Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Acta Neuropathol Commun ; 9(1): 17, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33509294

RESUMEN

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.


Asunto(s)
Cilio Conector de los Fotorreceptores/metabolismo , Proteínas PrPSc/metabolismo , Células Bipolares de la Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Segmento Interno de las Células Fotorreceptoras Retinianas/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , Muerte Celular , Progresión de la Enfermedad , Ratones , Microscopía Confocal , Microscopía Electrónica , Microscopía Fluorescente , Cilio Conector de los Fotorreceptores/patología , Cilio Conector de los Fotorreceptores/ultraestructura , Proteínas PrPSc/administración & dosificación , Células Bipolares de la Retina/patología , Células Bipolares de la Retina/ultraestructura , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Conos/ultraestructura , Segmento Interno de las Células Fotorreceptoras Retinianas/patología , Segmento Interno de las Células Fotorreceptoras Retinianas/ultraestructura , Segmento Externo de las Células Fotorreceptoras Retinianas/patología , Segmento Externo de las Células Fotorreceptoras Retinianas/ultraestructura , Células Fotorreceptoras Retinianas Bastones/patología , Células Fotorreceptoras Retinianas Bastones/ultraestructura , Scrapie/metabolismo , Scrapie/patología
2.
Dev Cell ; 53(3): 287-299.e5, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32275885

RESUMEN

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.


Asunto(s)
Cilios/patología , Histona Desacetilasa 6/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 5/metabolismo , Cilio Conector de los Fotorreceptores/patología , Proteolisis , Retinopatía de la Prematuridad/patología , Ubiquitinación , Animales , Cilios/metabolismo , Femenino , Histona Desacetilasa 6/genética , Histona Desacetilasa 6/metabolismo , MAP Quinasa Quinasa Quinasa 5/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/toxicidad , Fosforilación , Cilio Conector de los Fotorreceptores/metabolismo , Retinopatía de la Prematuridad/etiología , Retinopatía de la Prematuridad/metabolismo
3.
J Cell Biol ; 217(8): 2851-2865, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29899041

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Cilio Conector de los Fotorreceptores/metabolismo , Proteínas Adaptadoras Transductoras de Señales/análisis , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antígenos de Neoplasias , Proteínas Portadoras/análisis , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Proteínas del Citoesqueleto , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas del Ojo/análisis , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Cilio Conector de los Fotorreceptores/ultraestructura , Transporte de Proteínas/genética
4.
Br J Ophthalmol ; 99(12): 1725-31, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26294103

RESUMEN

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.


Asunto(s)
Mutación del Sistema de Lectura/genética , Genes Recesivos , Mutación Missense/genética , Cilio Conector de los Fotorreceptores/metabolismo , Proteínas/genética , Retina/patología , Distrofias Retinianas/genética , Adulto , Secuencia de Aminoácidos , Animales , Western Blotting , Niño , Consanguinidad , Proteínas del Citoesqueleto , Dilatación Patológica , Electrorretinografía , Femenino , Humanos , Imagen por Resonancia Magnética , Ratones , Datos de Secuencia Molecular , Linaje , Reacción en Cadena de la Polimerasa , Proteínas/metabolismo , Estudios Retrospectivos , Sus scrofa , Donantes de Tejidos , Tomografía de Coherencia Óptica
5.
Acta Ophthalmol ; 93(1): 83-94, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25385675

RESUMEN

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.


Asunto(s)
Codón sin Sentido , Mutación Missense , Cilio Conector de los Fotorreceptores/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Retinitis Pigmentosa/genética , Adulto , Anciano , Electrorretinografía , Femenino , Angiografía con Fluoresceína , Humanos , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Proteínas Serina-Treonina Quinasas/metabolismo , Retinitis Pigmentosa/patología , Encuestas y Cuestionarios , Tomografía de Coherencia Óptica , Pruebas del Campo Visual , Adulto Joven
6.
Hum Mol Genet ; 24(6): 1584-601, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25398945

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Cilio Conector de los Fotorreceptores/patología , Proteínas/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Animales , Apoptosis , Bovinos , Proteínas del Citoesqueleto , Proteínas de Unión al ADN/genética , Eliminación de Gen , Humanos , Ratones , Ratones Mutantes , Cilio Conector de los Fotorreceptores/metabolismo , Transporte de Proteínas , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Rodopsina/metabolismo
7.
Am J Hum Genet ; 95(2): 131-42, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25018096

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas del Ojo/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/genética , Secuencia de Aminoácidos , Animales , Cuerpos Basales , Secuencia de Bases , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Exoma/genética , Proteínas del Ojo/genética , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Masculino , Datos de Secuencia Molecular , Morfolinos/genética , Mutación Missense , Países Bajos , Cilio Conector de los Fotorreceptores/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/fisiología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Análisis de Secuencia de ADN , Turquía , Trastornos de la Visión/genética , Pez Cebra
8.
Am J Hum Genet ; 93(1): 110-7, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23746546

RESUMEN

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.


Asunto(s)
Genes Recesivos , Retinitis Pigmentosa/genética , Proteínas de Unión al GTP rab/genética , Adolescente , Adulto , Empalme Alternativo , Animales , Niño , Mapeo Cromosómico , Cilios/metabolismo , Cilios/patología , Codón sin Sentido/genética , Regulación de la Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Linaje , Cilio Conector de los Fotorreceptores/metabolismo , Cilio Conector de los Fotorreceptores/patología , Prenilación de Proteína , Transporte de Proteínas , Ratas , Retina/enzimología , Retina/patología , Epitelio Pigmentado de la Retina/enzimología , Epitelio Pigmentado de la Retina/patología , Células Fotorreceptoras Retinianas Bastones/enzimología , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/enzimología , Retinitis Pigmentosa/patología , Agudeza Visual , Proteínas de Unión al GTP rab/metabolismo
9.
Hum Mol Genet ; 22(8): 1507-15, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23297361

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/genética , Células Fotorreceptoras de Vertebrados , Proteínas Serina-Treonina Quinasas/genética , Retina/metabolismo , Degeneración Retiniana/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Modelos Animales de Enfermedad , Enanismo/genética , Enanismo/patología , Facies , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Microcefalia/genética , Microcefalia/patología , Mutación , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Cilio Conector de los Fotorreceptores/metabolismo , Cilio Conector de los Fotorreceptores/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Retina/crecimiento & desarrollo , Degeneración Retiniana/patología
10.
Hum Mol Genet ; 21(23): 5174-84, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22940612

RESUMEN

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.


Asunto(s)
Proteínas del Ojo/genética , Cilio Conector de los Fotorreceptores/metabolismo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Cilios/metabolismo , Proteínas del Ojo/metabolismo , Expresión Génica , Humanos , Ratones , Células Fotorreceptoras de Vertebrados/metabolismo , Unión Proteica , Transporte de Proteínas , Interferencia de ARN , Ratas , Retina/metabolismo , Retina/patología
11.
Vision Res ; 75: 2-4, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22884633

RESUMEN

The traffic of proteins to the outer segment of photoreceptors is a fundamentally important process, which when perturbed results in photoreceptor cell death. Recent reports have revealed a novel pathway for the traffic of lipid-modified proteins involving the small GTPase Arl3 and its effectors PDEδ and Unc119. The retinitis pigmentosa protein RP2 is a GTPase activating protein (GAP) for Arl3 and also appears to regulate the assembly and traffic of membrane associated protein complexes. We recently identified the Gß subunit of transducin (Gß1) as a novel RP2 interacting protein. Our data support a role for RP2 in facilitating membrane association and traffic of Gß1, potentially prior to the formation of the obligate Gß:Gγ heterodimer. Here, we review the recent evidence that suggests that RP2 co-operates with Arl3 and its effectors in protein complex assembly and membrane specification for lipid-modified proteins. This is exemplified by the co-ordination of cilia associated traffic for heterotrimeric G proteins and we propose a model for the role of Arl3 and RP2 in this process.


Asunto(s)
Factores de Ribosilacion-ADP/fisiología , Proteínas del Ojo/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/fisiología , Cilio Conector de los Fotorreceptores/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Animales , Proteínas de Unión al GTP , Humanos , Transporte de Proteínas/fisiología , Retinitis Pigmentosa/metabolismo , Transducina/metabolismo
12.
Cell Death Dis ; 3: e355, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22825473

RESUMEN

The retinitis pigmentosa GTPase regulator (RPGR) and nephrocystin-4 (NPHP4) comprise two key partners of the assembly complex of the RPGR-interacting protein 1 (RPGRIP1). Mutations in RPGR and NPHP4 are linked to severe multisystemic diseases with strong retinal involvement of photoreceptor neurons, whereas those in RPGRIP1 cause the fulminant photoreceptor dystrophy, Leber congenital amaurosis (LCA). Further, mutations in Rpgrip1 and Nphp4 suppress the elaboration of the outer segment compartment of photoreceptor neurons by elusive mechanisms, the understanding of which has critical implications in uncovering the pathogenesis of syndromic retinal dystrophies. Here we show RPGRIP1 localizes to the photoreceptor connecting cilium (CC) distally to the centriole/basal body marker, centrin-2 and the ciliary marker, acetylated-α-tubulin. NPHP4 abuts proximally RPGRIP1, RPGR and the serologically defined colon cancer antigen-8 (SDCCAG8), a protein thought to partake in the RPGRIP1 interactome and implicated also in retinal-renal ciliopathies. Ultrastructurally, RPGRIP1 localizes exclusively throughout the photoreceptor CC and Rpgrip1(nmf247) photoreceptors present shorter cilia with a ruffled membrane. Strikingly, Rpgrip1(nmf247) mice without RPGRIP1 expression lack NPHP4 and RPGR in photoreceptor cilia, whereas the SDCCAG8 and acetylated-α-tubulin ciliary localizations are strongly decreased, even though the NPHP4 and SDCCAG8 expression levels are unaffected and those of acetylated-α-tubulin and γ-tubulin are upregulated. Further, RPGRIP1 loss in photoreceptors shifts the subcellular partitioning of SDCCAG8 and NPHP4 to the membrane fraction associated to the endoplasmic reticulum. Conversely, the ciliary localization of these proteins is unaffected in glomeruli or tubular kidney cells of Rpgrip1(nmf247), but NPHP4 is downregulated developmentally and selectively in kidney cortex. Hence, RPGRIP1 presents cell type-dependent pathological effects crucial to the ciliary targeting and subcellular partitioning of NPHP4, RPGR and SDCCAG8, and acetylation of ciliary α-tubulin or its ciliary targeting, selectively in photoreceptors, but not kidney cells, and these pathological effects underlie photoreceptor degeneration and LCA.


Asunto(s)
Autoantígenos/metabolismo , Proteínas Portadoras/metabolismo , Proteínas del Ojo/metabolismo , Proteínas de Neoplasias/metabolismo , Cilio Conector de los Fotorreceptores/metabolismo , Proteínas/metabolismo , Animales , Proteínas de Unión al Calcio/metabolismo , Proteínas del Citoesqueleto , Túbulos Renales/metabolismo , Ratones , Mutación , Cilio Conector de los Fotorreceptores/ultraestructura , Proteínas/genética , Rodopsina/metabolismo , Tubulina (Proteína)/metabolismo , Regulación hacia Arriba
13.
Ophthalmic Genet ; 32(1): 1-11, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21174525

RESUMEN

Cilia are specialized dynamic organelles extending from the surface of almost all mammalian cells. Since proteins and protein precursors are transported across the ciliary compartments via intraflagellar transport (IFT), mutations in genes encoding proteins that participate in IFT can cause a spectrum of different ciliopathies. Photoreceptors of the mammalian retina contain ciliary structures that connect the inner (IS) with the outer segments (OS). This structure, the connecting cilium (CC), serves as the only junction between OS and IS, the correct passage of proteins through the CC is crucial for the functioning and maintenance of the cells. Therefore, any impairment of the IFT leads to severe malfunction of photoreceptors, and may induce apoptosis ultimately leading to the degeneration of the retina. The Retinitis Pigmentosa GTPase Regulator (RPGR), which is located in the CC, participates in the IFT and interacts with a variety of proteins, including RPGRIP-1, CEP290, NPM, SMC1 and 3 and IFT88. However, the function of RPGR through its interaction with these proteins is not yet entirely understood. Mutations in the RPGR gene lead to X-linked Retinitis pigmentosa (XLRP), one of the most severe and early onset forms of RP. Gene therapy is considered a potential therapeutic option and is currently under investigation in several animal models of XLRP. However, some of the currently available mouse models are only partially suitable for the development of therapeutic strategies and the quest for more appropriate small animal models is still an issue.


Asunto(s)
Proteínas del Ojo/fisiología , Terapia Genética , Cilio Conector de los Fotorreceptores/metabolismo , Retinitis Pigmentosa/genética , Animales , Antígenos de Neoplasias/metabolismo , Transporte Biológico Activo , Proteínas de Ciclo Celular/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas del Citoesqueleto , Humanos , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Nucleofosmina , Proteínas/metabolismo , Retinitis Pigmentosa/terapia , Proteínas Supresoras de Tumor/metabolismo
14.
Proc Natl Acad Sci U S A ; 107(52): 22671-6, 2010 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-21148103

RESUMEN

Cilia function as cell sensors in many organs, and their disorders are referred to as "ciliopathies." Although ciliary components and transport machinery have been well studied, regulatory mechanisms of ciliary formation and maintenance are poorly understood. Here we show that male germ cell-associated kinase (Mak) regulates retinal photoreceptor ciliary length and subcompartmentalization. Mak was localized both in the connecting cilia and outer-segment axonemes of photoreceptor cells. In the Mak-null retina, photoreceptors exhibit elongated cilia and progressive degeneration. We observed accumulation of intraflagellar transport 88 (IFT88) and IFT57, expansion of kinesin family member 3A (Kif3a), and acetylated α-tubulin signals in the Mak-null photoreceptor cilia. We found abnormal rhodopsin accumulation in the Mak-null photoreceptor cell bodies at postnatal day 14. In addition, overexpression of retinitis pigmentosa 1 (RP1), a microtubule-associated protein localized in outer-segment axonemes, induced ciliary elongation, and Mak coexpression rescued excessive ciliary elongation by RP1. The RP1 N-terminal portion induces ciliary elongation and increased intensity of acetylated α-tubulin labeling in the cells and is phosphorylated by Mak. These results suggest that Mak is essential for the regulation of ciliary length and is required for the long-term survival of photoreceptors.


Asunto(s)
Cilio Conector de los Fotorreceptores/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Retina/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Femenino , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Hibridación in Situ , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Células 3T3 NIH , Fosforilación , Cilio Conector de los Fotorreceptores/ultraestructura , Proteínas Serina-Treonina Quinasas/genética , Retina/embriología , Retina/crecimiento & desarrollo , Segmento Externo de las Células Fotorreceptoras Retinianas/ultraestructura , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Factores de Tiempo
15.
Hum Gene Ther ; 21(8): 993-1004, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20384479

RESUMEN

RPGR-interacting protein-1 (RPGRIP1) is localized in the photoreceptor-connecting cilium, where it anchors the RPGR (retinitis pigmentosa GTPase regulator) protein, and its function is essential for photoreceptor maintenance. Genetic defect in RPGRIP1 is a known cause of Leber congenital amaurosis (LCA), a severe, early-onset form of retinal degeneration. We evaluated the efficacy of replacement gene therapy in a murine model of LCA carrying a targeted disruption of RPGRIP1. The replacement construct, packaged in an adeno-associated virus serotype 8 (AAV8) vector, used a rhodopsin kinase gene promoter to drive RPGRIP1 expression. Both promoter and transgene were of human origin. After subretinal delivery of the replacement gene in the mutant mice, human RPGRIP1 was expressed specifically in photoreceptors, localized correctly in the connecting cilia, and restored the normal localization of RPGR. Electroretinogram and histological examinations showed better preservation of rod and cone photoreceptor function and improved photoreceptor survival in the treated eyes. This study demonstrates the efficacy of human gene replacement therapy and validates a gene therapy design for future clinical trials in patients afflicted with this condition. Our results also have therapeutic implications for other forms of retinal degenerations attributable to a ciliary defect.


Asunto(s)
Terapia Genética , Amaurosis Congénita de Leber/terapia , Cilio Conector de los Fotorreceptores/metabolismo , Proteínas/genética , Animales , Proteínas del Citoesqueleto , Modelos Animales de Enfermedad , Electrorretinografía , Quinasa 1 del Receptor Acoplado a Proteína-G/metabolismo , Vectores Genéticos , Humanos , Amaurosis Congénita de Leber/genética , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Proteínas/uso terapéutico , Degeneración Retiniana/genética , Degeneración Retiniana/terapia
16.
J Gen Physiol ; 135(3): 173-96, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20176852

RESUMEN

Transport of proteins to and from cilia is crucial for normal cell function and survival, and interruption of transport has been implicated in degenerative and neoplastic diseases. It has been hypothesized that the ciliary axoneme and structures adjacent to and including the basal bodies of cilia impose selective barriers to the movement of proteins into and out of the cilium. To examine this hypothesis, using confocal and multiphoton microscopy we determined the mobility of the highly soluble photoactivatable green fluorescent protein (PAGFP) in the connecting cilium (CC) of live Xenopus retinal rod photoreceptors, and in the contiguous subcellular compartments bridged by the CC, the inner segment (IS) and the outer segment (OS). The estimated axial diffusion coefficients are D(CC) = 2.8 +/- 0.3, D(IS) = 5.2 +/- 0.6, and D(OS) = 0.079 +/- 0.009 microm(2) s(-1). The results establish that the CC does not pose a major barrier to protein diffusion within the rod cell. However, the results also reveal that axial diffusion in each of the rod's compartments is substantially retarded relative to aqueous solution: the axial diffusion of PAGFP was retarded approximately 18-, 32- and 1,000-fold in the IS, CC, and OS, respectively, with approximately 20-fold of the reduction in the OS attributable to tortuosity imposed by the lamellar disc membranes. Previous investigation of PAGFP diffusion in passed, spherical Chinese hamster ovary cells yielded D(CHO) = 20 microm(2) s(-1), and estimating cytoplasmic viscosity as D(aq)/D(CHO) = 4.5, the residual 3- to 10-fold reduction in PAGFP diffusion is ascribed to sub-optical resolution structures in the IS, CC, and OS compartments.


Asunto(s)
Proteínas Fluorescentes Verdes/metabolismo , Cilio Conector de los Fotorreceptores/metabolismo , Segmento Interno de las Células Fotorreceptoras Retinianas/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Algoritmos , Animales , Animales Modificados Genéticamente , Transporte Biológico , Difusión , Colorantes Fluorescentes/metabolismo , Líquido Intracelular , Modelos Biológicos , Xenopus laevis
17.
Invest Ophthalmol Vis Sci ; 51(5): 2338-46, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19959638

RESUMEN

PURPOSE: Usher syndrome is the most common form of hereditary deaf-blindness. It is both clinically and genetically heterogeneous. The USH2D protein whirlin interacts via its PDZ domains with other Usher-associated proteins containing a C-terminal type I PDZ-binding motif. These proteins co-localize with whirlin at the region of the connecting cilium and at the synapse of photoreceptor cells. This study was undertaken to identify novel, Usher syndrome-associated, interacting partners of whirlin and thereby obtain more insights into the function of whirlin. METHODS: The database of ciliary proteins was searched for proteins that are present in both the retina and inner ear and contain a PDZ-binding motif. Interactions with whirlin were evaluated by yeast two-hybrid analyses and validated by glutathione S-transferase pull-down assays, co-immunoprecipitation, and co-localization in the retina with immunofluorescence and immunoelectron microscopy. RESULTS: The L-type calcium channel subunit Ca(v)1.3 (alpha(1D)) specifically interacts with whirlin. In adult photoreceptors, Ca(v)1.3 (alpha(1D)) and whirlin co-localize in the region of the connecting cilium and at the synapse. During murine embryonic development, the expression patterns of the Whrn and Cacna1d genes show significant overlap and include expression in the eye, the inner ear, and the central nervous system. CONCLUSIONS: The findings indicate that Ca(v)1.3 (alpha(1D)) is connected to the Usher protein network. This conclusion leads to the hypothesis that, in the retina, whirlin scaffolds Ca(v)1.3 (alpha(1D)) and therefore contributes to the organization of calcium channels in the photoreceptor cells, where both proteins may be involved in membrane fusions.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Proteínas de la Membrana/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Animales , Western Blotting , Células COS , Canales de Calcio Tipo L/genética , Chlorocebus aethiops , Biología Computacional , Bases de Datos de Proteínas , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Microscopía Inmunoelectrónica , Cilio Conector de los Fotorreceptores/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Retina/metabolismo , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...