Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 592
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 88: 691-724, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30601682

ABSTRACT

The centriole is an ancient microtubule-based organelle with a conserved nine-fold symmetry. Centrioles form the core of centrosomes, which organize the interphase microtubule cytoskeleton of most animal cells and form the poles of the mitotic spindle. Centrioles can also be modified to form basal bodies, which template the formation of cilia and play central roles in cellular signaling, fluid movement, and locomotion. In this review, we discuss developments in our understanding of the biogenesis of centrioles and cilia and the regulatory controls that govern their structure and number. We also discuss how defects in these processes contribute to a spectrum of human diseases and how new technologies have expanded our understanding of centriole and cilium biology, revealing exciting avenues for future exploration.


Subject(s)
Centrioles/physiology , Cilia/pathology , Organelle Biogenesis , Animals , Cell Cycle , Centrioles/metabolism , Centrioles/ultrastructure , Cilia/metabolism , Cilia/ultrastructure , Ciliopathies , Eukaryota/cytology , Eukaryota/physiology , Humans , Mitosis , Signal Transduction
2.
Am J Hum Genet ; 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39191256

ABSTRACT

Genome analysis of individuals affected by retinitis pigmentosa (RP) identified two rare nucleotide substitutions at the same genomic location on chromosome 11 (g.61392563 [GRCh38]), 69 base pairs upstream of the start codon of the ciliopathy gene TMEM216 (c.-69G>A, c.-69G>T [GenBank: NM_001173991.3]), in individuals of South Asian and African ancestry, respectively. Genotypes included 71 homozygotes and 3 mixed heterozygotes in trans with a predicted loss-of-function allele. Haplotype analysis showed single-nucleotide variants (SNVs) common across families, suggesting ancestral alleles within the two distinct ethnic populations. Clinical phenotype analysis of 62 available individuals from 49 families indicated a similar clinical presentation with night blindness in the first decade and progressive peripheral field loss thereafter. No evident systemic ciliopathy features were noted. Functional characterization of these variants by luciferase reporter gene assay showed reduced promotor activity. Nanopore sequencing confirmed the lower transcription of the TMEM216 c.-69G>T allele in blood-derived RNA from a heterozygous carrier, and reduced expression was further recapitulated by qPCR, using both leukocytes-derived RNA of c.-69G>T homozygotes and total RNA from genome-edited hTERT-RPE1 cells carrying homozygous TMEM216 c.-69G>A. In conclusion, these variants explain a significant proportion of unsolved cases, specifically in individuals of African ancestry, suggesting that reduced TMEM216 expression might lead to abnormal ciliogenesis and photoreceptor degeneration.

3.
Development ; 151(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38031972

ABSTRACT

Cilia are microtubule (MT)-based organelles present on the surface of nearly all vertebrate cells. MTs are polymers of α- and ß-tubulins that are each encoded by multiple, individual isotype genes. Tubulin isotype composition is thought to influence MT behaviors. Ciliary MTs differ from other MTs in the cell in terms of organization, stability and post-translational modifications. However, little is known about the tubulin isotypes that build ciliary MTs and the functional requirements for tubulin isotypes in cilia have not been examined in vertebrates. Here, we have tested the role of the ß-tubulin isotype genes in the mouse that harbor a conserved amino acid motif associated with ciliated organisms. We found that Tubb4b localizes to cilia in multi-ciliated cells (MCCs) specifically. In respiratory and oviduct MCCs, Tubb4b is asymmetrically localized within multi-cilia, indicating that the tubulin isotype composition changes along the length of the ciliary axonemal MTs. Deletion of Tubb4b resulted in striking structural defects within the axonemes of multi-cilia, without affecting primary cilia. These studies show that Tubb4b is essential for the formation of a specific MT-based subcellular organelle and sheds light on the requirements of tubulin isotypes in cilia.


Subject(s)
Cilia , Tubulin , Animals , Mice , Axoneme/metabolism , Cilia/metabolism , Microtubules/metabolism , Protein Processing, Post-Translational , Tubulin/genetics , Tubulin/metabolism
4.
Proc Natl Acad Sci U S A ; 121(27): e2314702121, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38916997

ABSTRACT

Enlargement of the cerebrospinal fluid (CSF)-filled brain ventricles (cerebral ventriculomegaly), the cardinal feature of congenital hydrocephalus (CH), is increasingly recognized among patients with autism spectrum disorders (ASD). KATNAL2, a member of Katanin family microtubule-severing ATPases, is a known ASD risk gene, but its roles in human brain development remain unclear. Here, we show that nonsense truncation of Katnal2 (Katnal2Δ17) in mice results in classic ciliopathy phenotypes, including impaired spermatogenesis and cerebral ventriculomegaly. In both humans and mice, KATNAL2 is highly expressed in ciliated radial glia of the fetal ventricular-subventricular zone as well as in their postnatal ependymal and neuronal progeny. The ventriculomegaly observed in Katnal2Δ17 mice is associated with disrupted primary cilia and ependymal planar cell polarity that results in impaired cilia-generated CSF flow. Further, prefrontal pyramidal neurons in ventriculomegalic Katnal2Δ17 mice exhibit decreased excitatory drive and reduced high-frequency firing. Consistent with these findings in mice, we identified rare, damaging heterozygous germline variants in KATNAL2 in five unrelated patients with neurosurgically treated CH and comorbid ASD or other neurodevelopmental disorders. Mice engineered with the orthologous ASD-associated KATNAL2 F244L missense variant recapitulated the ventriculomegaly found in human patients. Together, these data suggest KATNAL2 pathogenic variants alter intraventricular CSF homeostasis and parenchymal neuronal connectivity by disrupting microtubule dynamics in fetal radial glia and their postnatal ependymal and neuronal descendants. The results identify a molecular mechanism underlying the development of ventriculomegaly in a genetic subset of patients with ASD and may explain persistence of neurodevelopmental phenotypes in some patients with CH despite neurosurgical CSF shunting.


Subject(s)
Cilia , Hydrocephalus , Microtubules , Animals , Female , Humans , Male , Mice , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/metabolism , Cilia/metabolism , Cilia/pathology , Ependyma/metabolism , Ependyma/pathology , Hydrocephalus/genetics , Hydrocephalus/pathology , Hydrocephalus/metabolism , Katanin/metabolism , Katanin/genetics , Microtubules/metabolism , Neurons/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/pathology
5.
Genes Dev ; 33(19-20): 1381-1396, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31488579

ABSTRACT

Short telomere syndromes manifest as familial idiopathic pulmonary fibrosis; they are the most common premature aging disorders. We used genome-wide linkage to identify heterozygous loss of function of ZCCHC8, a zinc-knuckle containing protein, as a cause of autosomal dominant pulmonary fibrosis. ZCCHC8 associated with TR and was required for telomerase function. In ZCCHC8 knockout cells and in mutation carriers, genomically extended telomerase RNA (TR) accumulated at the expense of mature TR, consistent with a role for ZCCHC8 in mediating TR 3' end targeting to the nuclear RNA exosome. We generated Zcchc8-null mice and found that heterozygotes, similar to human mutation carriers, had TR insufficiency but an otherwise preserved transcriptome. In contrast, Zcchc8-/- mice developed progressive and fatal neurodevelopmental pathology with features of a ciliopathy. The Zcchc8-/- brain transcriptome was highly dysregulated, showing accumulation and 3' end misprocessing of other low-abundance RNAs, including those encoding cilia components as well as the intronless replication-dependent histones. Our data identify a novel cause of human short telomere syndromes-familial pulmonary fibrosis and uncover nuclear exosome targeting as an essential 3' end maturation mechanism that vertebrate TR shares with replication-dependent histones.


Subject(s)
Carrier Proteins/genetics , Idiopathic Pulmonary Fibrosis/genetics , Loss of Function Mutation , Nuclear Proteins/genetics , RNA/metabolism , Telomerase/metabolism , Animals , Brain/enzymology , Brain/physiopathology , Cell Line , Cilia/genetics , Female , Genetic Linkage , HCT116 Cells , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/physiopathology , Male , Mice , Mice, Knockout , Neurodevelopmental Disorders/genetics , Pedigree , RNA Processing, Post-Transcriptional/genetics , Telomere Shortening/genetics
6.
Traffic ; 25(1): e12929, 2024 01.
Article in English | MEDLINE | ID: mdl-38272449

ABSTRACT

Ciliary transport in eukaryotic cells is an intricate and conserved process involving the coordinated assembly and functioning of a multiprotein intraflagellar transport (IFT) complex. Among the various IFT proteins, intraflagellar transport 52 (IFT52) plays a crucial role in ciliary transport and is implicated in various ciliopathies. IFT52 is a core component of the IFT-B complex that facilitates movement of cargoes along the ciliary axoneme. Stable binding of the IFT-B1 and IFT-B2 subcomplexes by IFT52 in the IFT-B complex regulates recycling of ciliary components and maintenance of ciliary functions such as signal transduction and molecular movement. Mutations in the IFT52 gene can disrupt ciliary trafficking, resulting in dysfunctional cilia and affecting cellular processes in ciliopathies. Such ciliopathies caused by IFT52 mutations exhibit a wide range of clinical features, including skeletal developmental abnormalities, retinal degeneration, respiratory failure and neurological abnormalities in affected individuals. Therefore, IFT52 serves as a promising biomarker for the diagnosis of various ciliopathies, including short-rib thoracic dysplasia 16 with or without polydactyly. Here, we provide an overview of the IFT52-mediated molecular mechanisms underlying ciliary transport and describe the IFT52 mutations that cause different disorders associated with cilia dysfunction.


Subject(s)
Cilia , Ciliopathies , Humans , Biological Transport , Cilia/metabolism , Ciliopathies/genetics , Ciliopathies/metabolism , Flagella/genetics , Flagella/metabolism , Mutation , Protein Transport , Proteins/metabolism , Signal Transduction
7.
Hum Mol Genet ; 33(16): 1442-1453, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38751342

ABSTRACT

Primary cilia are antenna-like structures protruding from the surface of various eukaryotic cells, and have distinct protein compositions in their membranes. This distinct protein composition is maintained by the presence of the transition zone (TZ) at the ciliary base, which acts as a diffusion barrier between the ciliary and plasma membranes. Defects in cilia and the TZ are known to cause a group of disorders collectively called the ciliopathies, which demonstrate a broad spectrum of clinical features, such as perinatally lethal Meckel syndrome (MKS), relatively mild Joubert syndrome (JBTS), and nonsyndromic nephronophthisis (NPHP). Proteins constituting the TZ can be grouped into the MKS and NPHP modules. The MKS module is composed of several transmembrane proteins and three soluble proteins. TMEM218 was recently reported to be mutated in individuals diagnosed as MKS and JBTS. However, little is known about how TMEM218 mutations found in MKS and JBTS affect the functions of cilia. In this study, we found that ciliary membrane proteins were not localized to cilia in TMEM218-knockout cells, indicating impaired barrier function of the TZ. Furthermore, the exogenous expression of JBTS-associated TMEM218 variants but not MKS-associated variants in TMEM218-knockout cells restored the localization of ciliary membrane proteins. In particular, when expressed in TMEM218-knockout cells, the TMEM218(R115H) variant found in JBTS was able to restore the barrier function of cells, whereas the MKS variant TMEM218(R115C) could not. Thus, the severity of symptoms of MKS and JBTS individuals appears to correlate with the degree of their ciliary defects at the cellular level.


Subject(s)
Abnormalities, Multiple , Cilia , Ciliopathies , Encephalocele , Eye Abnormalities , Kidney Diseases, Cystic , Membrane Proteins , Mutation , Retina , Cilia/metabolism , Cilia/genetics , Cilia/pathology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Ciliopathies/genetics , Ciliopathies/metabolism , Ciliopathies/pathology , Encephalocele/genetics , Encephalocele/metabolism , Encephalocele/pathology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Eye Abnormalities/metabolism , Retina/metabolism , Retina/abnormalities , Retina/pathology , Cerebellum/abnormalities , Cerebellum/metabolism , Cerebellum/pathology , Cerebellar Diseases/genetics , Cerebellar Diseases/metabolism , Cerebellar Diseases/pathology , Animals , Cell Membrane/metabolism , Mice , Ciliary Motility Disorders , Polycystic Kidney Diseases , Retinitis Pigmentosa
8.
J Cell Sci ; 137(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39056167

ABSTRACT

The primary cilium is a small organelle protruding from the cell surface that receives signals from the extracellular milieu. Although dozens of studies have reported that several genetic factors can impair the structure of primary cilia, evidence for environmental stimuli affecting primary cilia structures is limited. Here, we investigated an extracellular stress that affected primary cilia morphology and its underlying mechanisms. Hyperosmotic shock induced reversible shortening and disassembly of the primary cilia of murine intramedullary collecting duct cells. The shortening of primary cilia caused by hyperosmotic shock followed delocalization of the pericentriolar material (PCM). Excessive microtubule and F-actin formation in the cytoplasm coincided with the hyperosmotic shock-induced changes to primary cilia and the PCM. Treatment with a microtubule-disrupting agent, nocodazole, partially prevented the hyperosmotic shock-induced disassembly of primary cilia and almost completely prevented delocalization of the PCM. An actin polymerization inhibitor, latrunculin A, also partially prevented the hyperosmotic shock-induced shortening and disassembly of primary cilia and almost completely prevented delocalization of the PCM. We demonstrate that hyperosmotic shock induces reversible morphological changes in primary cilia and the PCM in a manner dependent on excessive formation of microtubule and F-actin.


Subject(s)
Actins , Cilia , Microtubules , Osmotic Pressure , Cilia/metabolism , Cilia/drug effects , Animals , Microtubules/metabolism , Microtubules/drug effects , Actins/metabolism , Mice , Nocodazole/pharmacology , Thiazolidines/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/cytology
9.
J Cell Sci ; 137(13)2024 07 01.
Article in English | MEDLINE | ID: mdl-38841887

ABSTRACT

Centrosomal proteins play pivotal roles in orchestrating microtubule dynamics, and their dysregulation leads to disorders, including cancer and ciliopathies. Understanding the multifaceted roles of centrosomal proteins is vital to comprehend their involvement in disease development. Here, we report novel cellular functions of CEP41, a centrosomal and ciliary protein implicated in Joubert syndrome. We show that CEP41 is an essential microtubule-associated protein with microtubule-stabilizing activity. Purified CEP41 binds to preformed microtubules, promotes microtubule nucleation and suppresses microtubule disassembly. When overexpressed in cultured cells, CEP41 localizes to microtubules and promotes microtubule bundling. Conversely, shRNA-mediated knockdown of CEP41 disrupts the interphase microtubule network and delays microtubule reassembly, emphasizing its role in microtubule organization. Further, we demonstrate that the association of CEP41 with microtubules relies on its conserved rhodanese homology domain (RHOD) and the N-terminal region. Interestingly, a disease-causing mutation in the RHOD domain impairs CEP41-microtubule interaction. Moreover, depletion of CEP41 inhibits cell proliferation and disrupts cell cycle progression, suggesting its potential involvement in cell cycle regulation. These insights into the cellular functions of CEP41 hold promise for unraveling the impact of its mutations in ciliopathies.


Subject(s)
Cell Proliferation , Microtubules , Humans , Microtubules/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Centrosome/metabolism , Retina/metabolism , Retina/pathology , Retina/abnormalities , Ciliopathies/metabolism , Ciliopathies/genetics , Ciliopathies/pathology , Cerebellum/metabolism , Cerebellum/abnormalities , Cerebellum/pathology , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Cilia/metabolism , Cilia/pathology , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Animals , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Eye Abnormalities/metabolism , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Protein Binding , Cell Cycle/genetics , HEK293 Cells
10.
J Cell Sci ; 137(4)2024 02 15.
Article in English | MEDLINE | ID: mdl-38415788

ABSTRACT

The primary cilium is an antenna-like projection from the plasma membrane that serves as a sensor of the extracellular environment and a crucial signaling hub. Primary cilia are generated in most mammalian cells, and their physiological significance is highlighted by the large number of severe developmental disorders or ciliopathies that occur when primary ciliogenesis is impaired. Primary ciliogenesis is a tightly regulated process, and a central early regulatory step is the removal of a key mother centriole capping protein, CP110 (also known as CCP110). This uncapping allows vesicles docked on the distal appendages of the mother centriole to fuse to form a ciliary vesicle, which is bent into a ciliary sheath as the microtubule-based axoneme grows and extends from the mother centriole. When the mother centriole migrates toward the plasma membrane, the ciliary sheath fuses with the plasma membrane to form the primary cilium. In this Review, we outline key early steps of primary ciliogenesis, focusing on several novel mechanisms for removal of CP110. We also highlight examples of ciliopathies caused by genetic variants that encode key proteins involved in the early steps of ciliogenesis.


Subject(s)
Axoneme , Ciliopathies , Animals , Cell Membrane , Centrioles , Ciliopathies/genetics , Cytoplasmic Vesicles , Mammals
11.
Development ; 150(8)2023 04 15.
Article in English | MEDLINE | ID: mdl-36971348

ABSTRACT

Primary cilia are nearly ubiquitous organelles that transduce molecular and mechanical signals. Although the basic structure of the cilium and the cadre of genes that contribute to ciliary formation and function (the ciliome) are believed to be evolutionarily conserved, the presentation of ciliopathies with narrow, tissue-specific phenotypes and distinct molecular readouts suggests that an unappreciated heterogeneity exists within this organelle. Here, we provide a searchable transcriptomic resource for a curated primary ciliome, detailing various subgroups of differentially expressed genes within the ciliome that display tissue and temporal specificity. Genes within the differentially expressed ciliome exhibited a lower level of functional constraint across species, suggesting organism and cell-specific function adaptation. The biological relevance of ciliary heterogeneity was functionally validated by using Cas9 gene-editing to disrupt ciliary genes that displayed dynamic gene expression profiles during osteogenic differentiation of multipotent neural crest cells. Collectively, this novel primary cilia-focused resource will allow researchers to explore longstanding questions related to how tissue and cell-type specific functions and ciliary heterogeneity may contribute to the range of phenotypes associated with ciliopathies.


Subject(s)
Ciliopathies , Osteogenesis , Humans , Cilia/genetics , Cilia/metabolism , Ciliopathies/genetics , Embryonic Development/genetics , Cell Differentiation/genetics
12.
Mol Cell Proteomics ; 23(1): 100701, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38122899

ABSTRACT

Alström syndrome (ALMS) is a very rare autosomal-recessive disorder, causing a broad range of clinical defects most notably retinal degeneration, type 2 diabetes, and truncal obesity. The ALMS1 gene encodes a complex and huge ∼0.5 MDa protein, which has hampered analysis in the past. The ALMS1 protein is localized to the centrioles and the basal body of cilia and is involved in signaling processes, for example, TGF-ß signaling. However, the exact molecular function of ALMS1 at the basal body remains elusive and controversial. We recently demonstrated that protein complex analysis utilizing endogenously tagged cells provides an excellent tool to investigate protein interactions of ciliary proteins. Here, CRISPR/Cas9-mediated endogenously tagged ALMS1 cells were used for affinity-based protein complex analysis. Centrosomal and microtubule-associated proteins were identified, which are potential regulators of ALMS1 function, such as the centrosomal protein 70 kDa (CEP70). Candidate proteins were further investigated in ALMS1-deficient hTERT-RPE1 cells. Loss of ALMS1 led to shortened cilia with no change in structural protein localization, for example, acetylated and É£-tubulin, Centrin-3, or the novel interactor CEP70. Conversely, reduction of CEP70 resulted in decreased ALMS1 at the ciliary basal body. Complex analysis of CEP70 revealed domain-specific ALMS1 interaction involving the TPR-containing C-terminal (TRP-CT) fragment of CEP70. In addition to ALMS1, several ciliary proteins, including CEP135, were found to specifically bind to the TPR-CT domain. Data are available via ProteomeXchange with the identifier PXD046401. Protein interactors identified in this study provide candidate lists that help to understand ALMS1 and CEP70 function in cilia-related protein modification, cell death, and disease-related mechanisms.


Subject(s)
Alstrom Syndrome , Diabetes Mellitus, Type 2 , Humans , Alstrom Syndrome/genetics , Alstrom Syndrome/metabolism , Cell Cycle Proteins/genetics , Microtubule-Associated Proteins/metabolism , Obesity , Tubulin
13.
Proc Natl Acad Sci U S A ; 120(22): e2219686120, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37216541

ABSTRACT

Polycystins (PKD2, PKD2L1, and PKD2L2) are members of the transient receptor potential family, which form ciliary ion channels. Most notably, PKD2 dysregulation in the kidney nephron cilia is associated with polycystic kidney disease, but the function of PKD2L1 in neurons is undefined. In this report, we develop animal models to track the expression and subcellular localization of PKD2L1 in the brain. We discover that PKD2L1 localizes and functions as a Ca2+ channel in the primary cilia of hippocampal neurons that apically radiate from the soma. Loss of PKD2L1 expression ablates primary ciliary maturation and attenuates neuronal high-frequency excitability, which precipitates seizure susceptibility and autism spectrum disorder-like behavior in mice. The disproportionate impairment of interneuron excitability suggests that circuit disinhibition underlies the neurophenotypic features of these mice. Our results identify PKD2L1 channels as regulators of hippocampal excitability and the neuronal primary cilia as organelle mediators of brain electrical signaling.


Subject(s)
Autism Spectrum Disorder , Cilia , Mice , Animals , Cilia/metabolism , Autism Spectrum Disorder/metabolism , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , Neurons/metabolism , Hippocampus/metabolism , Receptors, Cell Surface/metabolism , Calcium Channels/metabolism
14.
Semin Cell Dev Biol ; 133: 20-31, 2023 01 15.
Article in English | MEDLINE | ID: mdl-35351373

ABSTRACT

Ciliogenesis is a complex multistep process used to describe assembly of cilia and flagella. These organelles play essential roles in motility and signaling on the surface of cells. Cilia are built at the distal ends of centrioles through the formation of an axoneme that is surrounded by the ciliary membrane. As is the case in the biogenesis of other cellular organelles, regulators of membrane trafficking play essential roles in ciliogenesis, albeit with a unique feature that membranes are organized around microtubule-based structures. Membrane association with the distal end of the centriole is a critical initiating step for ciliogenesis. Studies of this process in different cell types suggests that a singular mechanism may not be utilized to initiate cilium assembly. In this review, we focus on recent insights into cilium biogenesis and the roles membrane trafficking regulators play in described ciliogenesis mechanisms with relevance to human disease.


Subject(s)
Axoneme , Centrioles , Humans , Centrioles/metabolism , Axoneme/metabolism , Cilia/metabolism , Microtubules/metabolism , Flagella
15.
Trends Genet ; 38(6): 524-528, 2022 06.
Article in English | MEDLINE | ID: mdl-35115176

ABSTRACT

The etiology and heterogeneity of idiopathic scoliosis (IS) are poorly understood. Studies using scoliotic zebrafish models have indicated a potential link between ciliary defects and scoliosis. They may further explain the onset of IS partially. However, it is necessary to further interpret the link between this progress and clinical medicine.


Subject(s)
Scoliosis , Zebrafish , Animals , Disease Models, Animal , Scoliosis/genetics , Spine , Zebrafish/genetics , Zebrafish Proteins
16.
Am J Hum Genet ; 109(10): 1828-1849, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36084634

ABSTRACT

Orofaciodigital syndrome (OFD) is a genetically heterogeneous ciliopathy characterized by anomalies of the oral cavity, face, and digits. We describe individuals with OFD from three unrelated families having bi-allelic loss-of-function variants in SCNM1 as the cause of their condition. SCNM1 encodes a protein recently shown to be a component of the human minor spliceosome. However, so far the effect of loss of SCNM1 function on human cells had not been assessed. Using a comparative transcriptome analysis between fibroblasts derived from an OFD-affected individual harboring SCNM1 mutations and control fibroblasts, we identified a set of genes with defective minor intron (U12) processing in the fibroblasts of the affected subject. These results were reproduced in SCNM1 knockout hTERT RPE-1 (RPE-1) cells engineered by CRISPR-Cas9-mediated editing and in SCNM1 siRNA-treated RPE-1 cultures. Notably, expression of TMEM107 and FAM92A encoding primary cilia and basal body proteins, respectively, and that of DERL2, ZC3H8, and C17orf75, were severely reduced in SCNM1-deficient cells. Primary fibroblasts containing SCNM1 mutations, as well as SCNM1 knockout and SCNM1 knockdown RPE-1 cells, were also found with abnormally elongated cilia. Conversely, cilia length and expression of SCNM1-regulated genes were restored in SCNM1-deficient fibroblasts following reintroduction of SCNM1 via retroviral delivery. Additionally, functional analysis in SCNM1-retrotransduced fibroblasts showed that SCNM1 is a positive mediator of Hedgehog (Hh) signaling. Our findings demonstrate that defective U12 intron splicing can lead to a typical ciliopathy such as OFD and reveal that primary cilia length and Hh signaling are regulated by the minor spliceosome through SCNM1 activity.


Subject(s)
Ciliopathies , Orofaciodigital Syndromes , Cilia/genetics , Cilia/metabolism , Ciliopathies/genetics , Hedgehog Proteins/metabolism , Humans , Introns/genetics , Mutation/genetics , Orofaciodigital Syndromes/genetics , RNA Splicing/genetics , RNA Splicing Factors/metabolism , RNA, Small Interfering/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism
17.
Am J Hum Genet ; 109(1): 136-156, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34890546

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD), characterized by progressive cyst formation/expansion, results in enlarged kidneys and often end stage kidney disease. ADPKD is genetically heterogeneous; PKD1 and PKD2 are the common loci (∼78% and ∼15% of families) and GANAB, DNAJB11, and ALG9 are minor genes. PKD is a ciliary-associated disease, a ciliopathy, and many syndromic ciliopathies have a PKD phenotype. In a multi-cohort/-site collaboration, we screened ADPKD-diagnosed families that were naive to genetic testing (n = 834) or for whom no PKD1 and PKD2 pathogenic variants had been identified (n = 381) with a PKD targeted next-generation sequencing panel (tNGS; n = 1,186) or whole-exome sequencing (WES; n = 29). We identified monoallelic IFT140 loss-of-function (LoF) variants in 12 multiplex families and 26 singletons (1.9% of naive families). IFT140 is a core component of the intraflagellar transport-complex A, responsible for retrograde ciliary trafficking and ciliary entry of membrane proteins; bi-allelic IFT140 variants cause the syndromic ciliopathy, short-rib thoracic dysplasia (SRTD9). The distinctive monoallelic phenotype is mild PKD with large cysts, limited kidney insufficiency, and few liver cysts. Analyses of the cystic kidney disease probands of Genomics England 100K showed that 2.1% had IFT140 LoF variants. Analysis of the UK Biobank cystic kidney disease group showed probands with IFT140 LoF variants as the third most common group, after PKD1 and PKD2. The proximity of IFT140 to PKD1 (∼0.5 Mb) in 16p13.3 can cause diagnostic confusion, and PKD1 variants could modify the IFT140 phenotype. Importantly, our studies link a ciliary structural protein to the ADPKD spectrum.


Subject(s)
Alleles , Carrier Proteins , Genetic Predisposition to Disease , Mutation , Polycystic Kidney, Autosomal Dominant/genetics , Adult , Aged , Amino Acid Substitution , Biological Specimen Banks , Cilia/pathology , DNA Copy Number Variations , Female , Genetic Association Studies , Genetic Testing , High-Throughput Nucleotide Sequencing , Humans , Kidney Function Tests , Male , Middle Aged , Pedigree , Phenotype , Polycystic Kidney, Autosomal Dominant/diagnosis , Sequence Analysis, DNA , United Kingdom , Exome Sequencing
18.
Am J Hum Genet ; 109(5): 928-943, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35397207

ABSTRACT

Organ fibrosis is a shared endpoint of many diseases, yet underlying mechanisms are not well understood. Several pathways governed by the primary cilium, a sensory antenna present on most vertebrate cells, have been linked with fibrosis. Ciliopathies usually start early in life and represent a considerable disease burden. We performed massively parallel sequencing by using cohorts of genetically unsolved individuals with unexplained liver and kidney failure and correlated this with clinical, imaging, and histopathological analyses. Mechanistic studies were conducted with a vertebrate model and primary cells. We detected bi-allelic deleterious variants in TULP3, encoding a critical adaptor protein for ciliary trafficking, in a total of 15 mostly adult individuals, originating from eight unrelated families, with progressive degenerative liver fibrosis, fibrocystic kidney disease, and hypertrophic cardiomyopathy with atypical fibrotic patterns on histopathology. We recapitulated the human phenotype in adult zebrafish and confirmed disruption of critical ciliary cargo composition in several primary cell lines derived from affected individuals. Further, we show interaction between TULP3 and the nuclear deacetylase SIRT1, with roles in DNA damage repair and fibrosis, and report increased DNA damage ex vivo. Transcriptomic studies demonstrated upregulation of profibrotic pathways with gene clusters for hypertrophic cardiomyopathy and WNT and TGF-ß signaling. These findings identify variants in TULP3 as a monogenic cause for progressive degenerative disease of major organs in which affected individuals benefit from early detection and improved clinical management. Elucidation of mechanisms crucial for DNA damage repair and tissue maintenance will guide novel therapeutic avenues for this and similar genetic and non-genomic diseases.


Subject(s)
Cardiomyopathy, Hypertrophic , Cilia , Adult , Animals , Cardiomyopathy, Hypertrophic/metabolism , Child , Cilia/genetics , Cilia/metabolism , Fibrosis , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kidney , Liver , Mutation/genetics , Zebrafish/genetics
19.
J Cell Sci ; 136(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-36268591

ABSTRACT

The primary cilium is a sensory organelle, receiving signals from the external environment and relaying them into the cell. Mutations in proteins required for transport in the primary cilium result in ciliopathies, a group of genetic disorders that commonly lead to the malformation of organs such as the kidney, liver and eyes and skeletal dysplasias. The motor proteins dynein-2 and kinesin-2 mediate retrograde and anterograde transport, respectively, in the cilium. WDR34 (also known as DYNC2I2), a dynein-2 intermediate chain, is required for the maintenance of cilia function. Here, we investigated WDR34 mutations identified in Jeune syndrome, short-rib polydactyly syndrome and asphyxiating thoracic dysplasia patients. There is a poor correlation between genotype and phenotype in these cases, making diagnosis and treatment highly complex. We set out to define the biological impacts on cilia formation and function of WDR34 mutations by stably expressing the mutant proteins in WDR34-knockout cells. WDR34 mutations led to different spectrums of phenotypes. Quantitative proteomics demonstrated changes in dynein-2 assembly, whereas initiation and extension of the axoneme, localization of intraflagellar transport complex-B proteins, transition zone integrity and Hedgehog signalling were also affected.


Subject(s)
Dyneins , Ellis-Van Creveld Syndrome , Humans , Dyneins/genetics , Dyneins/metabolism , Carrier Proteins/metabolism , Hedgehog Proteins/metabolism , Ellis-Van Creveld Syndrome/genetics , Ellis-Van Creveld Syndrome/metabolism , Cilia/genetics , Cilia/metabolism , Mutation/genetics
20.
J Cell Sci ; 136(3)2023 02 01.
Article in English | MEDLINE | ID: mdl-36606424

ABSTRACT

The primary cilium is a microtubule-based organelle that serves as a hub for many signaling pathways. It functions as part of the centrosome or cilium complex, which also contains the basal body and the centriolar satellites. Little is known about the mechanisms by which the microtubule-based ciliary axoneme is assembled with a proper length and structure, particularly in terms of the activity of microtubule-associated proteins (MAPs) and the crosstalk between the different compartments of the centrosome or cilium complex. Here, we analyzed CCDC66, a MAP implicated in cilium biogenesis and ciliopathies. Live-cell imaging revealed that CCDC66 compartmentalizes between centrosomes, centriolar satellites, and the ciliary axoneme and tip during cilium biogenesis. CCDC66 depletion in human cells causes defects in cilium assembly, length and morphology. Notably, CCDC66 interacts with the ciliopathy-linked MAPs CEP104 and CSPP1, and regulates axonemal length and Hedgehog pathway activation. Moreover, CCDC66 is required for the basal body recruitment of transition zone proteins and intraflagellar transport B (IFT-B) machinery. Overall, our results establish CCDC66 as a multifaceted regulator of the primary cilium and provide insight into how ciliary MAPs and subcompartments cooperate to ensure assembly of functional cilia.


Subject(s)
Axoneme , Cilia , Humans , Cilia/metabolism , Axoneme/metabolism , Hedgehog Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Centrioles/metabolism , Eye Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL