLINE | ID: mdl-29281629

ABSTRACT

Ciliopathies are human disorders caused by dysfunction of primary cilia, ubiquitous organelles involved in transduction of environmental signals such as light sensation in photoreceptors. Concentration of signal detection proteins such as opsins in the ciliary membrane is achieved by RabGTPase-regulated polarized vesicle trafficking and by a selective barrier at the ciliary base, the transition zone (TZ). Dysfunction of the TZ protein CC2D2A causes Joubert/Meckel syndromes in humans and loss of ciliary protein localization in animal models, including opsins in retinal photoreceptors. The link between the TZ and upstream vesicle trafficking has been little explored to date. Moreover, the role of the small GTPase Rab8 in opsin-carrier vesicle (OCV) trafficking has been recently questioned in a mouse model. Using correlative light and electron microscopy and live imaging in zebrafish photoreceptors, we provide the first live characterization of Rab8-mediated trafficking in photoreceptors in vivo. Our results support a possibly redundant role for both Rab8a/b paralogs in OCV trafficking, based on co-localization of Rab8 and opsins in vesicular structures, and joint movement of Rab8-tagged particles with opsin. We further investigate the role of the TZ protein Cc2d2a in Rab8-mediated trafficking using cc2d2a zebrafish mutants and identify a requirement for Cc2d2a in the latest step of OCV trafficking, namely vesicle fusion. Progressive accumulation of opsin-containing vesicles in the apical portion of photoreceptors lacking Cc2d2a is caused by disorganization of the vesicle fusion machinery at the periciliary membrane with mislocalization and loss of the t-SNAREs SNAP25 and Syntaxin3 and of the exocyst component Exoc4. We further observe secondary defects on upstream Rab8-trafficking with cytoplasmic accumulation of Rab8. Taken together, our results support participation of Rab8 in OCV trafficking and identify a novel role for the TZ protein Cc2d2a in fusion of incoming ciliary-directed vesicles, through organization of the vesicle fusion machinery at the periciliary membrane.


Subject(s)
Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Animals, Genetically Modified , Biological Transport , Cell Movement , Cilia/genetics , Cilia/metabolism , Humans , Membranes/metabolism , Opsins/genetics , Opsins/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Protein Transport , Zebrafish , rab GTP-Binding Proteins/genetics
10.
Mol Med ; 25(1): 6, 2019 02 27.
Article in English | MEDLINE | ID: mdl-30813884

ABSTRACT

BACKGROUND: Deleterious variants in the voltage-gated sodium channel type 2 (Nav1.2) lead to a broad spectrum of phenotypes ranging from benign familial neonatal-infantile epilepsy (BFNIE), severe developmental and epileptic encephalopathy (DEE) and intellectual disability (ID) to autism spectrum disorders (ASD). Yet, the underlying mechanisms are still incompletely understood. METHODS: To further elucidate the genotype-phenotype correlation of SCN2A variants we investigated the functional effects of six variants representing the phenotypic spectrum by whole-cell patch-clamp studies in transfected HEK293T cells and in-silico structural modeling. RESULTS: The two variants p.L1342P and p.E1803G detected in patients with early onset epileptic encephalopathy (EE) showed profound and complex changes in channel gating, whereas the BFNIE variant p.L1563V exhibited only a small gain of channel function. The three variants identified in ID patients without seizures, p.R937C, p.L611Vfs*35 and p.W1716*, did not produce measurable currents. Homology modeling of the missense variants predicted structural impairments consistent with the electrophysiological findings. CONCLUSIONS: Our findings support the hypothesis that complete loss-of-function variants lead to ID without seizures, small gain-of-function variants cause BFNIE and EE variants exhibit variable but profound Nav1.2 gating changes. Moreover, structural modeling was able to predict the severity of the variant impact, supporting a potential role of structural modeling as a prognostic tool. Our study on the functional consequences of SCN2A variants causing the distinct phenotypes of EE, BFNIE and ID contributes to the elucidation of mechanisms underlying the broad phenotypic variability reported for SCN2A variants.


Subject(s)
Epilepsy, Benign Neonatal/genetics , Epileptic Syndromes/genetics , Intellectual Disability/genetics , NAV1.2 Voltage-Gated Sodium Channel/physiology , Adolescent , Child , Epilepsy, Benign Neonatal/physiopathology , Epileptic Syndromes/physiopathology , Genetic Association Studies , HEK293 Cells , Humans , Intellectual Disability/physiopathology , Phenotype , Young Adult
11.
Genet Med ; 21(9): 2043-2058, 2019 09.
Article in English | MEDLINE | ID: mdl-30842647

ABSTRACT

PURPOSE: Microcephaly is a sign of many genetic conditions but has been rarely systematically evaluated. We therefore comprehensively studied the clinical and genetic landscape of an unselected cohort of patients with microcephaly. METHODS: We performed clinical assessment, high-resolution chromosomal microarray analysis, exome sequencing, and functional studies in 62 patients (58% with primary microcephaly [PM], 27% with secondary microcephaly [SM], and 15% of unknown onset). RESULTS: We found severity of developmental delay/intellectual disability correlating with severity of microcephaly in PM, but not SM. We detected causative variants in 48.4% of patients and found divergent inheritance and variant pattern for PM (mainly recessive and likely gene-disrupting [LGD]) versus SM (all dominant de novo and evenly LGD or missense). While centrosome-related pathways were solely identified in PM, transcriptional regulation was the most frequently affected pathway in both SM and PM. Unexpectedly, we found causative variants in different mitochondria-related genes accounting for ~5% of patients, which emphasizes their role even in syndromic PM. Additionally, we delineated novel candidate genes involved in centrosome-related pathway (SPAG5, TEDC1), Wnt signaling (VPS26A, ZNRF3), and RNA trafficking (DDX1). CONCLUSION: Our findings enable improved evaluation and genetic counseling of PM and SM patients and further elucidate microcephaly pathways.


Subject(s)
Developmental Disabilities/genetics , Genetic Predisposition to Disease , Intellectual Disability/genetics , Microcephaly/genetics , Adolescent , Cell Cycle Proteins/genetics , Child , Child, Preschool , DEAD-box RNA Helicases/genetics , Developmental Disabilities/pathology , Exome/genetics , Female , Gene Expression Regulation/genetics , Humans , Infant , Intellectual Disability/pathology , Male , Microcephaly/pathology , Mutation , Pedigree , Phenotype , Ubiquitin-Protein Ligases/genetics , Exome Sequencing , Wnt Signaling Pathway
12.
Genet Med ; 20(2): 223-233, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28771248

ABSTRACT

PurposeNext-generation sequencing (NGS) often identifies multiple rare predicted-deleterious variants (RDVs) in different genes associated with a recessive disorder in a given patient. Such variants have been proposed to contribute to digenicity/oligogenicity or "triallelism" or to act as genetic modifiers.MethodsUsing the recessive ciliopathy Joubert syndrome (JBTS) as a model, we investigated these possibilities systematically, relying on NGS of known JBTS genes in a large JBTS and two control cohorts.Results65% of affected individuals had a recessive genetic cause, while 4.9% were candidates for di-/oligogenicity, harboring heterozygous RDVs in two or more genes, compared with 4.2-8% in controls (P = 0.66-0.21). Based on Exome Aggregation Consortium (ExAC) allele frequencies, the probability of cumulating RDVs in any two JBTS genes is 9.3%. We found no support for triallelism, as no unaffected siblings carried the same biallelic RDVs as their affected relative. Sixty percent of individuals sharing identical causal RDVs displayed phenotypic discordance. Although 38% of affected individuals harbored RDVs in addition to the causal mutations, their presence did not correlate with phenotypic severity.ConclusionOur data offer little support for triallelism or digenicity/oligogenicity as clinically relevant inheritance modes in JBTS. While phenotypic discordance supports the existence of genetic modifiers, identifying clinically relevant modifiers remains challenging.


Subject(s)
Genes, Recessive , Genetic Association Studies , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Genetic Predisposition to Disease , Genetic Variation , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Alleles , Cerebellum/abnormalities , Eye Abnormalities/diagnosis , Eye Abnormalities/genetics , Genes, Modifier , Genetic Association Studies/methods , Humans , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/genetics , Models, Genetic , Multifactorial Inheritance , Mutation , Phenotype , Retina/abnormalities
13.
PLoS Genet ; 11(10): e1005574, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26485514

ABSTRACT

Ciliopathies are Mendelian disorders caused by dysfunction of cilia, ubiquitous organelles involved in fluid propulsion (motile cilia) or signal transduction (primary cilia). Retinal dystrophy is a common phenotypic characteristic of ciliopathies since photoreceptor outer segments are specialized primary cilia. These ciliary structures heavily rely on intracellular minus-end directed transport of cargo, mediated at least in part by the cytoplasmic dynein 1 motor complex, for their formation, maintenance and function. Ninein-like protein (NINL) is known to associate with this motor complex and is an important interaction partner of the ciliopathy-associated proteins lebercilin, USH2A and CC2D2A. Here, we scrutinize the function of NINL with combined proteomic and zebrafish in vivo approaches. We identify Double Zinc Ribbon and Ankyrin Repeat domains 1 (DZANK1) as a novel interaction partner of NINL and show that loss of Ninl, Dzank1 or both synergistically leads to dysmorphic photoreceptor outer segments, accumulation of trans-Golgi-derived vesicles and mislocalization of Rhodopsin and Ush2a in zebrafish. In addition, retrograde melanosome transport is severely impaired in zebrafish lacking Ninl or Dzank1. We further demonstrate that NINL and DZANK1 are essential for intracellular dynein-based transport by associating with complementary subunits of the cytoplasmic dynein 1 motor complex, thus shedding light on the structure and stoichiometry of this important motor complex. Altogether, our results support a model in which the NINL-DZANK1 protein module is involved in the proper assembly and folding of the cytoplasmic dynein 1 motor complex in photoreceptor cells, a process essential for outer segment formation and function.


Subject(s)
Carrier Proteins/genetics , Dyneins/genetics , Larva/genetics , Microtubule-Associated Proteins/genetics , Nuclear Proteins/genetics , Photoreceptor Cells, Vertebrate , Retina/growth & development , Zebrafish Proteins/genetics , Animals , Biological Transport/genetics , Cilia/genetics , HEK293 Cells , Humans , Larva/growth & development , Neurogenesis/genetics , Proteomics , Signal Transduction , Zebrafish/genetics , Zebrafish/growth & development
14.
PLoS Genet ; 11(10): e1005575, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26485645

ABSTRACT

Ciliopathies are a group of human disorders caused by dysfunction of primary cilia, ubiquitous microtubule-based organelles involved in transduction of extra-cellular signals to the cell. This function requires the concentration of receptors and channels in the ciliary membrane, which is achieved by complex trafficking mechanisms, in part controlled by the small GTPase RAB8, and by sorting at the transition zone located at the entrance of the ciliary compartment. Mutations in the transition zone gene CC2D2A cause the related Joubert and Meckel syndromes, two typical ciliopathies characterized by central nervous system malformations, and result in loss of ciliary localization of multiple proteins in various models. The precise mechanisms by which CC2D2A and other transition zone proteins control protein entrance into the cilium and how they are linked to vesicular trafficking of incoming cargo remain largely unknown. In this work, we identify the centrosomal protein NINL as a physical interaction partner of CC2D2A. NINL partially co-localizes with CC2D2A at the base of cilia and ninl knockdown in zebrafish leads to photoreceptor outer segment loss, mislocalization of opsins and vesicle accumulation, similar to cc2d2a-/- phenotypes. Moreover, partial ninl knockdown in cc2d2a-/- embryos enhances the retinal phenotype of the mutants, indicating a genetic interaction in vivo, for which an illustration is found in patients from a Joubert Syndrome cohort. Similar to zebrafish cc2d2a mutants, ninl morphants display altered Rab8a localization. Further exploration of the NINL-associated interactome identifies MICAL3, a protein known to interact with Rab8 and to play an important role in vesicle docking and fusion. Together, these data support a model where CC2D2A associates with NINL to provide a docking point for cilia-directed cargo vesicles, suggesting a mechanism by which transition zone proteins can control the protein content of the ciliary compartment.


Subject(s)
Cerebellum/abnormalities , Ciliary Motility Disorders/genetics , Encephalocele/genetics , Microtubule-Associated Proteins/metabolism , Mixed Function Oxygenases/genetics , Nuclear Proteins/metabolism , Polycystic Kidney Diseases/genetics , Proteins/genetics , Retina/abnormalities , rab GTP-Binding Proteins/genetics , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Animals , Cerebellum/metabolism , Cerebellum/pathology , Cilia/genetics , Cilia/metabolism , Cilia/pathology , Ciliary Motility Disorders/metabolism , Ciliary Motility Disorders/pathology , Cytoskeletal Proteins , Encephalocele/metabolism , Encephalocele/pathology , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , Eye Abnormalities/pathology , Gene Knockdown Techniques , Humans , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Microtubule-Associated Proteins/genetics , Mixed Function Oxygenases/metabolism , Mutation , Nuclear Proteins/genetics , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , Protein Transport/genetics , Proteins/metabolism , Retina/metabolism , Retina/pathology , Retinitis Pigmentosa , Signal Transduction , Zebrafish , rab GTP-Binding Proteins/metabolism
15.
Am J Hum Genet ; 94(1): 62-72, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24360808

ABSTRACT

Joubert syndrome (JBTS) is a recessive ciliopathy in which a subset of affected individuals also have the skeletal dysplasia Jeune asphyxiating thoracic dystrophy (JATD). Here, we have identified biallelic truncating CSPP1 (centrosome and spindle pole associated protein 1) mutations in 19 JBTS-affected individuals, four of whom also have features of JATD. CSPP1 mutations explain ∼5% of JBTS in our cohort, and despite truncating mutations in all affected individuals, the range of phenotypic severity is broad. Morpholino knockdown of cspp1 in zebrafish caused phenotypes reported in other zebrafish models of JBTS (curved body shape, pronephric cysts, and cerebellar abnormalities) and reduced ciliary localization of Arl13b, further supporting loss of CSPP1 function as a cause of JBTS. Fibroblasts from affected individuals with CSPP1 mutations showed reduced numbers of primary cilia and/or short primary cilia, as well as reduced axonemal localization of ciliary proteins ARL13B and adenylyl cyclase III. In summary, CSPP1 mutations are a major cause of the Joubert-Jeune phenotype in humans; however, the mechanism by which these mutations lead to both JBTS and JATD remains unknown.


Subject(s)
Cell Cycle Proteins/genetics , Cerebellar Diseases/genetics , Cilia/genetics , Ellis-Van Creveld Syndrome/genetics , Eye Abnormalities/genetics , Kidney Diseases, Cystic/genetics , Microtubule-Associated Proteins/genetics , Mutation , Retina/abnormalities , Abnormalities, Multiple , Adolescent , Animals , Cerebellum/abnormalities , Child , Child, Preschool , Cilia/pathology , Exons , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Knockdown Techniques , Humans , Infant , Male , Phenotype , Sequence Analysis, DNA , Young Adult , Zebrafish/genetics
16.
Am J Med Genet A ; 173(5): 1237-1242, 2017 May.
Article in English | MEDLINE | ID: mdl-28371402

ABSTRACT

Joubert syndrome (JS) is a rare, recessively inherited neurodevelopmental disorder characterized by a distinctive mid-hindbrain malformation. Little is known about mortality in affected individuals. Identifying the timing and causes of death will allow for development of healthcare guidelines for families and providers and, thus, help to prolong and improve the lives of patients with JS. We evaluated information on 40 deceased individuals with JS to characterize age and cause of death. We compared this population with 525 living individuals with JS to estimate associations between risk of death and extra-neurological features. Genetic causes were examined in both groups. Mean age of death in this cohort was 7.2 years, and the most prevalent causes of death were respiratory failure (35%), particularly in individuals younger than 6 years, and kidney failure (37.5%), which was more common in older individuals. We identified possible associations between risk of death and kidney disease, liver fibrosis, polydactyly, occipital encephalocele, and genetic cause. This work highlights factors (genetic cause, extra-neurological organ involvement, and other malformations) likely to be associated with higher risk of mortality in JS, which should prompt increased monitoring for respiratory issues, kidney disease, and liver fibrosis.


Subject(s)
Abnormalities, Multiple/mortality , Cerebellum/abnormalities , Eye Abnormalities/mortality , Kidney Diseases, Cystic/mortality , Renal Insufficiency/mortality , Retina/abnormalities , Abnormalities, Multiple/genetics , Abnormalities, Multiple/physiopathology , Adolescent , Cerebellum/physiopathology , Child , Child, Preschool , Eye Abnormalities/complications , Eye Abnormalities/genetics , Eye Abnormalities/physiopathology , Female , Humans , Kidney Diseases, Cystic/complications , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/physiopathology , Male , Renal Insufficiency/complications , Renal Insufficiency/genetics , Renal Insufficiency/pathology , Retina/physiopathology , Rhombencephalon/abnormalities , Rhombencephalon/physiopathology
17.
Hum Mutat ; 36(9): 831-5, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26096313

ABSTRACT

Joubert syndrome (JS) is a recessive neurodevelopmental disorder characterized by a distinctive mid-hindbrain malformation. JS is part of a group of disorders called ciliopathies based on their overlapping phenotypes and common underlying pathophysiology linked to primary cilium dysfunction. Biallelic mutations in one of 28 genes, all encoding proteins localizing to the primary cilium or basal body, can cause JS. Despite this large number of genes, the genetic cause can currently be determined in about 62% of individuals with JS. To identify novel JS genes, we performed whole exome sequencing on 35 individuals with JS and found biallelic rare deleterious variants (RDVs) in KIAA0586, encoding a centrosomal protein required for ciliogenesis, in one individual. Targeted next-generation sequencing in a large JS cohort identified biallelic RDVs in eight additional families for an estimated prevalence of 2.5% (9/366 JS families). All affected individuals displayed JS phenotypes toward the mild end of the spectrum.


Subject(s)
Cell Cycle Proteins/genetics , Cerebellum/abnormalities , Mutation , Retina/abnormalities , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Adolescent , Adult , Alternative Splicing , Brain/pathology , Child , Child, Preschool , DNA Mutational Analysis , Eye Abnormalities/diagnosis , Eye Abnormalities/genetics , Gene Order , Genetic Association Studies , Humans , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/genetics , Magnetic Resonance Imaging , Phenotype , Young Adult
18.
Med Sci (Paris) ; 30(11): 1011-23, 2014 Nov.
Article in French | MEDLINE | ID: mdl-25388584

ABSTRACT

Ciliopathies are a large group of human disorders caused by dysfunction of primary or motile cilia and unified by their overlapping clinical features (brain malformations, retinal dystrophy, cystic kidney disease, liver fibrosis and skeletal abnormalities). Ciliopathies are mendelian disorders with prominent genetic heterogeneity and marked allelism between different clinical entities, which are in part explained by the recently identified functional modules and multi-protein complexes formed by ciliopathy-associated gene products. The current review provides an updated snapshot of this complex evolving field, highlighting the key phenotypic features and causative genes for commonly-studied ciliopathies and summarizing our emerging understanding of the correlations between the functions of subgroups of genes and clinical sub-types of ciliopathies. Using the example of Joubert syndrome, a ciliopathy characterized by a distinctive hindbrain malformation and caused by mutations in more than 20 different genes, this work also reviews the principal methods used for new gene identification, including candidate gene approaches, homozygosity mapping as well as high throughput next-generation and exome sequencing.


Subject(s)
Ciliary Motility Disorders/genetics , Genetic Association Studies , Genetic Diseases, Inborn/genetics , Genetic Heterogeneity , Abnormalities, Multiple , Animals , Cell Polarity , Cerebellar Diseases/genetics , Cerebellar Diseases/pathology , Cerebellum/abnormalities , Chromosome Mapping , Cilia/chemistry , Cilia/physiology , Cilia/ultrastructure , Ciliary Motility Disorders/classification , Disease Models, Animal , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Forecasting , Genes, Recessive , Genetic Diseases, Inborn/pathology , Humans , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/physiology , Microtubule Proteins/deficiency , Microtubule Proteins/genetics , Microtubule Proteins/physiology , Molecular Motor Proteins/deficiency , Molecular Motor Proteins/genetics , Molecular Motor Proteins/physiology , Phenotype , Polymorphism, Single Nucleotide , Proteomics , Retina/abnormalities , Retina/pathology , Sequence Analysis, DNA/methods , Syndrome , Systems Biology/methods
19.
Hum Mol Genet ; 20(20): 4041-55, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21816947

ABSTRACT

Ciliopathies are a genetically and phenotypically heterogeneous group of human developmental disorders whose root cause is the absence or dysfunction of primary cilia. Joubert syndrome is characterized by a distinctive hindbrain malformation variably associated with retinal dystrophy and cystic kidney disease. Mutations in CC2D2A are found in ∼10% of patients with Joubert syndrome. Here we describe the retinal phenotype of cc2d2a mutant zebrafish consisting of disorganized rod and cone photoreceptor outer segments resulting in abnormal visual function as measured by electroretinogram. Our analysis reveals trafficking defects in mutant photoreceptors affecting transmembrane outer segment proteins (opsins) and striking accumulation of vesicles, suggesting a role for Cc2d2a in vesicle trafficking and fusion. This is further supported by mislocalization of Rab8, a key regulator of opsin carrier vesicle trafficking, in cc2d2a mutant photoreceptors and by enhancement of the cc2d2a retinal and kidney phenotypes with partial knockdown of rab8. We demonstrate that Cc2d2a localizes to the connecting cilium in photoreceptors and to the transition zone in other ciliated cell types and that cilia are present in these cells in cc2d2a mutants, arguing against a primary function for Cc2d2a in ciliogenesis. Our data support a model where Cc2d2a, localized at the photoreceptor connecting cilium/transition zone, facilitates protein transport through a role in Rab8-dependent vesicle trafficking and fusion.


Subject(s)
Cilia/genetics , Retinal Photoreceptor Cell Outer Segment/metabolism , Transport Vesicles/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/physiology , Zebrafish Proteins/physiology , Zebrafish/genetics , rab GTP-Binding Proteins/genetics , Animals , Animals, Genetically Modified , Cilia/metabolism , Female , Gene Knockout Techniques , HEK293 Cells , Humans , Male , Membrane Proteins/metabolism , Protein Binding , Protein Transport , Transport Vesicles/ultrastructure , Vesicular Transport Proteins/metabolism , Vision, Ocular/genetics , Zebrafish/embryology , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , rab GTP-Binding Proteins/metabolism
20.
J Med Genet ; 49(2): 126-37, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22241855

ABSTRACT

BACKGROUND: Joubert syndrome (JS) is a ciliopathy characterised by a distinctive brain malformation (the 'molar tooth sign'), developmental delay, abnormal eye movements and abnormal breathing pattern. Retinal dystrophy, cystic kidney disease, liver fibrosis and polydactyly are variably present, resulting in significant phenotypic heterogeneity and overlap with other ciliopathies. JS is also genetically heterogeneous, resulting from mutations in 13 genes. These factors render clinical/molecular diagnosis and management challenging. CC2D2A mutations are a relatively common cause of JS and also cause Meckel syndrome. The clinical consequences of CC2D2A mutations in patients with JS have been incompletely reported. METHODS: Subjects with JS from 209 families were evaluated to identify mutations in CC2D2A. Clinical and imaging features in subjects with CC2D2A mutations were compared with those in subjects without CC2D2A mutations and reports in the literature. RESULTS: 10 novel CC2D2A mutations in 20 subjects were identified; a summary is provided of all published CC2D2A mutations. Subjects with CC2D2A-related JS were more likely to have ventriculomegaly (p<0.0001) and seizures (p=0.024) than subjects without CC2D2A mutations. No mutation-specific genotype-phenotype correlations could be identified, but the findings confirm the observation that mutations that cause CC2D2A-related JS are predicted to be less deleterious than mutations that cause CC2D2A-related Meckel syndrome. Missense variants in the coiled-coil and C2 domains, as well as the C-terminal region, identify these regions as important for the biological mechanisms underlying JS. CONCLUSIONS: CC2D2A testing should be prioritised in patients with JS and ventriculomegaly and/or seizures. Patients with CC2D2A-related JS should be monitored for hydrocephalus and seizures.


Subject(s)
Cerebellar Diseases/genetics , Eye Abnormalities/genetics , Genetic Association Studies , Hydrocephalus/genetics , Kidney Diseases, Cystic/genetics , Proteins/genetics , Seizures/genetics , Abnormalities, Multiple , Adolescent , Adult , Alleles , Cerebellar Diseases/diagnosis , Cerebellar Diseases/epidemiology , Cerebellum/abnormalities , Child , Child, Preschool , Cytoskeletal Proteins , Eye Abnormalities/diagnosis , Eye Abnormalities/epidemiology , Genotype , Humans , Hydrocephalus/diagnosis , Infant , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/epidemiology , Magnetic Resonance Imaging , Neuroimaging , Phenotype , Prevalence , Retina/abnormalities , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL