Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Cell Sci ; 136(5)2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35403186

RESUMO

Primary cilia are microtubule-based sensory organelles whose assembly and function rely on the conserved bidirectional intraflagellar transport (IFT) system, which is powered by anterograde kinesin-2 and retrograde cytoplasmic dynein-2 motors. Nematodes additionally employ a cell-type-specific kinesin-3 motor, KLP-6, which moves within cilia independently of IFT and regulates ciliary content and function. Here, we provide evidence that a KLP-6 homolog, KIF13B, undergoes bursts of bidirectional movement within primary cilia of cultured immortalized human retinal pigment epithelial (hTERT-RPE1) cells. Anterograde and retrograde intraciliary velocities of KIF13B were similar to those of IFT (as assayed using IFT172-eGFP), but intraciliary movement of KIF13B required its own motor domain and appeared to be cell-type specific. Our work provides the first demonstration of motor-driven, intraciliary movement by a vertebrate kinesin other than kinesin-2 motors.


Assuntos
Cílios , Cinesinas , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Transporte Biológico , Cílios/metabolismo , Proteínas do Citoesqueleto/metabolismo , Flagelos/metabolismo , Humanos , Cinesinas/genética , Microtúbulos
2.
Sci Rep ; 9(1): 16519, 2019 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-31712586

RESUMO

Intraflagellar transport (IFT) is essential for the formation and function of the microtubule-based primary cilium, which acts as a sensory and signalling device at the cell surface. Consisting of IFT-A/B and BBSome cargo adaptors that associate with molecular motors, IFT transports protein into (anterograde IFT) and out of (retrograde IFT) the cilium. In this study, we identify the mostly uncharacterised ERICH3 protein as a component of the mammalian primary cilium. Loss of ERICH3 causes abnormally short cilia and results in the accumulation of IFT-A/B proteins at the ciliary tip, together with reduced ciliary levels of retrograde transport regulators, ARL13B, INPP5E and BBS5. We also show that ERICH3 ciliary localisations require ARL13B and BBSome components. Finally, ERICH3 loss causes positive (Smoothened) and negative (GPR161) regulators of sonic hedgehog signaling (Shh) to accumulate at abnormally high levels in the cilia of pathway-stimulated cells. Together, these findings identify ERICH3 as a novel component of the primary cilium that regulates cilium length and the ciliary levels of Shh signaling molecules. We propose that ERICH3 functions within retrograde IFT-associated pathways to remove signaling proteins from cilia.


Assuntos
Biomarcadores , Cílios/metabolismo , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Linhagem Celular , Imunofluorescência , Genes Reporter , Humanos , Ligação Proteica , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
3.
EMBO Rep ; 20(10): e47625, 2019 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-31432619

RESUMO

Rab and Arl guanine nucleotide-binding (G) proteins regulate trafficking pathways essential for the formation, function and composition of primary cilia, which are sensory devices associated with Sonic hedgehog (Shh) signalling and ciliopathies. Here, using mammalian cells and zebrafish, we uncover ciliary functions for Rab35, a multitasking G protein with endocytic recycling, actin remodelling and cytokinesis roles. Rab35 loss via siRNAs, morpholinos or knockout reduces cilium length in mammalian cells and the zebrafish left-right organiser (Kupffer's vesicle) and causes motile cilia-associated left-right asymmetry defects. Consistent with these observations, GFP-Rab35 localises to cilia, as do GEF (DENND1B) and GAP (TBC1D10A) Rab35 regulators, which also regulate ciliary length and Rab35 ciliary localisation. Mammalian Rab35 also controls the ciliary membrane levels of Shh signalling regulators, promoting ciliary targeting of Smoothened, limiting ciliary accumulation of Arl13b and the inositol polyphosphate 5-phosphatase (INPP5E). Rab35 additionally regulates ciliary PI(4,5)P2 levels and interacts with Arl13b. Together, our findings demonstrate roles for Rab35 in regulating cilium length, function and membrane composition and implicate Rab35 in pathways controlling the ciliary levels of Shh signal regulators.


Assuntos
Cílios/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Padronização Corporal , Linhagem Celular , Células HEK293 , Humanos , Membranas/metabolismo , Camundongos , Modelos Biológicos , Células NIH 3T3 , Nucleotídeos/metabolismo , Ligação Proteica , Transporte Proteico , Telomerase/metabolismo
4.
Small GTPases ; 9(1-2): 76-94, 2018 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-29072526

RESUMO

Cilia are microtubule-based organelles extending from a basal body at the surface of eukaryotic cells. Cilia regulate cell and fluid motility, sensation and developmental signaling, and ciliary defects cause human diseases (ciliopathies) affecting the formation and function of many tissues and organs. Over the past decade, various Rab and Rab-like membrane trafficking proteins have been shown to regulate cilia-related processes such as basal body maturation, ciliary axoneme extension, intraflagellar transport and ciliary signaling. In this review, we provide a comprehensive overview of Rab protein ciliary associations, drawing on findings from multiple model systems, including mammalian cell culture, mice, zebrafish, C. elegans, trypanosomes, and green algae. We also discuss several emerging mechanistic themes related to ciliary Rab cascades and functional redundancy.


Assuntos
Cílios/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Humanos , Transporte Proteico
5.
Genetics ; 207(4): 1423-1440, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29021280

RESUMO

Motile and immotile (or primary) cilia are microtubule-based structures that mediate multiple cellular functions, including the transduction of environmental cues, developmental signaling, cellular motility, and modulation of fluid flow. Although their core architectures are similar, motile and primary cilia exhibit marked structural differences that underlie distinct functional properties. However, the extent to which ciliogenesis mechanisms are shared between these different cilia types is not fully described. Here, we report that the atypical MAP kinase MAPK15 (ERK7/8), implicated in the formation of vertebrate motile cilia, also regulates the formation of primary cilia in Caenorhabditis elegans sensory neurons and human cells. We find that MAPK15 localizes to a basal body subdomain with the ciliopathy protein BBS7 and to cell-cell junctions. MAPK15 also regulates the localization of ciliary proteins involved in cilium structure, transport, and signaling. Our results describe a primary cilia-related role for this poorly studied member of the MAPK family in vivo, and indicate a broad requirement for MAPK15 in the formation of multiple ciliary classes across species.


Assuntos
Caenorhabditis elegans/genética , Cílios/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Células Receptoras Sensoriais/metabolismo , Animais , Caenorhabditis elegans/crescimento & desenvolvimento , Movimento Celular/genética , Humanos , Microtúbulos , Transporte Proteico/genética , Transdução de Sinais
6.
Dev Cell ; 37(5): 395-6, 2016 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-27270038

RESUMO

Cilium formation depends on intraflagellar transport trains that move bidirectionally along ciliary microtubules. Reporting in Science, Stepanek and Pigino (2016) employ correlative light and electron microscopy in algae to determine the ultrastructure of anterograde and retrograde trains and discover that these trains avoid collision by running on B- and A-tubules, respectively.


Assuntos
Cílios/metabolismo , Animais , Transporte Biológico , Caenorhabditis elegans/metabolismo , Chlamydomonas reinhardtii/metabolismo , Flagelos/metabolismo
7.
Nat Cell Biol ; 18(1): 122-31, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26595381

RESUMO

The transition zone (TZ) ciliary subcompartment is thought to control cilium composition and signalling by facilitating a protein diffusion barrier at the ciliary base. TZ defects cause ciliopathies such as Meckel-Gruber syndrome (MKS), nephronophthisis (NPHP) and Joubert syndrome (JBTS). However, the molecular composition and mechanisms underpinning TZ organization and barrier regulation are poorly understood. To uncover candidate TZ genes, we employed bioinformatics (coexpression and co-evolution) and identified TMEM107 as a TZ protein mutated in oral-facial-digital syndrome and JBTS patients. Mechanistic studies in Caenorhabditis elegans showed that TMEM-107 controls ciliary composition and functions redundantly with NPHP-4 to regulate cilium integrity, TZ docking and assembly of membrane to microtubule Y-link connectors. Furthermore, nematode TMEM-107 occupies an intermediate layer of the TZ-localized MKS module by organizing recruitment of the ciliopathy proteins MKS-1, TMEM-231 (JBTS20) and JBTS-14 (TMEM237). Finally, MKS module membrane proteins are immobile and super-resolution microscopy in worms and mammalian cells reveals periodic localizations within the TZ. This work expands the MKS module of ciliopathy-causing TZ proteins associated with diffusion barrier formation and provides insight into TZ subdomain architecture.


Assuntos
Cerebelo/anormalidades , Cílios/metabolismo , Proteínas de Membrana/metabolismo , Retina/anormalidades , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Cerebelo/metabolismo , Anormalidades do Olho/genética , Anormalidades do Olho/metabolismo , Humanos , Doenças Renais Císticas/genética , Doenças Renais Císticas/metabolismo , Proteínas de Membrana/genética , Retina/metabolismo
8.
Genome Biol ; 16: 293, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26714646

RESUMO

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.


Assuntos
Corpos Basais/metabolismo , Cerebelo/anormalidades , Proteínas Associadas aos Microtúbulos/genética , Mutação , Retina/anormalidades , Fatores de Ribosilação do ADP/metabolismo , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Adenosina Trifosfatases/metabolismo , Adulto , Animais , Corpos Basais/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Células Cultivadas , Cerebelo/patologia , Criança , Pré-Escolar , Cílios/genética , Cílios/patologia , Exoma , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Feminino , Humanos , Katanina , Doenças Renais Císticas/genética , Doenças Renais Císticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Linhagem , Ligação Proteica , Retina/patologia
9.
Mol Biol Cell ; 25(4): 495-507, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24356449

RESUMO

In differentiated human cells, primary cilia fulfill essential functions in converting mechanical or chemical stimuli into intracellular signals. Formation and maintenance of cilia require multiple functions associated with the centriole-derived basal body, from which axonemal microtubules grow and which assembles a gate to maintain the specific ciliary proteome. Here we characterize the function of a novel centriolar satellite protein, synovial sarcoma X breakpoint-interacting protein 2 (SSX2IP), in the assembly of primary cilia. We show that SSX2IP localizes to the basal body of primary cilia in human and murine ciliated cells. Using small interfering RNA knockdown in human cells, we demonstrate the importance of SSX2IP for efficient recruitment of the ciliopathy-associated satellite protein Cep290 to both satellites and the basal body. Cep290 takes a central role in gating proteins to the ciliary compartment. Consistent with that, loss of SSX2IP drastically reduces entry of the BBSome, which functions to target membrane proteins to primary cilia, and interferes with efficient accumulation of the key regulator of ciliary membrane protein targeting, Rab8. Finally, we show that SSX2IP knockdown limits targeting of the ciliary membrane protein and BBSome cargo, somatostatin receptor 3, and significantly reduces axoneme length. Our data establish SSX2IP as a novel targeting factor for ciliary membrane proteins cooperating with Cep290, the BBSome, and Rab8.


Assuntos
Antígenos de Neoplasias/metabolismo , Proteínas de Transporte/metabolismo , Centríolos/metabolismo , Cílios/metabolismo , Células Epiteliais/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Antígenos de Neoplasias/genética , Axonema/metabolismo , Axonema/ultraestrutura , Corpos Basais/metabolismo , Corpos Basais/ultraestrutura , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Linhagem Celular , Centríolos/ultraestrutura , Cílios/ultraestrutura , Proteínas do Citoesqueleto , Células Epiteliais/ultraestrutura , Regulação da Expressão Gênica , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos , Células NIH 3T3 , Proteínas de Neoplasias/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/ultraestrutura , Transdução de Sinais , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
10.
J Cell Biol ; 200(4): 505-22, 2013 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-23400999

RESUMO

Despite the critical contributions of cilia to embryonic development and human health, key regulators of cilia formation await identification. In this paper, a functional RNA interference-based screen linked 30 novel protein kinases with ciliogenesis. Of them, we have studied the role of the microtubule (MT)-associated protein/MT affinity regulating kinase 4 (MARK4) in depth. MARK4 associated with the basal body and ciliary axoneme in human and murine cell lines. Ultrastructural and functional analyses established that MARK4 kinase activity was required for initiation of axoneme extension. We identified the mother centriolar protein ODF2 as an interaction partner of MARK4 and showed that ODF2 localization to the centriole partially depended on MARK4. Our data indicated that, upon MARK4 or ODF2 knockdown, the ciliary program arrested before the complete removal of the CP110-Cep97 inhibitory complex from the mother centriole, suggesting that these proteins act at this level of axonemal extension. We propose that MARK4 is a critical positive regulator of early steps in ciliogenesis.


Assuntos
Axonema/metabolismo , Cílios/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Axonema/ultraestrutura , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Cílios/ultraestrutura , Células HEK293 , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/fisiologia , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Células NIH 3T3 , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/genética , Interferência de RNA
11.
J Cell Biol ; 199(7): 1083-101, 2012 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-23253480

RESUMO

Cilia formation is a multi-step process that starts with the docking of a vesicle at the distal part of the mother centriole. This step marks the conversion of the mother centriole into the basal body, from which axonemal microtubules extend to form the ciliary compartment. How vesicles are stably attached to the mother centriole to initiate ciliary membrane biogenesis is unknown. Here, we investigate the molecular role of the mother centriolar component Cep164 in ciliogenesis. We show that Cep164 was indispensable for the docking of vesicles at the mother centriole. Using biochemical and functional assays, we identified the components of the vesicular transport machinery, the GEF Rabin8 and the GTPase Rab8, as interacting partners of Cep164. We propose that Cep164 is targeted to the apical domain of the mother centriole to provide the molecular link between the mother centriole and the membrane biogenesis machinery that initiates cilia formation.


Assuntos
Centríolos/metabolismo , Cílios/fisiologia , Proteínas dos Microtúbulos/fisiologia , Vesículas Transportadoras/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Autoantígenos/metabolismo , Sítios de Ligação , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Cílios/metabolismo , Proteínas do Citoesqueleto , Expressão Gênica , Quinases do Centro Germinativo , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Microtúbulos/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Supressoras de Tumor/metabolismo , Técnicas do Sistema de Duplo-Híbrido
12.
Biochem Biophys Res Commun ; 364(3): 483-7, 2007 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-17961512

RESUMO

Interleukin 6 (IL6)-type cytokines are major regulators of inflammation and thereby contribute to the neuropathology and pathophysiology associated with inflammation of the central nervous system (CNS). Furthermore, astrocyte development which is a key process in the development of the CNS is also controlled by cytokines of the IL6-family. Interleukin 27 (IL27) is a recently identified member of this family and has been implicated in the inhibition of TH17 T-cell-responses. Here we show that IL27 and the HHV8 encoded viral IL6 (vIL6) induce C6 glioma cells to differentiate into an astrocyte-like state. Cytokine stimulation led to STAT-factor phosphorylation and consequently to protein expression of the astrocyte marker glial fibrillary acidic protein (GFAP). These data could be confirmed by GFAP-immunostaining of stimulated cells. Taken together, IL27 and vIL6 can be considered as new astrocyte-inducing cytokines of the brain.


Assuntos
Astrócitos/citologia , Astrócitos/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Interleucina-17/imunologia , Interleucina-6/imunologia , Animais , Diferenciação Celular/imunologia , Linhagem Celular , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA