Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 11: 1113656, 2023.
Article in English | MEDLINE | ID: mdl-36776558

ABSTRACT

Establishment and maintenance of the primary cilium as a signaling-competent organelle requires a high degree of fine tuning, which is at least in part achieved by a variety of post-translational modifications. One such modification is ubiquitination. The small and highly conserved ubiquitin protein possesses a unique versatility in regulating protein function via its ability to build mono and polyubiquitin chains onto target proteins. We aimed to take an unbiased approach to generate a comprehensive blueprint of the ciliary ubiquitinome by deploying a multi-proteomics approach using both ciliary-targeted ubiquitin affinity proteomics, as well as ubiquitin-binding domain-based proximity labelling in two different mammalian cell lines. This resulted in the identification of several key proteins involved in signaling, cytoskeletal remodeling and membrane and protein trafficking. Interestingly, using two different approaches in IMCD3 and RPE1 cells, respectively, we uncovered several novel mechanisms that regulate cilia function. In our IMCD3 proximity labeling cell line model, we found a highly enriched group of ESCRT-dependent clathrin-mediated endocytosis-related proteins, suggesting an important and novel role for this pathway in the regulation of ciliary homeostasis and function. In contrast, in RPE1 cells we found that several structural components of caveolae (CAV1, CAVIN1, and EHD2) were highly enriched in our cilia affinity proteomics screen. Consistently, the presence of caveolae at the ciliary pocket and ubiquitination of CAV1 specifically, were found likely to play a role in the regulation of ciliary length in these cells. Cilia length measurements demonstrated increased ciliary length in RPE1 cells stably expressing a ubiquitination impaired CAV1 mutant protein. Furthermore, live cell imaging in the same cells revealed decreased CAV1 protein turnover at the cilium as the possible cause for this phenotype. In conclusion, we have generated a comprehensive list of cilia-specific proteins that are subject to regulation via ubiquitination which can serve to further our understanding of cilia biology in health and disease.

2.
Cells ; 12(2)2023 01 13.
Article in English | MEDLINE | ID: mdl-36672247

ABSTRACT

Mutations in PDE6D impair the function of its cognate protein, phosphodiesterase 6D (PDE6D), in prenylated protein trafficking towards the ciliary membrane, causing the human ciliopathy Joubert Syndrome (JBTS22) and retinal degeneration in mice. In this study, we purified the prenylated cargo of PDE6D by affinity proteomics to gain insight into PDE6D-associated disease mechanisms. By this approach, we have identified a specific set of PDE6D-interacting proteins that are involved in photoreceptor integrity, GTPase activity, nuclear import, or ubiquitination. Among these interacting proteins, we identified novel ciliary cargo proteins of PDE6D, including FAM219A, serine/threonine-protein kinase NIM1 (NIM1K), and ubiquitin-like protein 3 (UBL3). We show that NIM1K and UBL3 localize inside the cilium in a prenylation-dependent manner. Furthermore, UBL3 also localizes in vesicle-like structures around the base of the cilium. Through affinity proteomics of UBL3, we confirmed its strong interaction with PDE6D and its association with proteins that regulate small extracellular vesicles (sEVs) and ciliogenesis. Moreover, we show that UBL3 localizes in specific photoreceptor cilium compartments in a prenylation-dependent manner. Therefore, we propose that UBL3 may play a role in the sorting of proteins towards the photoreceptor outer segment, further explaining the development of PDE6D-associated retinal degeneration.


Subject(s)
Cilia , Retinal Degeneration , Humans , Animals , Mice , Cilia/metabolism , Retinal Degeneration/metabolism , Proteins/metabolism , Retina/metabolism , Protein Transport , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism
3.
J Clin Invest ; 130(8): 4423-4439, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32453716

ABSTRACT

Joubert syndrome (JBTS) is a recessive neurodevelopmental ciliopathy characterized by a pathognomonic hindbrain malformation. All known JBTS genes encode proteins involved in the structure or function of primary cilia, ubiquitous antenna-like organelles essential for cellular signal transduction. Here, we used the recently identified JBTS-associated protein armadillo repeat motif-containing 9 (ARMC9) in tandem-affinity purification and yeast 2-hybrid screens to identify a ciliary module whose dysfunction underlies JBTS. In addition to the known JBTS-associated proteins CEP104 and CSPP1, we identified coiled-coil domain containing 66 (CCDC66) and TOG array regulator of axonemal microtubules 1 (TOGARAM1) as ARMC9 interaction partners. We found that TOGARAM1 variants cause JBTS and disrupt TOGARAM1 interaction with ARMC9. Using a combination of protein interaction analyses, characterization of patient-derived fibroblasts, and analysis of CRISPR/Cas9-engineered zebrafish and hTERT-RPE1 cells, we demonstrated that dysfunction of ARMC9 or TOGARAM1 resulted in short cilia with decreased axonemal acetylation and polyglutamylation, but relatively intact transition zone function. Aberrant serum-induced ciliary resorption and cold-induced depolymerization in ARMC9 and TOGARAM1 patient cell lines suggest a role for this new JBTS-associated protein module in ciliary stability.


Subject(s)
Abnormalities, Multiple , Armadillo Domain Proteins , Cerebellum/abnormalities , Cilia , Eye Abnormalities , Kidney Diseases, Cystic , Retina/abnormalities , Zebrafish Proteins , Zebrafish , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Acetylation , Animals , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , CRISPR-Cas Systems , Cerebellum/metabolism , Cilia/genetics , Cilia/metabolism , Disease Models, Animal , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , Humans , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Peptides/genetics , Peptides/metabolism , Retina/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 117(18): 9922-9931, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32312818

ABSTRACT

The outer segments (OS) of rod and cone photoreceptor cells are specialized sensory cilia that contain hundreds of opsin-loaded stacked membrane disks that enable phototransduction. The biogenesis of these disks is initiated at the OS base, but the driving force has been debated. Here, we studied the function of the protein encoded by the photoreceptor-specific gene C2orf71, which is mutated in inherited retinal dystrophy (RP54). We demonstrate that C2orf71/PCARE (photoreceptor cilium actin regulator) can interact with the Arp2/3 complex activator WASF3, and efficiently recruits it to the primary cilium. Ectopic coexpression of PCARE and WASF3 in ciliated cells results in the remarkable expansion of the ciliary tip. This process was disrupted by small interfering RNA (siRNA)-based down-regulation of an actin regulator, by pharmacological inhibition of actin polymerization, and by the expression of PCARE harboring a retinal dystrophy-associated missense mutation. Using human retinal organoids and mouse retina, we observed that a similar actin dynamics-driven process is operational at the base of the photoreceptor OS where the PCARE module and actin colocalize, but which is abrogated in Pcare-/- mice. The observation that several proteins involved in retinal ciliopathies are translocated to these expansions renders it a potential common denominator in the pathomechanisms of these hereditary disorders. Together, our work suggests that PCARE is an actin-associated protein that interacts with WASF3 to regulate the actin-driven expansion of the ciliary membrane at the initiation of new outer segment disk formation.


Subject(s)
Cilia/genetics , Cone-Rod Dystrophies/genetics , Eye Proteins/genetics , Rod Cell Outer Segment/metabolism , Wiskott-Aldrich Syndrome Protein Family/genetics , Actin-Related Protein 2-3 Complex/genetics , Actins/genetics , Animals , Cilia/pathology , Cone-Rod Dystrophies/pathology , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Mice , Mice, Knockout , RNA, Small Interfering/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Rod Cell Outer Segment/pathology
5.
PLoS One ; 14(5): e0216705, 2019.
Article in English | MEDLINE | ID: mdl-31095607

ABSTRACT

The cilium is an essential organelle at the surface of mammalian cells whose dysfunction causes a wide range of genetic diseases collectively called ciliopathies. The current rate at which new ciliopathy genes are identified suggests that many ciliary components remain undiscovered. We generated and rigorously analyzed genomic, proteomic, transcriptomic and evolutionary data and systematically integrated these using Bayesian statistics into a predictive score for ciliary function. This resulted in 285 candidate ciliary genes. We generated independent experimental evidence of ciliary associations for 24 out of 36 analyzed candidate proteins using multiple cell and animal model systems (mouse, zebrafish and nematode) and techniques. For example, we show that OSCP1, which has previously been implicated in two distinct non-ciliary processes, causes ciliogenic and ciliopathy-associated tissue phenotypes when depleted in zebrafish. The candidate list forms the basis of CiliaCarta, a comprehensive ciliary compendium covering 956 genes. The resource can be used to objectively prioritize candidate genes in whole exome or genome sequencing of ciliopathy patients and can be accessed at http://bioinformatics.bio.uu.nl/john/syscilia/ciliacarta/.


Subject(s)
Cilia/genetics , Genomics , Animals , Bayes Theorem , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Molecular Sequence Annotation , Phenotype , Reproducibility of Results , Sensory Receptor Cells/metabolism , Zebrafish/genetics
6.
Am J Hum Genet ; 101(5): 824-832, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29106825

ABSTRACT

The Rab GTPase family comprises ∼70 GTP-binding proteins, functioning in vesicle formation, transport and fusion. They are activated by a conformational change induced by GTP-binding, allowing interactions with downstream effectors. Here, we report five individuals with two recurrent de novo missense mutations in RAB11B; c.64G>A; p.Val22Met in three individuals and c.202G>A; p.Ala68Thr in two individuals. An overlapping neurodevelopmental phenotype, including severe intellectual disability with absent speech, epilepsy, and hypotonia was observed in all affected individuals. Additionally, visual problems, musculoskeletal abnormalities, and microcephaly were present in the majority of cases. Re-evaluation of brain MRI images of four individuals showed a shared distinct brain phenotype, consisting of abnormal white matter (severely decreased volume and abnormal signal), thin corpus callosum, cerebellar vermis hypoplasia, optic nerve hypoplasia and mild ventriculomegaly. To compare the effects of both variants with known inactive GDP- and active GTP-bound RAB11B mutants, we modeled the variants on the three-dimensional protein structure and performed subcellular localization studies. We predicted that both variants alter the GTP/GDP binding pocket and show that they both have localization patterns similar to inactive RAB11B. Evaluation of their influence on the affinity of RAB11B to a series of binary interactors, both effectors and guanine nucleotide exchange factors (GEFs), showed induction of RAB11B binding to the GEF SH3BP5, again similar to inactive RAB11B. In conclusion, we report two recurrent dominant mutations in RAB11B leading to a neurodevelopmental syndrome, likely caused by altered GDP/GTP binding that inactivate the protein and induce GEF binding and protein mislocalization.


Subject(s)
Epilepsy/genetics , Intellectual Disability/genetics , Muscle Hypotonia/genetics , Mutation , Optic Nerve Diseases/congenital , rab GTP-Binding Proteins/genetics , Adolescent , Amino Acid Sequence , Binding Sites , Cerebellar Vermis/diagnostic imaging , Cerebellar Vermis/metabolism , Cerebellar Vermis/pathology , Child , Child, Preschool , Corpus Callosum/diagnostic imaging , Corpus Callosum/metabolism , Corpus Callosum/pathology , Epilepsy/diagnostic imaging , Epilepsy/pathology , Female , Gene Expression , Guanosine Diphosphate/chemistry , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/pathology , Magnetic Resonance Imaging , Male , Models, Molecular , Muscle Hypotonia/diagnostic imaging , Muscle Hypotonia/pathology , Optic Nerve Diseases/diagnostic imaging , Optic Nerve Diseases/genetics , Optic Nerve Diseases/pathology , Phenotype , Protein Binding , White Matter/diagnostic imaging , White Matter/metabolism , White Matter/pathology , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/deficiency
7.
J Med Genet ; 54(9): 624-632, 2017 09.
Article in English | MEDLINE | ID: mdl-28442542

ABSTRACT

BACKGROUND: Recent findings suggesting that Abelson helper integration site 1 (AHI1) is involved in non-syndromic retinal disease have been debated, as the functional significance of identified missense variants was uncertain. We assessed whether AHI1 variants cause non-syndromic retinitis pigmentosa (RP). METHODS: Exome sequencing was performed in three probands with RP. The effects of the identified missense variants in AHI1 were predicted by three-dimensional structure homology modelling. Ciliary parameters were evaluated in patient's fibroblasts, and recombinant mutant proteins were expressed in ciliated retinal pigmented epithelium cells. RESULTS: In the three patients with RP, three sets of compound heterozygous variants were detected in AHI1 (c.2174G>A; p.Trp725* and c.2258A>T; p.Asp753Val, c.660delC; p.Ser221Glnfs*10 and c.2090C>T; p.Pro697Leu, c.2087A>G; p.His696Arg and c.2429C>T; p.Pro810Leu). All four missense variants were present in the conserved WD40 domain of Jouberin, the ciliary protein encoded by AHI1, with variable predicted implications for the domain structure. No significant changes in the percentage of ciliated cells, nor in cilium length or intraflagellar transport were detected. However, expression of mutant recombinant Jouberin in ciliated cells showed a significantly decreased enrichment at the ciliary base. CONCLUSIONS: This report confirms that mutations in AHI1 can underlie autosomal recessive RP. Moreover, it structurally and functionally validates the effect of the RP-associated AHI1 variants on protein function, thus proposing a new genotype-phenotype correlation for AHI1 mutation associated retinal ciliopathies.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Mutation, Missense , Retinitis Pigmentosa/genetics , Abnormalities, Multiple/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Vesicular Transport , Adult , Cerebellum/abnormalities , Eye Abnormalities/genetics , Female , Humans , Kidney Diseases, Cystic/genetics , Male , Middle Aged , Pedigree , Protein Domains/genetics , Retina/abnormalities
8.
Nat Commun ; 7: 11491, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27173435

ABSTRACT

Cellular organelles provide opportunities to relate biological mechanisms to disease. Here we use affinity proteomics, genetics and cell biology to interrogate cilia: poorly understood organelles, where defects cause genetic diseases. Two hundred and seventeen tagged human ciliary proteins create a final landscape of 1,319 proteins, 4,905 interactions and 52 complexes. Reverse tagging, repetition of purifications and statistical analyses, produce a high-resolution network that reveals organelle-specific interactions and complexes not apparent in larger studies, and links vesicle transport, the cytoskeleton, signalling and ubiquitination to ciliary signalling and proteostasis. We observe sub-complexes in exocyst and intraflagellar transport complexes, which we validate biochemically, and by probing structurally predicted, disruptive, genetic variants from ciliary disease patients. The landscape suggests other genetic diseases could be ciliary including 3M syndrome. We show that 3M genes are involved in ciliogenesis, and that patient fibroblasts lack cilia. Overall, this organelle-specific targeting strategy shows considerable promise for Systems Medicine.


Subject(s)
Cilia/metabolism , Ciliopathies/genetics , Dwarfism/genetics , Muscle Hypotonia/genetics , Protein Interaction Maps , Proteins/metabolism , Spine/abnormalities , Biological Transport/physiology , Chromatography, Affinity/methods , Ciliopathies/pathology , Ciliopathies/therapy , DNA Mutational Analysis , Datasets as Topic , Dwarfism/pathology , Dwarfism/therapy , Fibroblasts , HEK293 Cells , Humans , Mass Spectrometry , Molecular Targeted Therapy/methods , Muscle Hypotonia/pathology , Muscle Hypotonia/therapy , Protein Interaction Mapping/methods , Proteins/genetics , Proteins/isolation & purification , Proteomics/methods , Spine/pathology , Systems Analysis
10.
Genome Biol ; 16: 293, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26714646

ABSTRACT

BACKGROUND: Joubert syndrome (JBTS) and related disorders are defined by cerebellar malformation (molar tooth sign), together with neurological symptoms of variable expressivity. The ciliary basis of Joubert syndrome related disorders frequently extends the phenotype to tissues such as the eye, kidney, skeleton and craniofacial structures. RESULTS: Using autozygome and exome analyses, we identified a null mutation in KIAA0556 in a multiplex consanguineous family with hallmark features of mild Joubert syndrome. Patient-derived fibroblasts displayed reduced ciliogenesis potential and abnormally elongated cilia. Investigation of disease pathophysiology revealed that Kiaa0556 (-/-) null mice possess a Joubert syndrome-associated brain-restricted phenotype. Functional studies in Caenorhabditis elegans nematodes and cultured human cells support a conserved ciliary role for KIAA0556 linked to microtubule regulation. First, nematode KIAA0556 is expressed almost exclusively in ciliated cells, and the worm and human KIAA0556 proteins are enriched at the ciliary base. Second, C. elegans KIAA0056 regulates ciliary A-tubule number and genetically interacts with an ARL13B (JBTS8) orthologue to control cilium integrity. Third, human KIAA0556 binds to microtubules in vitro and appears to stabilise microtubule networks when overexpressed. Finally, human KIAA0556 biochemically interacts with ciliary proteins and p60/p80 katanins. The latter form a microtubule-severing enzyme complex that regulates microtubule dynamics as well as ciliary functions. CONCLUSIONS: We have identified KIAA0556 as a novel microtubule-associated ciliary base protein mutated in Joubert syndrome. Consistent with the mild patient phenotype, our nematode, mice and human cell data support the notion that KIAA0556 has a relatively subtle and variable cilia-related function, which we propose is related to microtubule regulation.


Subject(s)
Basal Bodies/metabolism , Cerebellum/abnormalities , Microtubule-Associated Proteins/genetics , Mutation , Retina/abnormalities , ADP-Ribosylation Factors/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Adenosine Triphosphatases/metabolism , Adult , Animals , Basal Bodies/pathology , Brain/metabolism , Brain/pathology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cells, Cultured , Cerebellum/pathology , Child , Child, Preschool , Cilia/genetics , Cilia/pathology , Exome , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Female , Humans , Katanin , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Pedigree , Protein Binding , Retina/pathology
11.
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
12.
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
13.
Structure ; 23(11): 2122-32, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26455799

ABSTRACT

Cilia are small antenna-like cellular protrusions critical for many developmental signaling pathways. The ciliary protein Arl3 has been shown to act as a specific release factor for myristoylated and farnesylated ciliary cargo molecules by binding to the effectors Unc119 and PDE6δ. Here we describe a newly identified Arl3 binding partner, CCDC104/CFAP36. Biochemical and structural analyses reveal that the protein contains a BART-like domain and is called BARTL1. It recognizes an LLxILxxL motif at the N-terminal amphipathic helix of Arl3, which is crucial for the interaction with the BART-like domain but also for the ciliary localization of Arl3 itself. These results seem to suggest a ciliary role of BARTL1, and possibly link it to the Arl3 transport network. We thus speculate on a regulatory mechanism whereby BARTL1 aids the presentation of active Arl3 to its GTPase-activating protein RP2 or hinders Arl3 membrane binding in the area of the transition zone.


Subject(s)
ADP-Ribosylation Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cilia/metabolism , Mice , Molecular Sequence Data , Protein Binding , Protein Transport , Proteins
14.
Nat Commun ; 6: 7074, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-26044572

ABSTRACT

The analysis of individuals with ciliary chondrodysplasias can shed light on sensitive mechanisms controlling ciliogenesis and cell signalling that are essential to embryonic development and survival. Here we identify TCTEX1D2 mutations causing Jeune asphyxiating thoracic dystrophy with partially penetrant inheritance. Loss of TCTEX1D2 impairs retrograde intraflagellar transport (IFT) in humans and the protist Chlamydomonas, accompanied by destabilization of the retrograde IFT dynein motor. We thus define TCTEX1D2 as an integral component of the evolutionarily conserved retrograde IFT machinery. In complex with several IFT dynein light chains, it is required for correct vertebrate skeletal formation but may be functionally redundant under certain conditions.


Subject(s)
Dyneins/genetics , Ellis-Van Creveld Syndrome/genetics , Flagella/physiology , Animals , Chlamydomonas reinhardtii , Cytoskeletal Proteins , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Mutation , Penetrance , Zebrafish
15.
Hum Mol Genet ; 24(6): 1584-601, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25398945

ABSTRACT

Leber congenital amaurosis (LCA) and juvenile retinitis pigmentosa (RP) are severe hereditary diseases that causes visual impairment in infants and children. SPATA7 has recently been identified as the LCA3 and juvenile RP gene in humans, whose function in the retina remains elusive. Here, we show that SPATA7 localizes at the primary cilium of cells and at the connecting cilium (CC) of photoreceptor cells, indicating that SPATA7 is a ciliary protein. In addition, SPATA7 directly interacts with the retinitis pigmentosa GTPase regulator interacting protein 1 (RPGRIP1), a key connecting cilium protein that has also been linked to LCA. In the retina of Spata7 null mutant mice, a substantial reduction of RPGRIP1 levels at the CC of photoreceptor cells is observed, suggesting that SPATA7 is required for the stable assembly and localization of the ciliary RPGRIP1 protein complex. Furthermore, our results pinpoint a role of this complex in protein trafficking across the CC to the outer segments, as we identified that rhodopsin accumulates in the inner segments and around the nucleus of photoreceptors. This accumulation then likely triggers the apoptosis of rod photoreceptors that was observed. Loss of Spata7 function in mice indeed results in a juvenile RP-like phenotype, characterized by progressive degeneration of photoreceptor cells and a strongly decreased light response. Together, these results indicate that SPATA7 functions as a key member of a retinal ciliopathy-associated protein complex, and that apoptosis of rod photoreceptor cells triggered by protein mislocalization is likely the mechanism of disease progression in LCA3/ juvenile RP patients.


Subject(s)
DNA-Binding Proteins/metabolism , Photoreceptor Connecting Cilium/pathology , Proteins/metabolism , Retinal Rod Photoreceptor Cells/pathology , Animals , Apoptosis , Cattle , Cytoskeletal Proteins , DNA-Binding Proteins/genetics , Gene Deletion , Humans , Mice , Mice, Mutant Strains , Photoreceptor Connecting Cilium/metabolism , Protein Transport , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/metabolism , Rhodopsin/metabolism
16.
PLoS Genet ; 9(12): e1003977, 2013.
Article in English | MEDLINE | ID: mdl-24339792

ABSTRACT

Cilia are microtubule-based cell appendages, serving motility, chemo-/mechano-/photo- sensation, and developmental signaling functions. Cilia are comprised of distinct structural and functional subregions including the basal body, transition zone (TZ) and inversin (Inv) compartments, and defects in this organelle are associated with an expanding spectrum of inherited disorders including Bardet-Biedl syndrome (BBS), Meckel-Gruber Syndrome (MKS), Joubert Syndrome (JS) and Nephronophthisis (NPHP). Despite major advances in understanding ciliary trafficking pathways such as intraflagellar transport (IFT), how proteins are transported to subciliary membranes remains poorly understood. Using Caenorhabditis elegans and mammalian cells, we investigated the transport mechanisms underlying compartmentalization of JS-associated ARL13B/ARL-13, which we previously found is restricted at proximal ciliary membranes. We now show evolutionary conservation of ARL13B/ARL-13 localisation to an Inv-like subciliary membrane compartment, excluding the TZ, in many C. elegans ciliated neurons and in a subset of mammalian ciliary subtypes. Compartmentalisation of C. elegans ARL-13 requires a C-terminal RVVP motif and membrane anchoring to prevent distal cilium and nuclear targeting, respectively. Quantitative imaging in more than 20 mutants revealed differential contributions for IFT and ciliopathy modules in defining the ARL-13 compartment; IFT-A/B, IFT-dynein and BBS genes prevent ARL-13 accumulation at periciliary membranes, whereas MKS/NPHP modules additionally inhibit ARL-13 association with TZ membranes. Furthermore, in vivo FRAP analyses revealed distinct roles for IFT and MKS/NPHP genes in regulating a TZ barrier to ARL-13 diffusion, and intraciliary ARL-13 diffusion. Finally, C. elegans ARL-13 undergoes IFT-like motility and quantitative protein complex analysis of human ARL13B identified functional associations with IFT-B complexes, mapped to IFT46 and IFT74 interactions. Together, these findings reveal distinct requirements for sequence motifs, IFT and ciliopathy modules in defining an ARL-13 subciliary membrane compartment. We conclude that MKS/NPHP modules comprise a TZ barrier to ARL-13 diffusion, whereas IFT genes predominantly facilitate ARL-13 ciliary entry and/or retention via active transport mechanisms.


Subject(s)
ADP-Ribosylation Factors/genetics , Caenorhabditis elegans/genetics , Cerebellar Diseases/genetics , Cilia/genetics , Eye Abnormalities/genetics , Kidney Diseases, Cystic/genetics , Retina/abnormalities , ADP-Ribosylation Factors/metabolism , Abnormalities, Multiple , Animals , Bardet-Biedl Syndrome/genetics , Bardet-Biedl Syndrome/pathology , Biological Transport, Active/genetics , Caenorhabditis elegans/metabolism , Cerebellar Diseases/pathology , Cerebellum/abnormalities , Cilia/metabolism , Ciliary Motility Disorders/genetics , Ciliary Motility Disorders/pathology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Encephalocele/genetics , Encephalocele/pathology , Eye Abnormalities/pathology , Humans , Kidney Diseases, Cystic/pathology , Membranes/metabolism , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/pathology , Retina/pathology , Retinitis Pigmentosa , Transcription Factors/genetics , Transcription Factors/metabolism
17.
PLoS One ; 8(11): e79369, 2013.
Article in English | MEDLINE | ID: mdl-24223178

ABSTRACT

Leber congenital amaurosis (LCA) is the most severe form of retinal dystrophy with an onset in the first year of life. The most frequent genetic cause of LCA, accounting for up to 15% of all LCA cases in Europe and North-America, is a mutation (c.2991+1655AG) in intron 26 of CEP290. This mutation generates a cryptic splice donor site resulting in the insertion of an aberrant exon (exon X) containing a premature stop codon to CEP290 mRNA. In order to study the pathophysiology of the intronic CEP290 mutation, we generated two humanized knock-in mouse models each carrying ~6.3 kb of the human CEP290 gene, either with or without the intronic mutation. Transcriptional characterization of these mouse models revealed an unexpected splice pattern of CEP290 mRNA, especially in the retina. In both models, a new cryptic exon (coined exon Y) was identified in ~5 to 12% of all Cep290 transcripts. This exon Y was expressed in all murine tissues analyzed but not detected in human retina or fibroblasts of LCA patients. In addition, exon x that is characteristic of LCA in humans, was expressed at only very low levels in the retina of the LCA mouse model. Western blot and immunohistochemical analyses did not reveal any differences between the two transgenic models and wild-type mice. Together, our results show clear differences in the recognition of splice sites between mice and humans, and emphasize that care is warranted when generating animal models for human genetic diseases caused by splice mutations.


Subject(s)
Antigens, Neoplasm/genetics , Disease Models, Animal , Gene Knock-In Techniques , Leber Congenital Amaurosis/genetics , Neoplasm Proteins/genetics , RNA Splicing , Animals , Cell Cycle Proteins , Cell Line , Cytoskeletal Proteins , Exons/genetics , Female , Humans , Mice , Pregnancy , Retina/metabolism , Species Specificity , Transcription, Genetic/genetics
18.
J Med Genet ; 48(6): 390-5, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21378380

ABSTRACT

BACKGROUND: Sensenbrenner syndrome is a heterogeneous ciliopathy that is characterised by skeletal and ectodermal anomalies, accompanied by chronic renal failure, heart defects, liver fibrosis and other features. OBJECTIVE: To identify an additional causative gene in Sensenbrenner syndrome. METHODS: Single nucleotide polymorphism array analysis and standard sequencing techniques were applied to identify the causative gene. The effect of the identified mutation on protein translation was determined by western blot analysis. Antibodies against intraflagellar transport (IFT) proteins were used in ciliated fibroblast cell lines to investigate the molecular consequences of the mutation on ciliary transport. RESULTS: Homozygosity mapping and positional candidate gene sequence analysis were performed in two siblings with Sensenbrenner syndrome of a consanguineous Moroccan family. In both siblings, a homozygous mutation in the initiation codon of C14ORF179 was identified. C14ORF179 encodes IFT43, a subunit of the IFT complex A (IFT-A) machinery of primary cilia. Western blots showed that the mutation disturbs translation of IFT43, inducing the initiation of translation of a shorter protein product from a downstream ATG. The IFT-A protein complex is implicated in retrograde ciliary transport along axonemal microtubules. It was shown that in fibroblasts of one of the siblings affected by Sensenbrenner syndrome, disruption of IFT43 disturbs this transport from the ciliary tip to its base. As anterograde transport in the opposite direction apparently remains functional, the IFT complex B proteins accumulate in the ciliary tip. Interestingly, similar results were obtained using fibroblasts from a patient with Sensenbrenner syndrome with mutations in WDR35/IFT121, encoding another IFT-A subunit. CONCLUSIONS: The results indicate that Sensenbrenner syndrome is caused by disrupted IFT-A-mediated retrograde ciliary transport.


Subject(s)
Carrier Proteins/genetics , Cilia/metabolism , Craniofacial Abnormalities/genetics , Ectodermal Dysplasia/genetics , Flagella/metabolism , Protein Transport/genetics , Recombinant Proteins/genetics , Animals , Base Sequence , Carrier Proteins/metabolism , Child , Cilia/genetics , Craniofacial Abnormalities/ethnology , Ectodermal Dysplasia/ethnology , Fibroblasts/physiology , Flagella/genetics , HEK293 Cells , Humans , Male , Molecular Sequence Data , Morocco/ethnology , Mutation , Netherlands/epidemiology , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Recombinant Proteins/metabolism , Siblings , Syndrome , Transfection
19.
Am J Hum Genet ; 83(5): 559-71, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18950740

ABSTRACT

Joubert syndrome and related disorders (JSRD) are primarily autosomal-recessive conditions characterized by hypotonia, ataxia, abnormal eye movements, and intellectual disability with a distinctive mid-hindbrain malformation. Variable features include retinal dystrophy, cystic kidney disease, and liver fibrosis. JSRD are included in the rapidly expanding group of disorders called ciliopathies, because all six gene products implicated in JSRD (NPHP1, AHI1, CEP290, RPGRIP1L, TMEM67, and ARL13B) function in the primary cilium/basal body organelle. By using homozygosity mapping in consanguineous families, we identify loss-of-function mutations in CC2D2A in JSRD patients with and without retinal, kidney, and liver disease. CC2D2A is expressed in all fetal and adult tissues tested. In ciliated cells, we observe localization of recombinant CC2D2A at the basal body and colocalization with CEP290, whose cognate gene is mutated in multiple hereditary ciliopathies. In addition, the proteins can physically interact in vitro, as shown by yeast two-hybrid and GST pull-down experiments. A nonsense mutation in the zebrafish CC2D2A ortholog (sentinel) results in pronephric cysts, a hallmark of ciliary dysfunction analogous to human cystic kidney disease. Knockdown of cep290 function in sentinel fish results in a synergistic pronephric cyst phenotype, revealing a genetic interaction between CC2D2A and CEP290 and implicating CC2D2A in cilium/basal body function. These observations extend the genetic spectrum of JSRD and provide a model system for studying extragenic modifiers in JSRD and other ciliopathies.


Subject(s)
Abnormalities, Multiple/genetics , Antigens, Neoplasm/metabolism , Mutation , Neoplasm Proteins/metabolism , Proteins/genetics , Proteins/metabolism , Antigens, Neoplasm/genetics , Ataxia/genetics , Cell Cycle Proteins , Cerebellum/abnormalities , Cerebellum/diagnostic imaging , Chromosome Mapping , Chromosomes, Human, Pair 4 , Cilia/genetics , Cohort Studies , Consanguinity , Cytoskeletal Proteins , Exons , Genetic Markers , Haplotypes , Homozygote , Humans , Immunohistochemistry , Kidney Diseases, Cystic/genetics , Male , Microsatellite Repeats , Muscle Hypotonia/genetics , Neoplasm Proteins/genetics , Ocular Motility Disorders/genetics , Pedigree , Polymorphism, Single Nucleotide , Radiography , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Syndrome , Two-Hybrid System Techniques
20.
Nat Genet ; 39(7): 889-95, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17546029

ABSTRACT

Leber congenital amaurosis (LCA) causes blindness or severe visual impairment at or within a few months of birth. Here we show, using homozygosity mapping, that the LCA5 gene on chromosome 6q14, which encodes the previously unknown ciliary protein lebercilin, is associated with this disease. We detected homozygous nonsense and frameshift mutations in LCA5 in five families affected with LCA. In a sixth family, the LCA5 transcript was completely absent. LCA5 is expressed widely throughout development, although the phenotype in affected individuals is limited to the eye. Lebercilin localizes to the connecting cilia of photoreceptors and to the microtubules, centrioles and primary cilia of cultured mammalian cells. Using tandem affinity purification, we identified 24 proteins that link lebercilin to centrosomal and ciliary functions. Members of this interactome represent candidate genes for LCA and other ciliopathies. Our findings emphasize the emerging role of disrupted ciliary processes in the molecular pathogenesis of LCA.


Subject(s)
Eye Proteins/genetics , Microtubule-Associated Proteins/genetics , Optic Atrophy, Hereditary, Leber/genetics , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Cilia/genetics , Codon, Nonsense , Eye Proteins/metabolism , Female , Frameshift Mutation , Humans , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Pedigree , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...