Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
2.
Hum Mol Genet ; 26(24): 4896-4905, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29036441

RESUMEN

Mutations in rhodopsin, the light-sensitive protein of rod cells, are the most common cause of dominant retinitis pigmentosa (RP), a type of inherited blindness caused by the dysfunction and death of photoreceptor cells. The P23H mutation, the most frequent single cause of RP in the USA, causes rhodopsin misfolding and induction of the unfolded protein response (UPR), an adaptive ER stress response and signalling network that aims to enhance the folding and degradation of misfolded proteins to restore proteostasis. Prolonged UPR activation, and in particular the PERK branch, can reduce protein synthesis and initiate cell death through induction of pro-apoptotic pathways. Here, we investigated the effect of pharmacological PERK inhibition on retinal disease process in the P23H-1 transgenic rat model of retinal degeneration. PERK inhibition with GSK2606414A led to an inhibition of eIF2α phosphorylation, which correlated with reduced ERG function and decreased photoreceptor survival at both high and low doses of PERK inhibitor. Additionally, PERK inhibition increased the incidence of inclusion formation in cultured cells overexpressing P23H rod opsin, and increased rhodopsin aggregation in the P23H-1 rat retina, suggesting enhanced P23H misfolding and aggregation. In contrast, treatment of P23H-1 rats with an inhibitor of eIF2α phosphatase, salubrinal, led to improved photoreceptor survival. Collectively, these data suggest the activation of PERK is part of a protective response to mutant rhodopsin that ultimately limits photoreceptor cell death.


Asunto(s)
Retinitis Pigmentosa/metabolismo , Rodopsinas Sensoriales/metabolismo , eIF-2 Quinasa/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Línea Celular Transformada , Línea Celular Tumoral , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Humanos , Indoles/farmacología , Pliegue de Proteína , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Células Fotorreceptoras Retinianas Bastones/metabolismo , Retinitis Pigmentosa/genética , Rodopsinas Sensoriales/genética , Estrés Fisiológico/fisiología , Respuesta de Proteína Desplegada , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/genética
3.
Hum Mol Genet ; 26(2): 305-319, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28065882

RESUMEN

Protein misfolding caused by inherited mutations leads to loss of protein function and potentially toxic 'gain of function', such as the dominant P23H rhodopsin mutation that causes retinitis pigmentosa (RP). Here, we tested whether the AMPK activator metformin could affect the P23H rhodopsin synthesis and folding. In cell models, metformin treatment improved P23H rhodopsin folding and traffic. In animal models of P23H RP, metformin treatment successfully enhanced P23H traffic to the rod outer segment, but this led to reduced photoreceptor function and increased photoreceptor cell death. The metformin-rescued P23H rhodopsin was still intrinsically unstable and led to increased structural instability of the rod outer segments. These data suggest that improving the traffic of misfolding rhodopsin mutants is unlikely to be a practical therapy, because of their intrinsic instability and long half-life in the outer segment, but also highlights the potential of altering translation through AMPK to improve protein function in other protein misfolding diseases.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Metformina/administración & dosificación , Degeneración Retiniana/genética , Retinitis Pigmentosa/genética , Rodopsina/genética , Proteínas Quinasas Activadas por AMP/biosíntesis , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Proteínas Mutantes/genética , Células Fotorreceptoras/efectos de los fármacos , Células Fotorreceptoras/patología , Pliegue de Proteína/efectos de los fármacos , Deficiencias en la Proteostasis/genética , Deficiencias en la Proteostasis/patología , Ratas , Degeneración Retiniana/tratamiento farmacológico , Degeneración Retiniana/patología , Células Fotorreceptoras Retinianas Bastones/efectos de los fármacos , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/tratamiento farmacológico , Retinitis Pigmentosa/patología , Rodopsina/química , Segmento Externo de la Célula en Bastón/efectos de los fármacos , Segmento Externo de la Célula en Bastón/patología , Activación Transcripcional/efectos de los fármacos
4.
Cell Stem Cell ; 18(6): 769-781, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27151457

RESUMEN

Leber congenital amaurosis (LCA) is an inherited retinal dystrophy that causes childhood blindness. Photoreceptors are especially sensitive to an intronic mutation in the cilia-related gene CEP290, which causes missplicing and premature termination, but the basis of this sensitivity is unclear. Here, we generated differentiated photoreceptors in three-dimensional optic cups and retinal pigment epithelium (RPE) from iPSCs with this common CEP290 mutation to investigate disease mechanisms and evaluate candidate therapies. iPSCs differentiated normally into RPE and optic cups, despite abnormal CEP290 splicing and cilia defects. The highest levels of aberrant splicing and cilia defects were observed in optic cups, explaining the retinal-specific manifestation of this CEP290 mutation. Treating optic cups with an antisense morpholino effectively blocked aberrant splicing and restored expression of full-length CEP290, restoring normal cilia-based protein trafficking. These results provide a mechanistic understanding of the retina-specific phenotypes in CEP290 LCA patients and potential strategies for therapeutic intervention.


Asunto(s)
Ceguera/patología , Ceguera/terapia , Células Madre Pluripotentes Inducidas/citología , Patrón de Herencia/genética , Disco Óptico/citología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Proteínas de Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Cilios/efectos de los fármacos , Cilios/metabolismo , Proteínas del Citoesqueleto , Exones/genética , Proteínas del Ojo/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Amaurosis Congénita de Leber/patología , Masculino , Morfolinos/farmacología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Opsinas/metabolismo , Organogénesis/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Células Fotorreceptoras de Vertebrados/ultraestructura , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Epitelio Pigmentado de la Retina/ultraestructura , Proteínas de Unión al GTP rab/metabolismo
5.
Hum Mol Genet ; 24(4): 972-86, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25292197

RESUMEN

Mutations in the RP2 gene lead to a severe form of X-linked retinitis pigmentosa. RP2 patients frequently present with nonsense mutations and no treatments are currently available to restore RP2 function. In this study, we reprogrammed fibroblasts from an RP2 patient carrying the nonsense mutation c.519C>T (p.R120X) into induced pluripotent stem cells (iPSC), and differentiated these cells into retinal pigment epithelial cells (RPE) to study the mechanisms of disease and test potential therapies. RP2 protein was undetectable in the RP2 R120X patient cells, suggesting a disease mechanism caused by complete lack of RP2 protein. The RP2 patient fibroblasts and iPSC-derived RPE cells showed phenotypic defects in IFT20 localization, Golgi cohesion and Gß1 trafficking. These phenotypes were corrected by over-expressing GFP-tagged RP2. Using the translational read-through inducing drugs (TRIDs) G418 and PTC124 (Ataluren), we were able to restore up to 20% of endogenous, full-length RP2 protein in R120X cells. This level of restored RP2 was sufficient to reverse the cellular phenotypic defects observed in both the R120X patient fibroblasts and iPSC-RPE cells. This is the first proof-of-concept study to demonstrate successful read-through and restoration of RP2 function for the R120X nonsense mutation. The ability of the restored RP2 protein level to reverse the observed cellular phenotypes in cells lacking RP2 indicates that translational read-through could be clinically beneficial for patients.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas del Ojo/genética , Células Madre Pluripotentes Inducidas/citología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Mutación , Biosíntesis de Proteínas , Epitelio Pigmentado de la Retina/citología , Diferenciación Celular , Reprogramación Celular , Cilios/metabolismo , Cilios/patología , Proteínas del Ojo/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Proteínas de Unión al GTP , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Oxadiazoles/farmacología , Fenotipo , Biosíntesis de Proteínas/efectos de los fármacos , Transporte de Proteínas , Adulto Joven
6.
Hum Mutat ; 35(11): 1354-62, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25168334

RESUMEN

Mutations in the OPN1LW (L-) and OPN1MW (M-)cone opsin genes underlie a spectrum of cone photoreceptor defects from stationary loss of color vision to progressive retinal degeneration. Genotypes of 22 families with a range of cone disorders were grouped into three classes: deletions of the locus control region (LCR); missense mutation (p.Cys203Arg) in an L-/M-hybrid gene; and exon 3 single-nucleotide polymorphism (SNP) interchange haplotypes in an otherwise normal gene array. Moderate-to-high myopia was observed in all mutation categories. Individuals with LCR deletions or p.Cys203Arg mutations were more likely to have nystagmus and poor vision, with disease progression in some p.Cys203Arg patients. Three disease-associated exon 3 SNP haplotypes encoding LIAVA, LVAVA, or MIAVA were identified in our cohort. These patients were less likely to have nystagmus but more likely to show progression, with all patients over the age of 40 years having marked macular abnormalities. Previously, the haplotype LIAVA has been shown to result in exon 3 skipping. Here, we show that haplotypes LVAVA and MIAVA also result in aberrant splicing, with a residual low level of correctly spliced cone opsin. The OPN1LW/OPN1MW:c.532A>G SNP, common to all three disease-associated haplotypes, appears to be principally responsible for this mutational mechanism.


Asunto(s)
Opsinas de los Conos/genética , Estudios de Asociación Genética , Genotipo , Mutación , Fenotipo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Sustitución de Aminoácidos , Niño , Preescolar , Orden Génico , Silenciador del Gen , Enfermedades Genéticas Ligadas al Cromosoma X/diagnóstico , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Haplotipos , Hemicigoto , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Oftalmoscopios , Linaje , Polimorfismo de Nucleótido Simple , Empalme del ARN , Retinitis Pigmentosa/diagnóstico , Retinitis Pigmentosa/genética , Eliminación de Secuencia , Adulto Joven
7.
Hum Mol Genet ; 23(24): 6594-606, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25055872

RESUMEN

Mutations in rhodopsin, the light-sensitive protein of rod cells, are the most common cause of autosomal dominant retinitis pigmentosa (ADRP). Many rod opsin mutations, such as P23H, lead to misfolding of rod opsin with detrimental effects on photoreceptor function and viability. Misfolded P23H rod opsin and other mutations in the intradiscal domain are characterized by the formation of an incorrect disulphide bond between C185 and C187, as opposed to the correct and highly conserved C110-C187 disulphide bond. Therefore, we tested the hypothesis that incorrect disulphide bond formation might be a factor that affects the biogenesis of rod opsin by studying wild-type (WT) or P23H rod opsin in combination with amino acid substitutions that prevent the formation of incorrect disulphide bonds involving C185. These mutants had altered traffic dynamics, suggesting a requirement for regulation of disulphide bond formation/reduction during rod opsin biogenesis. Here, we show that the BiP co-chaperone and reductase protein ERdj5 (DNAJC10) regulates this process. ERdj5 overexpression promoted the degradation, improved the endoplasmic reticulum mobility and prevented the aggregation of P23H rod opsin. ERdj5 reduction by shRNA delayed rod opsin degradation and promoted aggregation. The reductase and co-chaperone activity of ERdj5 were both required for these effects on P23H rod opsin. Furthermore, mutations in these functional domains acted as dominant negatives that affected WT rod opsin biogenesis. Collectively, these data identify ERdj5 as a member of the proteostasis network that regulates rod opsin biogenesis and supports a role for disulphide bond formation/reduction in rod opsin biogenesis and disease.


Asunto(s)
Proteínas del Choque Térmico HSP40/genética , Chaperonas Moleculares/genética , Neuronas/metabolismo , Rodopsina/genética , Línea Celular Tumoral , Disulfuros/química , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica , Proteínas del Choque Térmico HSP40/antagonistas & inhibidores , Proteínas del Choque Térmico HSP40/metabolismo , Humanos , Chaperonas Moleculares/antagonistas & inhibidores , Chaperonas Moleculares/metabolismo , Mutación , Neuronas/citología , Plásmidos/química , Plásmidos/metabolismo , Agregado de Proteínas , Pliegue de Proteína , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Rodopsina/metabolismo , Transducción de Señal , Transfección
8.
Hum Mol Genet ; 23(8): 2164-75, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24301679

RESUMEN

The molecular chaperone Hsp90 is important for the functional maturation of many client proteins, and inhibitors are in clinical trials for multiple indications in cancer. Hsp90 inhibition activates the heat shock response and can improve viability in a cell model of the P23H misfolding mutation in rhodopsin that causes autosomal dominant retinitis pigmentosa (adRP). Here, we show that a single low dose of the Hsp90 inhibitor HSP990 enhanced visual function and delayed photoreceptor degeneration in a P23H transgenic rat model. This was associated with the induction of heat shock protein expression and reduced rhodopsin aggregation. We then investigated the effect of Hsp90 inhibition on a different type of rod opsin mutant, R135L, which is hyperphosphorylated, binds arrestin and disrupts vesicular traffic. Hsp90 inhibition with 17-AAG reduced the intracellular accumulation of R135L and abolished arrestin binding in cells. Hsf-1(-/-) cells revealed that the effect of 17-AAG on P23H aggregation was dependent on HSF-1, whereas the effect on R135L was HSF-1 independent. Instead, the effect on R135L was mediated by a requirement of Hsp90 for rhodopsin kinase (GRK1) maturation and function. Importantly, Hsp90 inhibition restored R135L rod opsin localization to wild-type (WT) phenotype in vivo in rat retina. Prolonged Hsp90 inhibition with HSP990 in vivo led to a posttranslational reduction in GRK1 and phosphodiesterase (PDE6) protein levels, identifying them as Hsp90 clients. These data suggest that Hsp90 represents a potential therapeutic target for different types of rhodopsin adRP through distinct mechanisms, but also indicate that sustained Hsp90 inhibition might adversely affect visual function.


Asunto(s)
Predisposición Genética a la Enfermedad , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Mutación/genética , Piridonas/farmacología , Pirimidinas/farmacología , Retinitis Pigmentosa/prevención & control , Rodopsina/metabolismo , Animales , Western Blotting , Células Cultivadas , Electrorretinografía , Femenino , Quinasa 1 del Receptor Acoplado a Proteína-G/genética , Quinasa 1 del Receptor Acoplado a Proteína-G/metabolismo , Genes Dominantes , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/efectos de los fármacos , Retina/metabolismo , Retina/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rodopsina/genética , Tomografía de Coherencia Óptica , Visión Ocular/efectos de los fármacos , Visión Ocular/fisiología
9.
PLoS One ; 8(8): e73944, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24023695

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by the selective loss of motor neurons in the spinal cord, brain stem, and motor cortex. Mutations in superoxide dismutase (SOD1) are associated with familial ALS and lead to SOD1 protein misfolding and aggregation. Here we show that the molecular chaperone, HSJ1 (DNAJB2), mutations in which cause distal hereditary motor neuropathy, can reduce mutant SOD1 aggregation and improve motor neuron survival in mutant SOD1 models of ALS. Overexpression of human HSJ1a (hHSJ1a) in vivo in motor neurons of SOD1(G93A) transgenic mice ameliorated disease. In particular, there was a significant improvement in muscle force, increased motor unit number and enhanced motor neuron survival. hHSJ1a was present in a complex with SOD1(G93A) and led to reduced SOD1 aggregation at late stages of disease progression. We also observed altered ubiquitin immunoreactivity in the double transgenic animals, suggesting that ubiquitin modification might be important for the observed improvements. In a cell model of SOD1(G93A) aggregation, HSJ1a preferentially bound to mutant SOD1, enhanced SOD1 ubiquitylation and reduced SOD1 aggregation in a J-domain and ubiquitin interaction motif (UIM) dependent manner. Collectively, the data suggest that HSJ1a acts on mutant SOD1 through a combination of chaperone, co-chaperone and pro-ubiquitylation activity. These results show that targeting SOD1 protein misfolding and aggregation in vivo can be neuroprotective and suggest that manipulation of DnaJ molecular chaperones might be useful in the treatment of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Proteínas del Choque Térmico HSP40/metabolismo , Chaperonas Moleculares/metabolismo , Fármacos Neuroprotectores/metabolismo , Superóxido Dismutasa/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Peso Corporal , Bovinos , Supervivencia Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Longevidad , Masculino , Ratones , Ratones Transgénicos , Modelos Biológicos , Neuronas Motoras/patología , Músculos/fisiopatología , Tamaño de los Órganos , Estructura Cuaternaria de Proteína , Médula Espinal/metabolismo , Médula Espinal/patología , Médula Espinal/fisiopatología , Superóxido Dismutasa/química , Superóxido Dismutasa/metabolismo , Ubiquitinación
10.
Mol Biol Cell ; 23(18): 3522-31, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22855534

RESUMEN

Mutations in rod opsin-the light-sensitive protein of rod cells-cause retinitis pigmentosa. Many rod opsin mutations lead to protein misfolding, and therefore it is important to understand the role of molecular chaperones in rod opsin biogenesis. We show that BiP (HSPA5) prevents the aggregation of rod opsin. Cleavage of BiP with the subtilase cytotoxin SubAB results in endoplasmic reticulum (ER) retention and ubiquitylation of wild-type (WT) rod opsin (WT-green fluorescent protein [GFP]) at the ER. Fluorescence recovery after photobleaching reveals that WT-GFP is usually mobile in the ER. By contrast, depletion of BiP activity by treatment with SubAB or coexpression of a BiP ATPase mutant, BiP(T37G), decreases WT-GFP mobility to below that of the misfolding P23H mutant of rod opsin (P23H-GFP), which is retained in the ER and can form cytoplasmic ubiquitylated inclusions. SubAB treatment of P23H-GFP-expressing cells decreases the mobility of the mutant protein further and leads to ubiquitylation throughout the ER. Of interest, BiP overexpression increases the mobility of P23H-GFP, suggesting that it can reduce mutant rod opsin aggregation. Therefore inhibition of BiP function results in aggregation of rod opsin in the ER, which suggests that BiP is important for maintaining the solubility of rod opsin in the ER.


Asunto(s)
Citoplasma/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de Choque Térmico/metabolismo , Opsinas de Bastones/metabolismo , Western Blotting , Línea Celular Tumoral , Chaperón BiP del Retículo Endoplásmico , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/farmacología , Recuperación de Fluorescencia tras Fotoblanqueo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Choque Térmico/genética , Humanos , Microscopía Confocal , Mutación , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Opsinas de Bastones/genética , Subtilisinas/metabolismo , Subtilisinas/farmacología , Transfección , Ubiquitinación/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos
11.
Hum Mol Genet ; 21(16): 3647-54, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22619378

RESUMEN

X-linked retinitis pigmentosa (XLRP) is genetically heterogeneous with two causative genes identified, RPGR and RP2. We previously mapped a locus for a severe form of XLRP, RP23, to a 10.71 Mb interval on Xp22.31-22.13 containing 62 genes. Candidate gene screening failed to identify a causative mutation, so we adopted targeted genomic next-generation sequencing of the disease interval to determine the molecular cause of RP23. No coding variants or variants within or near splice sites were identified. In contrast, a variant deep within intron 9 of OFD1 increased the splice site prediction score 4 bp upstream of the variant. Mutations in OFD1 cause the syndromic ciliopathies orofaciodigital syndrome-1, which is male lethal, Simpson-Golabi-Behmel syndrome type 2 and Joubert syndrome. We tested the effect of the IVS9+706A>G variant on OFD1 splicing in vivo. In RP23 patient-derived RNA, we detected an OFD1 transcript with the insertion of a cryptic exon spliced between exons 9 and 10 causing a frameshift, p.N313fs.X330. Correctly spliced OFD1 was also detected in patient-derived RNA, although at reduced levels (39%), hence the mutation is not male lethal. Our data suggest that photoreceptors are uniquely susceptible to reduced expression of OFD1 and that an alternative disease mechanism can cause XLRP. This disease mechanism of reduced expression for a syndromic ciliopathy gene causing isolated retinal degeneration is reminiscent of CEP290 intronic mutations that cause Leber congenital amaurosis, and we speculate that reduced dosage of correctly spliced ciliopathy genes may be a common disease mechanism in retinal degenerations.


Asunto(s)
Mutación del Sistema de Lectura , Proteínas/genética , Retinitis Pigmentosa/etiología , Secuencia de Aminoácidos , Secuencia de Bases , Cromosomas Humanos X , Exones , Humanos , Intrones , Masculino , Datos de Secuencia Molecular , Sitios de Empalme de ARN , Retinitis Pigmentosa/genética , Análisis de Secuencia de ADN
12.
PLoS One ; 7(2): e30866, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22347407

RESUMEN

Mutations in AIPL1 cause the inherited blindness Leber congenital amaurosis (LCA). AIPL1 has previously been shown to interact with NUB1, which facilitates the proteasomal degradation of proteins modified with the ubiquitin-like protein FAT10. Here we report that AIPL1 binds non-covalently to free FAT10 and FAT10ylated proteins and can form a ternary complex with FAT10 and NUB1. In addition, AIPL1 antagonised the NUB1-mediated degradation of the model FAT10 conjugate, FAT10-DHFR, and pathogenic mutations of AIPL1 were defective in inhibiting this degradation. While all AIPL1 mutants tested still bound FAT10-DHFR, there was a close correlation between the ability of the mutants to interact with NUB1 and their ability to prevent NUB1-mediated degradation. Interestingly, AIPL1 also co-immunoprecipitated the E1 activating enzyme for FAT10, UBA6, suggesting AIPL1 may have a role in directly regulating the FAT10 conjugation machinery. These studies are the first to implicate FAT10 in retinal cell biology and LCA pathogenesis, and reveal a new role of AIPL1 in regulating the FAT10 pathway.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas del Ojo/fisiología , Ubiquitinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Línea Celular Tumoral , Humanos , Amaurosis Congénita de Leber , Mutación , Retina/patología , Factores de Transcripción
14.
Am J Hum Genet ; 87(1): 26-39, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20579627

RESUMEN

X-linked cone and cone-rod dystrophies (XLCOD and XLCORD) are a heterogeneous group of progressive disorders that solely or primarily affect cone photoreceptors. Mutations in exon ORF15 of the RPGR gene are the most common underlying cause. In a previous study, we excluded RPGR exon ORF15 in some families with XLCOD. Here, we report genetic mapping of XLCOD to Xq26.1-qter. A significant LOD score was detected with marker DXS8045 (Z(max) = 2.41 [theta = 0.0]). The disease locus encompasses the cone opsin gene array on Xq28. Analysis of the array revealed a missense mutation (c. 529T>C [p. W177R]) in exon 3 of both the long-wavelength-sensitive (LW, red) and medium-wavelength-sensitive (MW, green) cone opsin genes that segregated with disease. Both exon 3 sequences were identical and were derived from the MW gene as a result of gene conversion. The amino acid W177 is highly conserved in visual and nonvisual opsins across species. We show that W177R in MW opsin and the equivalent W161R mutation in rod opsin result in protein misfolding and retention in the endoplasmic reticulum. We also demonstrate that W177R misfolding, unlike the P23H mutation in rod opsin that causes retinitis pigmentosa, is not rescued by treatment with the pharmacological chaperone 9-cis-retinal. Mutations in the LW/MW cone opsin gene array can, therefore, lead to a spectrum of disease, ranging from color blindness to progressive cone dystrophy (XLCOD5).


Asunto(s)
Opsinas de los Conos/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Células Fotorreceptoras Retinianas Conos/patología , Enfermedades de la Retina/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Cromosomas Humanos X/genética , Femenino , Estudios de Asociación Genética , Ligamiento Genético , Sitios Genéticos , Haplotipos , Humanos , Escala de Lod , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación Missense , Linaje , Estructura Secundaria de Proteína , Enfermedades de la Retina/patología , Enfermedades de la Retina/fisiopatología
15.
Hum Mol Genet ; 19(12): 2421-32, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20332100

RESUMEN

Nance-Horan syndrome (NHS) is an X-linked developmental disorder, characterized by bilateral congenital cataracts, dental anomalies, facial dysmorphism and mental retardation. Null mutations in a novel gene, NHS, cause the syndrome. The NHS gene appears to have multiple isoforms as a result of alternative transcription, but a cellular function for the NHS protein has yet to be defined. We describe NHS as a founder member of a new protein family (NHS, NHSL1 and NHSL2). Here, we demonstrate that NHS is a novel regulator of actin remodelling and cell morphology. NHS localizes to sites of cell-cell contact, the leading edge of lamellipodia and focal adhesions. The N-terminus of isoforms NHS-A and NHS-1A, implicated in the pathogenesis of NHS, have a functional WAVE homology domain that interacts with the Abi protein family, haematopoietic stem/progenitor cell protein 300 (HSPC300), Nap1 and Sra1. NHS knockdown resulted in the disruption of the actin cytoskeleton. We show that NHS controls cell morphology by maintaining the integrity of the circumferential actin ring and controlling lamellipod formation. NHS knockdown led to a striking increase in cell spreading. Conversely, ectopic overexpression of NHS inhibited lamellipod formation. Remodelling of the actin cytoskeleton and localized actin polymerization into branched actin filaments at the plasma membrane are essential for mediating changes in cell shape, migration and cell contact. Our data identify NHS as a new regulator of actin remodelling. We suggest that NHS orchestrates actin regulatory protein function in response to signalling events during development.


Asunto(s)
Actinas/metabolismo , Adhesiones Focales/metabolismo , Proteínas Nucleares/metabolismo , Seudópodos/metabolismo , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Secuencia de Aminoácidos , Animales , Células CACO-2 , Citoesqueleto/metabolismo , Adhesiones Focales/ultraestructura , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Estructura Terciaria de Proteína/genética , Seudópodos/ultraestructura , Ratas , Familia de Proteínas del Síndrome de Wiskott-Aldrich/genética
16.
J Cell Sci ; 122(Pt 24): 4465-72, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19934218

RESUMEN

Mutations in rod opsin, the archetypal G-protein-coupled receptor, cause retinitis pigmentosa. The majority of mutations, e.g. P23H, cause protein misfolding, resulting in ER retention, induction of the unfolded protein response and degradation by ERAD. If misfolded rod opsin escapes degradation, it aggregates and forms intracellular inclusions. Therefore, it is important to identify the chaperones that mediate the folding or degradation of rod opsin. ER degradation enhancing alpha-mannosidase-like 1 (EDEM1) can enhance the release of terminally misfolded glycoproteins from the calnexin chaperone system. Here, we identify EDEM1 as a novel chaperone of rod opsin. EDEM1 expression promoted the degradation of P23H rod opsin and decreased its aggregation. By contrast, shRNA-mediated knockdown of EDEM1 increased both the amount of P23H rod opsin and its aggregation into inclusions. EDEM1 was detected in rod photoreceptor inner segments and EndoH-sensitive rod opsin co-immunoprecipitated with EDEM1 from retina, suggesting that rod opsin is a physiological EDEM1 client. Unexpectedly, EDEM1 binding to rod opsin was independent of mannose trimming and EDEM1 promoted the cell-surface expression of mutant rod opsin. Collectively, the data suggest that EDEM1 is a chaperone for rod opsin and that expression of EDEM1 can be used to promote correct folding, as well as enhanced degradation, of mutant proteins in the ER to combat protein-misfolding disease.


Asunto(s)
Proteínas de la Membrana/metabolismo , Procesamiento Proteico-Postraduccional , Rodopsina/metabolismo , Humanos , Proteínas de la Membrana/genética , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Pliegue de Proteína , Transporte de Proteínas , Retina/metabolismo , Rodopsina/química , Rodopsina/genética
17.
Mol Vis ; 15: 876-84, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19421413

RESUMEN

PURPOSE: To perform a phenotypic assessment of members of three British families with blue cone monochromatism (BCM), and to determine the underlying molecular genetic basis of disease. METHODS: Affected members of three British families with BCM were examined clinically and underwent detailed electrophysiological and psychophysical testing. Blood samples were taken for DNA extraction. Molecular analysis involved the amplification of the coding regions of the long (L) and medium (M) wave cone opsin genes and the upstream locus control region (LCR) by polymerase chain reaction (PCR). Gene products were directly sequenced and analyzed. RESULTS: In all three families, genetic analysis identified that the underlying cause of BCM involved an unequal crossover within the opsin gene array, with an inactivating mutation. Family 1 had a single 5'-L-M-3' hybrid gene, with an inactivating Cys203Arg (C203R) mutation. Family 3 had an array composed of a C203R inactivated 5'-L-M-3' hybrid gene followed by a second inactive gene. Families 1 and 3 had typical clinical, electrophysiological, and psychophysical findings consistent with stationary BCM. A novel mutation was detected in Family 2 that had a single hybrid gene lacking exon 2. This family presented clinical and psychophysical evidence of a slowly progressive phenotype. CONCLUSIONS: Two of the BCM-causing family genotypes identified in this study comprised different hybrid genes, each of which contained the commonly described C203R inactivating mutation. The genotype in the family with evidence of a slowly progressive phenotype represents a novel BCM mutation. The deleted exon 2 in this family is not predicted to result in a shift in the reading frame, therefore we hypothesize that an abnormal opsin protein product may accumulate and lead to cone cell loss over time. This is the first report of slow progression associated with this class of mutation in the L or M opsin genes in BCM.


Asunto(s)
Defectos de la Visión Cromática/genética , Opsinas/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Electrorretinografía , Familia , Femenino , Eliminación de Gen , Silenciador del Gen , Humanos , Masculino , Persona de Mediana Edad , Mutación , Linaje , Fenotipo , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN , Reino Unido
18.
Hum Mol Genet ; 18(14): 2643-55, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19414485

RESUMEN

Nance-Horan syndrome (NHS) is an X-linked developmental disorder characterized by congenital cataract, dental anomalies, facial dysmorphism and, in some cases, mental retardation. Protein truncation mutations in a novel gene (NHS) have been identified in patients with this syndrome. We previously mapped X-linked congenital cataract (CXN) in one family to an interval on chromosome Xp22.13 which encompasses the NHS locus; however, no mutations were identified in the NHS gene. In this study, we show that NHS and X-linked cataract are allelic diseases. Two CXN families, which were negative for mutations in the NHS gene, were further analysed using array comparative genomic hybridization. CXN was found to be caused by novel copy number variations: a complex duplication-triplication re-arrangement and an intragenic deletion, predicted to result in altered transcriptional regulation of the NHS gene. Furthermore, we also describe the clinical and molecular analysis of seven families diagnosed with NHS, identifying four novel protein truncation mutations and a novel large deletion encompassing the majority of the NHS gene, all leading to no functional protein. We therefore show that different mechanisms, aberrant transcription of the NHS gene or no functional NHS protein, lead to different diseases. Our data highlight the importance of copy number variation and non-recurrent re-arrangements leading to different severity of disease and describe the potential mechanisms involved.


Asunto(s)
Catarata/genética , Genes Ligados a X , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Proteínas Nucleares/genética , Adulto , Secuencia de Bases , Catarata/congénito , Catarata/metabolismo , Niño , Preescolar , Femenino , Dosificación de Gen , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Humanos , Recién Nacido , Masculino , Proteínas de la Membrana , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Linaje , Adulto Joven
19.
Invest Ophthalmol Vis Sci ; 45(8): 2786-94, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15277505

RESUMEN

PURPOSE: To use porcine lens capsule (PLC) as basement membrane for ARPE-19 cells and to characterize its effects on cell differentiation and gene expression. METHODS: Postconfluent cultures of ARPE-19 cells were established on either porous polyester filters or PLC membranes and characterized by electron microscopy, immunocytochemistry, and transepithelial electrical resistance measurements. Metabolic activity was assessed by measuring phagocytosis of rod outer segments. mRNA populations of ARPE-19 cells grown on polyester and PLC membranes were compared by suppressive subtractive hybridization. Differentially regulated messages were subsequently identified by DNA sequencing and their altered expression confirmed by Northern or virtual Northern blot analysis. RESULTS: Culture of ARPE-19 cells on PLC membrane induced the formation of apical microvilli and the ability to phagocytose rod outer segments. These culture conditions also led to enhanced junctional distribution of ZO-1 and occludin, the formation of polarized membrane domains, and a significant increase in transepithelial resistance. Gene expression was significantly altered by growth on PLC membranes and 29 differentially expressed transcripts were identified. CONCLUSIONS: Culture of ARPE-19 cells on PLC membranes resulted in a more differentiated phenotype and in expression of a specific set of transcripts encoding protein products that may affect epithelial differentiation, polarity and survival.


Asunto(s)
Membrana Basal/fisiología , Proteínas del Ojo/genética , Regulación de la Expresión Génica/fisiología , Cápsula del Cristalino/fisiología , Epitelio Pigmentado Ocular/citología , Animales , Northern Blotting , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Línea Celular , Polaridad Celular/fisiología , Supervivencia Celular/fisiología , Conductividad Eléctrica , Proteínas del Ojo/metabolismo , Biblioteca de Genes , Humanos , Inmunohistoquímica , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microscopía Confocal , Microscopía Electrónica , Ocludina , Fagocitosis/fisiología , Fenotipo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Epitelio Pigmentado Ocular/metabolismo , ARN Mensajero/metabolismo , Segmento Externo de la Célula en Bastón/fisiología , Porcinos , Proteína de la Zonula Occludens-1
20.
Invest Ophthalmol Vis Sci ; 45(2): 675-84, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14744914

RESUMEN

PURPOSE: Oxidative stress has been implicated in the pathogenesis of age-related macular degeneration. The cell line ARPE-19 was therefore examined for response to oxidative stress and its effect on stress protein induction and junctional integrity. METHODS: ARPE-19 cell viability after 1 week or 5 weeks in culture was assessed in response to different concentrations of hydrogen peroxide. The response to sublethal doses was assessed by examination of heme oxygenase (HO)-1, Hsp27 and Hsp70 by immunofluorescence and Western blot analysis. Immunofluorescence was used to investigate the localization of the junctional proteins zonula occludens (ZO)-1, occludin, and N-cadherin, and beta-catenin. Subcellular fractionation was used to assess any redistribution of beta-catenin. Monolayer integrity was examined by measurement of flux of rhodamine-conjugated dextrans from the apical to basal aspect of cells. RESULTS: ARPE-19 cells cultured for 5 weeks were less sensitive to chronic oxidative stress induced by hydrogen peroxide than those cultured for 1 week. The more differentiated ARPE-19 cells had higher steady state levels of Hsp27 and Hsp70. The response to stress also differed with time in culture. The localization of junctional proteins, which became strongly peripheral after 5 weeks in culture, became disrupted after oxidative stress, and cytosolic beta-catenin increased. Chronic oxidative stress also increased paracellular flux across the monolayer. CONCLUSIONS: Increased resistance to chronic oxidative stress with differentiation in ARPE-19 cells correlated with higher steady state levels of Hsp27 and Hsp70. Oxidative stress disrupted RPE cell junction and barrier integrity, which may contribute to the pathogenesis of diseases related to RPE through disruption of the blood-retinal barrier.


Asunto(s)
Proteínas de Choque Térmico , Estrés Oxidativo , Epitelio Pigmentado Ocular/patología , Uniones Estrechas/patología , Barrera Hematorretinal/efectos de los fármacos , Western Blotting , Cadherinas/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Proteínas del Citoesqueleto/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Proteínas de Choque Térmico HSP27 , Proteínas HSP70 de Choque Térmico/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1 , Humanos , Peróxido de Hidrógeno/toxicidad , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares , Proteínas de Neoplasias/metabolismo , Ocludina , Oxidantes/toxicidad , Fosfoproteínas/metabolismo , Epitelio Pigmentado Ocular/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Uniones Estrechas/metabolismo , Transactivadores/metabolismo , Proteína de la Zonula Occludens-1 , beta Catenina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA