Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(15): e2109934119, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35394880

RESUMO

Photoreceptor connecting cilium (CC) is structurally analogous to the transition zone (TZ) of primary cilia and gates the molecular trafficking between the inner and the outer segment (OS). Retinal dystrophies with underlying CC defects are manifested in a broad array of syndromic conditions known as ciliopathies as well as nonsyndromic retinal degenerations. Despite extensive studies, many questions remain in the mechanism of protein trafficking across the photoreceptor CC. Here, we genetically inactivated mouse Tmem138, a gene encoding a putative transmembrane protein localized to the ciliary TZ and linked to ciliopathies. Germline deletion of Tmem138 abolished OS morphogenesis, followed by rapid photoreceptor degeneration. Tmem138 was found localized to the photoreceptor CC and was required for localization of Ahi1 to the distal subdomain of the CC. Among the examined set of OS proteins, rhodopsin was mislocalized throughout the mutant cell body prior to OS morphogenesis. Ablation of Tmem138 in mature rods recapitulated the molecular changes in the germline mutants, causing failure of disc renewal and disintegration of the OS. Furthermore, Tmem138 interacts reciprocally with rhodopsin and a related protein Tmem231, and the ciliary localization of the latter was also altered in the mutant photoreceptors. Taken together, these results suggest a crucial role of Tmem138 in the functional organization of the CC, which is essential for rhodopsin localization and OS biogenesis.


Assuntos
Ciliopatias , Degeneração Retiniana , Cílios/metabolismo , Ciliopatias/metabolismo , Humanos , Proteínas de Membrana , Cílio Conector dos Fotorreceptores , Degeneração Retiniana/metabolismo , Rodopsina/genética , Rodopsina/metabolismo
2.
Acta Neuropathol Commun ; 9(1): 17, 2021 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33509294

RESUMO

Accumulation of misfolded host proteins is central to neuropathogenesis of numerous human brain diseases including prion and prion-like diseases. Neurons of retina are also affected by these diseases. Previously, our group and others found that prion-induced retinal damage to photoreceptor cells in mice and humans resembled pathology of human retinitis pigmentosa caused by mutations in retinal proteins. Here, using confocal, epifluorescent and electron microscopy we followed deposition of disease-associated prion protein (PrPSc) and its association with damage to critical retinal structures following intracerebral prion inoculation. The earliest time and place of retinal PrPSc deposition was 67 days post-inoculation (dpi) on the inner segment (IS) of cone photoreceptors. At 104 and 118 dpi, PrPSc was associated with the base of cilia and swollen cone inner segments, suggesting ciliopathy as a pathogenic mechanism. By 118 dpi, PrPSc was deposited in both rods and cones which showed rootlet damage in the IS, and photoreceptor cell death was indicated by thinning of the outer nuclear layer. In the outer plexiform layer (OPL) in uninfected mice, normal host PrP (PrPC) was mainly associated with cone bipolar cell processes, but in infected mice, at 118 dpi, PrPSc was detected on cone and rod bipolar cell dendrites extending into ribbon synapses. Loss of ribbon synapses in cone pedicles and rod spherules in the OPL was observed to precede destruction of most rods and cones over the next 2-3 weeks. However, bipolar cells and horizontal cells were less damaged, indicating high selectivity among neurons for injury by prions. PrPSc deposition in cone and rod inner segments and on the bipolar cell processes participating in ribbon synapses appear to be critical early events leading to damage and death of photoreceptors after prion infection. These mechanisms may also occur in human retinitis pigmentosa and prion-like diseases, such as AD.


Assuntos
Cílio Conector dos Fotorreceptores/metabolismo , Proteínas PrPSc/metabolismo , Células Bipolares da Retina/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Segmento Interno das Células Fotorreceptoras da Retina/metabolismo , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Animais , Morte Celular , Progressão da Doença , Camundongos , Microscopia Confocal , Microscopia Eletrônica , Microscopia de Fluorescência , Cílio Conector dos Fotorreceptores/patologia , Cílio Conector dos Fotorreceptores/ultraestrutura , Proteínas PrPSc/administração & dosagem , Células Bipolares da Retina/patologia , Células Bipolares da Retina/ultraestrutura , Células Fotorreceptoras Retinianas Cones/patologia , Células Fotorreceptoras Retinianas Cones/ultraestrutura , Segmento Interno das Células Fotorreceptoras da Retina/patologia , Segmento Interno das Células Fotorreceptoras da Retina/ultraestrutura , Segmento Externo das Células Fotorreceptoras da Retina/patologia , Segmento Externo das Células Fotorreceptoras da Retina/ultraestrutura , Células Fotorreceptoras Retinianas Bastonetes/patologia , Células Fotorreceptoras Retinianas Bastonetes/ultraestrutura , Scrapie/metabolismo , Scrapie/patologia
3.
Dev Cell ; 53(3): 287-299.e5, 2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32275885

RESUMO

Retinopathy of prematurity (ROP) is a leading cause of childhood blindness. However, the pathogenesis and molecular mechanisms underlying ROP remain elusive. Herein, using the oxygen-induced retinopathy (OIR) mouse model of ROP, we demonstrate that disassembly of photoreceptor connecting cilia is an early event in response to oxygen changes. Histone deacetylase 6 (HDAC6) is upregulated in the retina of OIR mice and accumulates in the transition zone of connecting cilia. We also show that in response to oxygen changes, apoptosis signal-regulating kinase 1 (ASK1) is activated and phosphorylates HDAC6, blocking its ubiquitination by von Hippel-Lindau and subsequent degradation by the proteasome. Moreover, depletion of HDAC6 or inhibition of the ASK1/HDAC6 axis protects mice from oxygen-change-induced pathological changes of photoreceptors. These findings reveal a critical role for ASK1/HDAC6-mediated connecting cilium disassembly in the OIR mouse model of ROP and suggest a potential value of ASK1/HDAC6-targeted agents for prevention of this disease.


Assuntos
Cílios/patologia , Desacetilase 6 de Histona/antagonistas & inibidores , MAP Quinase Quinase Quinase 5/metabolismo , Cílio Conector dos Fotorreceptores/patologia , Proteólise , Retinopatia da Prematuridade/patologia , Ubiquitinação , Animais , Cílios/metabolismo , Feminino , Desacetilase 6 de Histona/genética , Desacetilase 6 de Histona/metabolismo , MAP Quinase Quinase Quinase 5/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/toxicidade , Fosforilação , Cílio Conector dos Fotorreceptores/metabolismo , Retinopatia da Prematuridade/etiologia , Retinopatia da Prematuridade/metabolismo
4.
Proc Natl Acad Sci U S A ; 116(47): 23562-23572, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31690665

RESUMO

Primary cilia carry out numerous signaling and sensory functions, and defects in them, "ciliopathies," cause a range of symptoms, including blindness. Understanding of their nanometer-scale ciliary substructures and their disruptions in ciliopathies has been hindered by limitations of conventional microscopic techniques. We have combined cryoelectron tomography, enhanced by subtomogram averaging, with superresolution stochastic optical reconstruction microscopy (STORM) to define subdomains within the light-sensing rod sensory cilium of mouse retinas and reveal previously unknown substructures formed by resident proteins. Domains are demarcated by structural features such as the axoneme and its connections to the ciliary membrane, and are correlated with molecular markers of subcompartments, including the lumen and walls of the axoneme, the membrane glycocalyx, and the intervening cytoplasm. Within this framework, we report spatial distributions of key proteins in wild-type (WT) mice and the effects on them of genetic deficiencies in 3 models of Bardet-Biedl syndrome.


Assuntos
Síndrome de Bardet-Biedl/patologia , Cílios/ultraestrutura , Microscopia Crioeletrônica/métodos , Tomografia com Microscopia Eletrônica/métodos , Microscopia de Fluorescência/métodos , Nanotecnologia/métodos , Cílio Conector dos Fotorreceptores/ultraestrutura , Segmento Externo da Célula Bastonete/ultraestrutura , Imagem Individual de Molécula/métodos , Animais , Axonema/química , Axonema/ultraestrutura , Centríolos/ultraestrutura , Modelos Animais de Doenças , Proteínas do Olho/análise , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/análise , Microtúbulos/ultraestrutura , Complexos Multiproteicos , Proteínas Musculares/análise , Cílio Conector dos Fotorreceptores/química , Proteínas Qa-SNARE/análise , Proteínas Supressoras de Tumor/análise
5.
J Cell Biol ; 217(8): 2851-2865, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-29899041

RESUMO

Photoreceptor-specific ciliopathies often affect a structure that is considered functionally homologous to the ciliary transition zone (TZ) called the connecting cilium (CC). However, it is unclear how mutations in certain ciliary genes disrupt the photoreceptor CC without impacting the primary cilia systemically. By applying stochastic optical reconstruction microscopy technology in different genetic models, we show that the CC can be partitioned into two regions: the proximal CC (PCC), which is homologous to the TZ of primary cilia, and the distal CC (DCC), a photoreceptor-specific extension of the ciliary TZ. This specialized distal zone of the CC in photoreceptors is maintained by SPATA7, which interacts with other photoreceptor-specific ciliary proteins such as RPGR and RPGRIP1. The absence of Spata7 results in the mislocalization of DCC proteins without affecting the PCC protein complexes. This collapse results in destabilization of the axonemal microtubules, which consequently results in photoreceptor degeneration. These data provide a novel mechanism to explain how genetic disruption of ubiquitously present ciliary proteins exerts tissue-specific ciliopathy phenotypes.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Cílio Conector dos Fotorreceptores/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/análise , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Antígenos de Neoplasias , Proteínas de Transporte/análise , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas do Olho/análise , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia , Cílio Conector dos Fotorreceptores/ultraestrutura , Transporte Proteico/genética
6.
J Neurosci ; 38(20): 4708-4723, 2018 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-29669747

RESUMO

Microglia are the resident immune cells of the CNS, and their response to infection, injury and disease is well documented. More recently, microglia have been shown to play a role in normal CNS development, with the fractalkine-Cx3cr1 signaling pathway of particular importance. This work describes the interaction between the light-sensitive photoreceptors and microglia during eye opening, a time of postnatal photoreceptor maturation. Genetic removal of Cx3cr1 (Cx3cr1GFP/GFP ) led to an early retinal dysfunction soon after eye opening [postnatal day 17 (P17)] and cone photoreceptor loss (P30 onward) in mice of either sex. This dysfunction occurred at a time when fractalkine expression was predominantly outer retinal, when there was an increased microglial presence near the photoreceptor layer and increased microglial-cone photoreceptor contacts. Photoreceptor maturation and outer segment elongation was coincident with increased opsin photopigment expression in wild-type retina, while this was aberrant in the Cx3cr1GFP/GFP retina and outer segment length was reduced. A beadchip array highlighted Cx3cr1 regulation of genes involved in the photoreceptor cilium, a key structure that is important for outer segment elongation. This was confirmed with quantitative PCR with specific cilium-related genes, Rpgr and Rpgrip1, downregulated at eye opening (P14). While the overall cilium structure was unaffected, expression of Rpgr, Rpgrip1, and centrin were restricted to more proximal regions of the transitional zone. This study highlighted a novel role for microglia in postnatal neuronal development within the retina, with loss of fractalkine-Cx3cr1 signaling leading to an altered distribution of cilium proteins, failure of outer segment elongation and ultimately cone photoreceptor loss.SIGNIFICANCE STATEMENT Microglia are involved in CNS development and disease. This work highlights the role of microglia in postnatal development of the light-detecting photoreceptor neurons within the mouse retina. Loss of the microglial Cx3cr1 signaling pathway resulted in specific alterations in the cilium, a key structure in photoreceptor outer segment elongation. The distribution of key components of the cilium transitional zone, Rpgr, Rpgrip1, and centrin, were altered in retinae lacking Cx3cr1 with reduced outer segment length and cone photoreceptor death observed at later postnatal ages. This work identifies a novel role for microglia in the postnatal maturation of retinal photoreceptors.


Assuntos
Receptor 1 de Quimiocina CX3C/fisiologia , Células Fotorreceptoras de Vertebrados/fisiologia , Retina/crescimento & desenvolvimento , Retina/fisiologia , Transdução de Sinais/fisiologia , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Proteínas do Citoesqueleto , Olho/crescimento & desenvolvimento , Proteínas do Olho/genética , Proteínas do Olho/fisiologia , Feminino , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microglia , Cílio Conector dos Fotorreceptores/fisiologia , Proteínas/genética , Proteínas/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Segmento Externo das Células Fotorreceptoras da Retina/fisiologia
7.
Br J Ophthalmol ; 99(12): 1725-31, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26294103

RESUMO

BACKGROUND/AIM: We have noted a phenotype of early-onset retinal dystrophy with macular staphyloma but without high myopia. The aim of this study is to report the underlying genetic mutations and the subcellular localisation of the gene product in the retina. METHODS: Retrospective case series (2012-2015); immunohistochemical analyses of mammalian retina for in situ protein localisation. RESULTS: All three probands were first noted to have decreased vision at 3-6 years old which worsened over time. At ages 39, 37 and 12 years old, all had similar retinal findings: dystrophic changes (retinal pigment epithelium mottling, vessel narrowing), macular staphyloma (despite only mild myopia or high hyperopia), and non-recordable electroretinography. All harboured homozygous mutations in C21orf2, a gene recently suggested to be associated with retinal dystrophy but of unknown function. Two had a frameshift deletion c.436_466del (p.Glu146Serfs*6). The third had a missense mutation affecting a highly conserved residue (p.Cys61Tyr) and was short (below the 3rd percentile) and obese (50th percentile for weight despite short stature). Immunohistochemical studies in human, pig and mouse retinas localised C21orf2 protein to the ciliary structures of the photoreceptor cell (the daughter basal body, the centriole adjacent to the basal body, and the connecting cilium). CONCLUSIONS: This retinal dystrophy phenotype is caused by recessive mutations in C21orf2 and can be considered a retinal ciliopathy as C21orf2 encodes a protein that localises to photoreceptor ciliary structures. The short stature and obesity noted in the youngest girl suggest that for some patients biallelic C21orf2 mutations may result in syndromic ciliopathy.


Assuntos
Mutação da Fase de Leitura/genética , Genes Recessivos , Mutação de Sentido Incorreto/genética , Cílio Conector dos Fotorreceptores/metabolismo , Proteínas/genética , Retina/patologia , Distrofias Retinianas/genética , Adulto , Sequência de Aminoácidos , Animais , Western Blotting , Criança , Consanguinidade , Proteínas do Citoesqueleto , Dilatação Patológica , Eletrorretinografia , Feminino , Humanos , Imageamento por Ressonância Magnética , Camundongos , Dados de Sequência Molecular , Linhagem , Reação em Cadeia da Polimerase , Proteínas/metabolismo , Estudos Retrospectivos , Sus scrofa , Doadores de Tecidos , Tomografia de Coerência Óptica
8.
Biochem Biophys Res Commun ; 465(4): 651-7, 2015 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-26188096

RESUMO

Proteomic analysis of the mouse photoreceptor sensory cilium identified a set of cilia proteins, including Poc1 centriolar protein b (Poc1b). Previous functional studies in human cells and zebrafish embryos implicated that Poc1b plays important roles in centriole duplication and length control, as well as ciliogenesis. To study the function of Poc1b in photoreceptor sensory cilia and other primary cilia, we expressed a tagged recombinant Poc1b protein in cultured renal epithelial cells and rat retina. Poc1b was localized to the centrioles and spindle bundles during cell cycle progression, and to the basal body of photoreceptor sensory cilia. A morpholino knockdown and complementation assay of poc1b in zebrafish showed that loss of poc1b led to a range of morphological anomalies of cilia commonly associated with human ciliopathies. In the retina, the development of retinal laminae was significantly delayed and the length of photoreceptor outer segments was shortened. Visual behavior studies revealed impaired visual function in the poc1b morphants. In addition, ciliopathy-associated developmental defects, such as small eyes, curved body axis, heart defects, and shortened cilia in Kupffer's vesicle, were observed as well. These data suggest that poc1b is required for normal development and ciliogenesis of retinal photoreceptor sensory cilia and other cilia. Furthermore, this conclusion is supported by recent findings that mutations in POC1B gene have been identified in patients with inherited retinal dystrophy and syndromic retinal ciliopathy.


Assuntos
Cílio Conector dos Fotorreceptores/fisiologia , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/fisiologia , Animais , Células CHO , Proteínas de Ciclo Celular/genética , Centríolos/fisiologia , Cílios/fisiologia , Cricetulus , Técnicas de Silenciamento de Genes , Teste de Complementação Genética , Cardiopatias Congênitas/genética , Humanos , Camundongos , Mutação , Proteômica , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Distrofias Retinianas/genética , Transtornos da Visão/genética , Transtornos da Visão/fisiopatologia , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
9.
Exp Eye Res ; 138: 32-41, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26093275

RESUMO

Mammalian photoreceptors contain specialised connecting cilia that connect the inner (IS) to the outer segments (OS). Dysfunction of the connecting cilia due to mutations in ciliary proteins are a common cause of the inherited retinal dystrophy retinitis pigmentosa (RP). Mutations affecting the Retinitis Pigmentosa GTPase Regulator (RPGR) protein is one such cause, affecting 10-20% of all people with RP and the majority of those with X-linked RP. RPGR is located in photoreceptor connecting cilia. It interacts with a wide variety of ciliary proteins, but its exact function is unknown. Recently, there have been important advances both in our understanding of RPGR function and towards the development of a therapy. This review summarises the existing literature on human RPGR function and dysfunction, and suggests that RPGR plays a role in the function of the ciliary gate, which controls access of both membrane and soluble proteins to the photoreceptor outer segment. We discuss key models used to investigate and treat RPGR disease and suggest that gene augmentation therapy offers a realistic therapeutic approach, although important questions still remain to be answered, while cell replacement therapy based on retinal progenitor cells represents a more distant prospect.


Assuntos
Proteínas do Olho/fisiologia , Terapia Genética , Células Fotorreceptoras de Vertebrados/fisiologia , Retinose Pigmentar/fisiopatologia , Retinose Pigmentar/terapia , Animais , Modelos Animais de Doenças , Humanos , Cílio Conector dos Fotorreceptores/fisiologia
10.
Acta Ophthalmol ; 93(1): 83-94, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25385675

RESUMO

PURPOSE: Defects in MAK, encoding a protein localized to the photoreceptor connecting cilium, have recently been associated with autosomal recessive retinitis pigmentosa (RP). The aim of this study is to describe our detailed clinical observations in patients with MAK-associated RP, including an assessment of syndromic symptoms frequently observed in ciliopathies. METHODS: In this international collaborative study, 11 patients carrying nonsense or missense mutations in MAK were clinically evaluated, including extensive assessment of the medical history, slit-lamp biomicroscopy, ophthalmoscopy, kinetic perimetry, electroretinography (ERG), spectral-domain optical coherence tomography (SD-OCT), autofluorescence imaging and fundus photography. Additionally, we used a questionnaire to evaluate the presence of syndromic features and tested the olfactory function. RESULTS: MAK-associated RP is not associated with syndromic features, not even with subclinical dysfunction of the olfactory apparatus. All patients experienced typical RP symptoms of night blindness followed by visual field constriction. Symptoms initiated between childhood and the age of 43 (mean: 23 years). Although some patients experienced vision loss, the visual acuity remained normal in most patients. ERG and ophthalmoscopy revealed classic RP characteristics, and SD-OCT demonstrated thinning of the overall retina, outer nuclear layer and photoreceptor-pigment epithelium complex. CONCLUSION: Nonsense and missense mutations in MAK give rise to a non-syndromic recessive RP phenotype without apparent extra-ocular features. When compared to other retinal ciliopathies, MAK-associated RP appears to be relatively mild and shows remarkable resemblance to RP1-associated RP, which could be explained by the close functional relation of these proteins.


Assuntos
Códon sem Sentido , Mutação de Sentido Incorreto , Cílio Conector dos Fotorreceptores/metabolismo , Proteínas Serina-Treonina Quinases/genética , Retinose Pigmentar/genética , Adulto , Idoso , Eletrorretinografia , Feminino , Angiofluoresceinografia , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Proteínas Serina-Treonina Quinases/metabolismo , Retinose Pigmentar/patologia , Inquéritos e Questionários , Tomografia de Coerência Óptica , Testes de Campo Visual , Adulto Jovem
11.
Hum Mol Genet ; 24(6): 1584-601, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25398945

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Cílio Conector dos Fotorreceptores/patologia , Proteínas/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/patologia , Animais , Apoptose , Bovinos , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/genética , Deleção de Genes , Humanos , Camundongos , Camundongos Mutantes , Cílio Conector dos Fotorreceptores/metabolismo , Transporte Proteico , Células Fotorreceptoras Retinianas Cones/patologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Rodopsina/metabolismo
12.
Am J Hum Genet ; 95(2): 131-42, 2014 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25018096

RESUMO

Exome sequencing revealed a homozygous missense mutation (c.317C>G [p.Arg106Pro]) in POC1B, encoding POC1 centriolar protein B, in three siblings with autosomal-recessive cone dystrophy or cone-rod dystrophy and compound-heterozygous POC1B mutations (c.199_201del [p.Gln67del] and c.810+1G>T) in an unrelated person with cone-rod dystrophy. Upon overexpression of POC1B in human TERT-immortalized retinal pigment epithelium 1 cells, the encoded wild-type protein localized to the basal body of the primary cilium, whereas this localization was lost for p.Arg106Pro and p.Gln67del variant forms of POC1B. Morpholino-oligonucleotide-induced knockdown of poc1b translation in zebrafish resulted in a dose-dependent small-eye phenotype, impaired optokinetic responses, and decreased length of photoreceptor outer segments. These ocular phenotypes could partially be rescued by wild-type human POC1B mRNA, but not by c.199_201del and c.317C>G mutant human POC1B mRNAs. Yeast two-hybrid screening of a human retinal cDNA library revealed FAM161A as a binary interaction partner of POC1B. This was confirmed in coimmunoprecipitation and colocalization assays, which both showed loss of FAM161A interaction with p.Arg106Pro and p.Gln67del variant forms of POC1B. FAM161A was previously implicated in autosomal-recessive retinitis pigmentosa and shown to be located at the base of the photoreceptor connecting cilium, where it interacts with several other ciliopathy-associated proteins. Altogether, this study demonstrates that POC1B mutations result in a defect of the photoreceptor sensory cilium and thus affect cone and rod photoreceptors.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas do Olho/metabolismo , Células Fotorreceptoras Retinianas Cones/patologia , Células Fotorreceptoras Retinianas Bastonetes/patologia , Retinose Pigmentar/genética , Sequência de Aminoácidos , Animais , Corpos Basais , Sequência de Bases , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Exoma/genética , Proteínas do Olho/genética , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Masculino , Dados de Sequência Molecular , Morfolinos/genética , Mutação de Sentido Incorreto , Países Baixos , Cílio Conector dos Fotorreceptores/metabolismo , Segmento Externo das Células Fotorreceptoras da Retina/fisiologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Análise de Sequência de DNA , Turquia , Transtornos da Visão/genética , Peixe-Zebra
13.
Prog Retin Eye Res ; 38: 1-19, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24135424

RESUMO

Rhodopsin is a key molecular constituent of photoreceptor cells, yet understanding of how it regulates photoreceptor membrane trafficking and biogenesis of light-sensing organelles, the rod outer segments (ROS) is only beginning to emerge. Recently identified sequence of well-orchestrated molecular interactions of rhodopsin with the functional networks of Arf and Rab GTPases at multiple stages of intracellular targeting fits well into the complex framework of the biogenesis and maintenance of primary cilia, of which the ROS is one example. This review will discuss the latest progress in dissecting the molecular complexes that coordinate rhodopsin incorporation into ciliary-targeted carriers with the recruitment and activation of membrane tethering complexes and regulators of fusion with the periciliary plasma membrane. In addition to revealing the fundamental principals of ciliary membrane renewal, recent advances also provide molecular insight into the ways by which disruptions of the exquisitely orchestrated interactions lead to cilia dysfunction and result in human retinal dystrophies and syndromic diseases that affect multiple organs, including the eyes.


Assuntos
Substâncias Macromoleculares/metabolismo , Cílio Conector dos Fotorreceptores/fisiologia , Rodopsina/metabolismo , Segmento Externo da Célula Bastonete/fisiologia , Animais , Transporte Biológico/fisiologia , Humanos , Transporte Proteico/fisiologia
14.
Am J Hum Genet ; 93(1): 110-7, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23746546

RESUMO

The majority of the genetic causes of autosomal-recessive (ar) cone-rod dystrophy (CRD) are currently unknown. A combined approach of homozygosity mapping and exome sequencing revealed a homozygous nonsense mutation (c.565C>T [p.Glu189*]) in RAB28 in a German family with three siblings with arCRD. Another homozygous nonsense mutation (c.409C>T [p.Arg137*]) was identified in a family of Moroccan Jewish descent with two siblings affected by arCRD. All five affected individuals presented with hyperpigmentation in the macula, progressive loss of the visual acuity, atrophy of the retinal pigment epithelium, and severely reduced cone and rod responses on the electroretinogram. RAB28 encodes a member of the Rab subfamily of the RAS-related small GTPases. Alternative RNA splicing yields three predicted protein isoforms with alternative C-termini, which are all truncated by the nonsense mutations identified in the arCRD families in this report. Opposed to other Rab GTPases that are generally geranylgeranylated, RAB28 is predicted to be farnesylated. Staining of rat retina showed localization of RAB28 to the basal body and the ciliary rootlet of the photoreceptors. Analogous to the function of other RAB family members, RAB28 might be involved in ciliary transport in photoreceptor cells. This study reveals a crucial role for RAB28 in photoreceptor function and suggests that mutations in other Rab proteins may also be associated with retinal dystrophies.


Assuntos
Genes Recessivos , Retinose Pigmentar/genética , Proteínas rab de Ligação ao GTP/genética , Adolescente , Adulto , Processamento Alternativo , Animais , Criança , Mapeamento Cromossômico , Cílios/metabolismo , Cílios/patologia , Códon sem Sentido/genética , Regulação da Expressão Gênica , Estudos de Associação Genética , Predisposição Genética para Doença , Homozigoto , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Linhagem , Cílio Conector dos Fotorreceptores/metabolismo , Cílio Conector dos Fotorreceptores/patologia , Prenilação de Proteína , Transporte Proteico , Ratos , Retina/enzimologia , Retina/patologia , Epitélio Pigmentado da Retina/enzimologia , Epitélio Pigmentado da Retina/patologia , Células Fotorreceptoras Retinianas Bastonetes/enzimologia , Células Fotorreceptoras Retinianas Bastonetes/patologia , Retinose Pigmentar/enzimologia , Retinose Pigmentar/patologia , Acuidade Visual , Proteínas rab de Ligação ao GTP/metabolismo
15.
Hum Mol Genet ; 22(8): 1507-15, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23297361

RESUMO

Ataxia-telangiectasia and Rad3 (ATR), a sensor of DNA damage, is associated with the regulation and control of cell division. ATR deficit is known to cause Seckel syndrome, characterized by severe proportionate short stature and microcephaly. We used a mouse model for Seckel disease to study the effect of ATR deficit on retinal development and function and we have found a new role for ATR, which is critical for the postnatal development of the photoreceptor (PR) layer in mouse retina. The structural and functional characterization of the ATR(+/s) mouse retinas displayed a specific, severe and early degeneration of rod and cone cells resembling some characteristics of human retinal degenerations. A new localization of ATR in the cilia of PRs and the fact that mutant mice have shorter cilia suggests that the PR degeneration here described results from a ciliary defect.


Assuntos
Proteínas de Ciclo Celular/genética , Células Fotorreceptoras de Vertebrados , Proteínas Serina-Treonina Quinases/genética , Retina/metabolismo , Degeneração Retiniana/genética , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Dano ao DNA , Modelos Animais de Doenças , Nanismo/genética , Nanismo/patologia , Fácies , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Microcefalia/genética , Microcefalia/patologia , Mutação , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Cílio Conector dos Fotorreceptores/metabolismo , Cílio Conector dos Fotorreceptores/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Retina/crescimento & desenvolvimento , Degeneração Retiniana/patologia
17.
Hum Mol Genet ; 21(23): 5174-84, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22940612

RESUMO

Retinitis pigmentosa (RP) is a retinal degenerative disease characterized by the progressive loss of photoreceptors. We have previously demonstrated that RP can be caused by recessive mutations in the human FAM161A gene, encoding a protein with unknown function that contains a conserved region shared only with a distant paralog, FAM161B. In this study, we show that FAM161A localizes at the base of the photoreceptor connecting cilium in human, mouse and rat. Furthermore, it is also present at the ciliary basal body in ciliated mammalian cells, both in native conditions and upon the expression of recombinant tagged proteins. Yeast two-hybrid analysis of binary interactions between FAM161A and an array of ciliary and ciliopathy-associated proteins reveals direct interaction with lebercilin, CEP290, OFD1 and SDCCAG8, all involved in hereditary retinal degeneration. These interactions are mediated by the C-terminal moiety of FAM161A, as demonstrated by pull-down experiments in cultured cell lines and in bovine retinal extracts. As other ciliary proteins, FAM161A can also interact with the microtubules and organize itself into microtubule-dependent intracellular networks. Moreover, small interfering RNA-mediated depletion of FAM161A transcripts in cultured cells causes the reduction in assembled primary cilia. Taken together, these data indicate that FAM161A-associated RP can be considered as a novel retinal ciliopathy and that its molecular pathogenesis may be related to other ciliopathies.


Assuntos
Proteínas do Olho/genética , Cílio Conector dos Fotorreceptores/metabolismo , Retinose Pigmentar/genética , Retinose Pigmentar/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Cílios/metabolismo , Proteínas do Olho/metabolismo , Expressão Gênica , Humanos , Camundongos , Células Fotorreceptoras de Vertebrados/metabolismo , Ligação Proteica , Transporte Proteico , Interferência de RNA , Ratos , Retina/metabolismo , Retina/patologia
18.
EMBO J ; 31(20): 4057-71, 2012 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-22983554

RESUMO

Dysfunctional trafficking to primary cilia is a frequent cause of human diseases known as ciliopathies, yet molecular mechanisms for specific targeting of sensory receptors to cilia are largely unknown. Here, we show that the targeting of ciliary cargo, represented by rhodopsin, is mediated by a specialized system, the principal component of which is the Arf GAP ASAP1. Ablation of ASAP1 abolishes ciliary targeting and causes formation of actin-rich periciliary membrane projections that accumulate mislocalized rhodopsin. We find that ASAP1 serves as a scaffold that brings together the proteins necessary for transport to the cilia including the GTP-binding protein Arf4 and the two G proteins of the Rab family--Rab11 and Rab8--linked by the Rab8 guanine nucleotide exchange factor Rabin8. ASAP1 recognizes the FR ciliary targeting signal of rhodopsin. Rhodopsin FR-AA mutant, defective in ASAP1 binding, fails to interact with Rab8 and translocate across the periciliary diffusion barrier. Our study implies that other rhodopsin-like sensory receptors may interact with this conserved system and reach the cilia using the same platform.


Assuntos
Fatores de Ribosilação do ADP/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Cílios/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Segmento Externo da Célula Bastonete/fisiologia , Proteínas rab de Ligação ao GTP/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Bovinos , Células Cultivadas , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Quinases do Centro Germinativo , Complexo de Golgi/metabolismo , Túbulos Renais Coletores/citologia , Camundongos , Camundongos Knockout , Modelos Moleculares , Cílio Conector dos Fotorreceptores/fisiologia , Ligação Proteica , Mapeamento de Interação de Proteínas , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Ranidae , Proteínas Recombinantes de Fusão/metabolismo , Segmento Externo das Células Fotorreceptoras da Retina , Rodopsina/química , Rodopsina/genética , Rodopsina/metabolismo , Segmento Externo da Célula Bastonete/ultraestrutura
19.
Vision Res ; 75: 5-10, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22892112

RESUMO

This review will focus on the conserved molecular mechanisms for the specific targeting of rhodopsin and rhodopsin-like sensory receptors to the primary cilia. We will discuss the molecular assemblies that control the movement of rhodopsin from the central sorting station of the cell, the trans-Golgi network (TGN), into membrane-enclosed rhodopsin transport carriers (RTCs), and their delivery to the primary cilia and the cilia-derived sensory organelle, the rod outer segment (ROS). Recent studies reveal that these processes are initiated by the synergistic interaction of rhodopsin with the active form of the G-protein Arf4 and the Arf GTPase activating protein (GAP) ASAP1. During rhodopsin progression, ASAP1 serves as an activation platform that brings together the proteins necessary for transport to the cilia, including the Rab11a-Rabin8-Rab8 complex involved in ciliogenesis. These specialized molecular assemblies, through successive action of discrete modules, cooperatively determine how rhodopsin and other rhodopsin-like signaling receptors gain access to primary cilia.


Assuntos
Cílio Conector dos Fotorreceptores/fisiologia , Segmento Externo das Células Fotorreceptoras da Retina/fisiologia , Rodopsina/metabolismo , Fatores de Ribosilação do ADP/metabolismo , Animais , Transporte Biológico/fisiologia , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Transporte Proteico/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo , Rede trans-Golgi/metabolismo
20.
Vision Res ; 75: 112-6, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22921640

RESUMO

For being a polarized neuron and having a sensory cilium, photoreceptors attract remarkable attention. This is due their highly polarized structure and active visual signal transduction cascades and for the enrichment of complex networks of proteins in the cilium. Structural and functional maintenance of the photoreceptor sensory cilium, also called outer segment, ensures that light signal is received and relayed appropriately to the brain. Any perturbations in the protein content of the outer segment result in photoreceptor dysfunction, degeneration and eventually, blindness. This review focuses on the importance of photoreceptor sensory cilium to carry out signal transduction cascade for vision.


Assuntos
Células Fotorreceptoras de Vertebrados/fisiologia , Cílio Conector dos Fotorreceptores/fisiologia , Segmento Externo das Células Fotorreceptoras da Retina/fisiologia , Transdução de Sinais/fisiologia , Visão Ocular/fisiologia , Animais , Transporte Biológico/fisiologia , Humanos , Proteínas de Membrana/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...