Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 29(22): 3706-3716, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33355362

RESUMEN

Mutations in retinitis pigmentosa GTPase regulator (RPGR) cause severe retinal ciliopathy, X-linked retinitis pigmentosa. Although two major alternatively spliced isoforms, RPGRex1-19 and RPGRORF15, are expressed, the relative importance of these isoforms in disease pathogenesis is unclear. Here, we analyzed fibroblast samples from eight patients and found that all of them form longer cilia than normal controls, albeit to different degrees. Although all mutant RPGRORF15 messenger RNAs (mRNAs) are unstable, their steady-state levels were similar or higher than those in the control cells, suggesting there may be increased transcription. Three of the fibroblasts that had higher levels of mutant RPGRORF15 mRNA also exhibited significantly higher levels of RPGRex1-19 mRNA. Four samples with unaltered RPGRex1-19 levels carried mutations in RPGRORF15 that resulted in this isoform being relatively less stable. Thus, in all cases, the RPGRex1-19/RPGRORF15 isoform ratio was increased, and this was highly correlative to the cilia extension defect. Moreover, overexpression of RPGRex1-19 (mimicking the increase in RPGRex1-19 to RPGRORF15 isoform ratio) or RPGRORF15 (mimicking reduction of the ratio) resulted in significantly longer or shorter cilia, respectively. Notably, the cilia length defect appears to be attributable to both the loss of the wild-type RPGRORF15 protein and to the higher levels of the RPGRex1-19 isoform, indicating that the observed defect is due to the altered isoform ratios. These results suggest that maintaining the optimal RPGRex1-9 to RPGRORF15 ratio is critical for cilia growth and that designing strategies that focus on the best ways to restore the RPGRex1-19/RPGRORF15 ratio may lead to better therapeutic outcomes.


Asunto(s)
Proteínas del Ojo/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Retinitis Pigmentosa/genética , Empalme Alternativo/genética , Proteínas Portadoras/genética , Cilios/genética , Cilios/patología , Exones/genética , Femenino , Fibroblastos , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Humanos , Masculino , Mutación/genética , Isoformas de Proteínas/genética , Retina/metabolismo , Retina/patología , Retinitis Pigmentosa/patología
2.
Hum Mol Genet ; 25(20): 4533-4545, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28172980

RESUMEN

Ciliary trafficking defects underlie the pathogenesis of severe human ciliopathies, including Joubert Syndrome (JBTS), Bardet-Biedl Syndrome, and some forms of retinitis pigmentosa (RP). Mutations in the ciliary protein RPGR (retinitis pigmentosa GTPase regulator) are common causes of RP-associated photoreceptor degeneration worldwide. While previous work has suggested that the localization of RPGR to cilia is critical to its functions, the mechanism by which RPGR and its associated cargo are trafficked to the cilia is unclear. Using proteomic and biochemical approaches, we show that RPGR interacts with two JBTS-associated ciliary proteins: PDE6δ (delta subunit of phosphodiesterase; a prenyl-binding protein) and INPP5E (inositol polyphosphate-5-phosphatase 5E). We find that PDE6δ binds selectively to the C-terminus of RPGR and that this interaction is critical for RPGR's localization to cilia. Furthermore, we show that INPP5E associates with the N-terminus of RPGR and trafficking of INPP5E to cilia is dependent upon the ciliary localization of RPGR. These results implicate prenylation of RPGR as a critical modification for its localization to cilia and, in turn suggest that trafficking of INPP5E to cilia depends upon the interaction of RPGR with PDE6δ. Finally, our results implicate INPP5E, a novel RPGR-interacting protein, in the pathogenesis of RPGR-associated ciliopathies.


Asunto(s)
Cilios/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Proteínas del Ojo/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Anomalías Múltiples/metabolismo , Animales , Proteínas Portadoras/metabolismo , Línea Celular , Cerebelo/anomalías , Cerebelo/metabolismo , Anomalías del Ojo/metabolismo , Humanos , Enfermedades Renales Quísticas/metabolismo , Masculino , Ratones , Dominios y Motivos de Interacción de Proteínas , Prenilación de Proteína , Transporte de Proteínas , Retina/anomalías , Retina/metabolismo , Retinitis Pigmentosa/metabolismo
3.
Hum Mol Genet ; 25(10): 2005-2012, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26936822

RESUMEN

Mutations in RPGR (retinitis pigmentosa GTPase regulator) are the most common cause of X-linked RP, a severe blindness disorder. RPGR mutations result in clinically variable disease with early- to late-onset phenotypic presentation. Molecular mechanisms underlying such heterogeneity are unclear. Here we show that phenotypic expression of Rpgr-loss in mice is influenced genetically by the loss of Cep290, a human ciliopathy gene. We found that Rpgrko/Y mice with a heterozygous hypomorphic allele of Cep290 (Cep290rd16/+) but not of a heterozygous null allele of Cep290 (Cep290null/+) or of other ciliopathy genes, Rpgrip1, Nphp1, Nphp4 and Nphp5, exhibit relatively early onset (by 3 months of age) retinal degeneration and dysfunction when compared with the onset at ∼7 months of age in the Rpgrko/Y mice. We also observed disorganized photoreceptor outer-segment morphology and defective trafficking of opsins in the Rpgrko/Y::Cep290rd16/+ mice. Together with a physical interaction between RPGR and the C-terminal domain of CEP290, our data suggest that RPGR and CEP290 genetically interact and highlight the involvement of hypomorphic alleles of genes as potential modifiers of heterogeneous retinal ciliopathies.


Asunto(s)
Antígenos de Neoplasias/genética , Ciliopatías/genética , Proteínas del Ojo/genética , Proteínas de Neoplasias/genética , Degeneración Retiniana/genética , Alelos , Animales , Antígenos de Neoplasias/biosíntesis , Proteínas de Ciclo Celular , Cilios/genética , Cilios/patología , Ciliopatías/patología , Proteínas del Citoesqueleto , Modelos Animales de Enfermedad , Proteínas del Ojo/biosíntesis , Regulación de la Expresión Génica , Heterocigoto , Humanos , Ratones , Mutación , Proteínas de Neoplasias/biosíntesis , Células Fotorreceptoras/patología , Mapas de Interacción de Proteínas/genética , Retina/metabolismo , Retina/patología , Degeneración Retiniana/patología , Índice de Severidad de la Enfermedad
4.
Hum Mol Genet ; 25(7): 1345-56, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26908598

RESUMEN

It is unclear how genes, such as RPGR (retinitis pigmentosa guanine triphosphatase regulator) that are expressed in both rods and cones, cause variable disease pathogenesis. Using transcriptomic analysis, we show that loss of RPGR in a rod-dominant mouse retina (Rpgr(ko)) results in predominant alterations in genes involved in actin cytoskeletal dynamics, prior to onset of degeneration. We validated these findings and found an increase in activated RhoA-GTP levels and polymerized F-actin in the Rpgr(ko) mouse retina. To assess the effect of the loss of RPGR in the all-cone region of the human retina, we used Nrl(-/-) (neural retina leucine zipper) mice, to generate Rpgr(ko)::Nrl(-/-) double-knock-out (Rpgr-DKO) mice. These mice exhibited supranormal cone response to light and substantially retained retinal architecture. Transcriptomic analysis revealed predominant up-regulation of retinal pigmented epithelium (RPE)-specific genes associated with visual cycle, whereas fatty acid analysis showed mild decrease in docosahexaenoic acid in the retina of the Rpgr-DKO mice when compared with the Nrl(-/-) mice. Our data reveal new insights into distinct intracellular pathways that are involved in RPGR-associated rod and cone dysfunction and provide a platform to design new treatment modalities.


Asunto(s)
Proteínas Portadoras/genética , Proteínas del Ojo/genética , Eliminación de Gen , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Animales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología
5.
Hum Mol Genet ; 25(24): 5444-5459, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27798110

RESUMEN

Mutations in the ORF15 exon of the RPGR gene cause a common form of X-linked retinitis pigmentosa, which often results in severe loss of vision. In dogs and mice, gene augmentation therapy has been shown to arrest the progressive degeneration of rod and cone photoreceptors. However, the distribution of potentially treatable photoreceptors across the human retinas and the rate of degeneration are not known. Here, we have defined structural and functional features of the disease in 70 individuals with ORF15 mutations. We also correlated the features observed in patients with those of three Rpgr-mutant (Rpgr-ko, Rd9, and Rpgr-cko) mice. In patients, there was pronounced macular disease. Across the retina, rod and cone dysfunction showed a range of patterns and a spectrum of severity between individuals, but a high symmetry was observed between eyes of each individual. Genotype was not related to disease expression. In the Rpgr-ko mice, there were intra-retinal differences in rhodopsin and cone opsin trafficking. In Rd9 and Rpgr-cko mice, retinal degeneration showed inter-ocular symmetry. Longitudinal results in patients revealed localized rod and cone dysfunction with progression rates of 0.8 to 1.3 log per decade in sensitivity loss. Relatively retained rod and cone photoreceptors in mid- and far-peripheral temporal-inferior and nasal-inferior visual field regions should be good targets for future localized gene therapies in patients.


Asunto(s)
Proteínas del Ojo/genética , Degeneración Retiniana/genética , Retinosquisis/genética , Rodopsina/genética , Adolescente , Adulto , Anciano , Animales , Niño , Heterocigoto , Humanos , Ratones , Ratones Noqueados , Persona de Mediana Edad , Mutación , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Degeneración Retiniana/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Retinosquisis/patología , Rodopsina/metabolismo , Adulto Joven
6.
Adv Exp Med Biol ; 1074: 521-538, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721984

RESUMEN

This article summarizes the recent advances in our understanding of a major retinal disease gene RPGR (retinitis pigmentosa GTPase regulator), mutations in which are associated with majority of X-linked forms of retinal degenerations. A great deal of work has been done to uncover the ciliary localization of RPGR and its interacting proteins in the retina. However, the molecular mechanisms of action of RPGR in the photoreceptors are still unclear. Recent studies have begun to shed light on the intracellular pathways in which RPGR is likely involved. The deregulation of such pathways may underlie the pathogenesis of severe retinal degeneration associated with RPGR. With the recent advances in the gene augmentation therapy for RPGR-associated disease, there is a lot of excitement in the field. Patients with RPGR mutations, however, present with clinically heterogeneous manifestations. It is therefore imperative to examine the function of RPGR in detail, so that we can design patient-oriented therapeutic strategies for this disease.


Asunto(s)
Cilios/fisiología , Proteínas del Ojo/fisiología , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Transporte de Proteínas/fisiología , Retinitis Pigmentosa/genética , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Cilios/ultraestructura , Exones , Proteínas del Ojo/química , Proteínas del Ojo/genética , Pleiotropía Genética , Humanos , Ratones Noqueados , Modelos Biológicos , Proteínas de Unión al GTP Monoméricas/metabolismo , Complejos Multiproteicos , Dominios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Células Fotorreceptoras Retinianas Conos/fisiología , Células Fotorreceptoras Retinianas Bastones/fisiología , Células Fotorreceptoras Retinianas Bastones/ultraestructura , Retinitis Pigmentosa/patología , Transducción de Señal/genética
7.
Hum Mol Genet ; 24(22): 6446-58, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26358772

RESUMEN

Retinal neurodegenerative diseases are especially attractive targets for gene replacement therapy, which appears to be clinically effective for several monogenic diseases. X-linked forms of retinitis pigmentosa (XLRP) are relatively severe blinding disorders, resulting from progressive photoreceptor dysfunction primarily caused by mutations in RPGR or RP2 gene. With a goal to develop gene therapy for the XLRP-RP2 disease, we first performed detailed characterization of the Rp2-knockout (Rp2-KO) mice and observed early-onset cone dysfunction, which was followed by progressive cone degeneration, mimicking cone vision impairment in XLRP patients. The mice also exhibited distinct and significantly delayed falling phase of photopic b-wave of electroretinogram (ERG). Concurrently, we generated a self-complementary adeno-associated viral (AAV) vector carrying human RP2-coding sequence and demonstrated its ability to mediate stable RP2 protein expression in mouse photoreceptors. A long-term efficacy study was then conducted in Rp2-KO mice following AAV-RP2 vector administration. Preservation of cone function was achieved with a wide dose range over 18-month duration, as evidenced by photopic ERG and optomotor tests. The slower b-wave kinetics was also completely restored. Morphologically, the treatment preserved cone viability, corrected mis-trafficking of M-cone opsin and restored cone PDE6 expression. The therapeutic effect was achieved even in mice that received treatment at an advanced disease stage. The highest AAV-RP2 dose group demonstrated retinal toxicity, highlighting the importance of careful vector dosing in designing future human trials. The wide range of effective dose, a broad treatment window and long-lasting therapeutic effects should make the RP2 gene therapy attractive for clinical development.


Asunto(s)
Proteínas del Ojo/genética , Terapia Genética/métodos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Células Fotorreceptoras Retinianas Conos/fisiología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Animales , Electrorretinografía , Proteínas del Ojo/biosíntesis , Proteínas de Unión al GTP , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Vectores Genéticos , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Noqueados , Mutación , Pirofosfatasas/deficiencia , Pirofosfatasas/genética , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Degeneración Retiniana/genética , Retinitis Pigmentosa/metabolismo
8.
Hum Genet ; 136(8): 941-949, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28620713

RESUMEN

Primary congenital glaucoma (PCG) is a severe autosomal recessive ocular disorder associated with considerable clinical and genetic heterogeneity. Recently, rare heterozygous alleles in the angiopoietin receptor-encoding gene TEK were implicated in PCG. We undertook this study to ascertain the second mutant allele in a large cohort (n = 337) of autosomal recessive PCG cases that carried heterozygous TEK mutations. Our investigations revealed 12 rare heterozygous missense mutations in TEK by targeted sequencing. Interestingly, four of these TEK mutations (p.E103D, p.I148T, p.Q214P, and p.G743A) co-occurred with three heterozygous mutations in another major PCG gene CYP1B1 (p.A115P, p.E229K, and p.R368H) in five families. The parents of these probands harbored either of the heterozygous TEK or CYP1B1 alleles and were asymptomatic, indicating a potential digenic mode of inheritance. Furthermore, we ascertained the interactions of TEK and CYP1B1 by co-transfection and pull-down assays in HEK293 cells. Ligand responsiveness of the wild-type and mutant TEK proteins was assessed in HUVECs using immunofluorescence analysis. We observed that recombinant TEK and CYP1B1 proteins interact with each other, while the disease-associated allelic combinations of TEK (p.E103D)::CYP1B1 (p.A115P), TEK (p.Q214P)::CYP1B1 (p.E229K), and TEK (p.I148T)::CYP1B1 (p.R368H) exhibit perturbed interaction. The mutations also diminished the ability of TEK to respond to ligand stimulation, indicating perturbed TEK signaling. Overall, our data suggest that interaction of TEK and CYP1B1 contributes to PCG pathogenesis and argue that TEK-CYP1B1 may perform overlapping as well as distinct functions in manifesting the disease etiology.


Asunto(s)
Citocromo P-450 CYP1B1/genética , Glaucoma/congénito , Glaucoma/genética , Receptor TIE-2/genética , Alelos , Estudios de Cohortes , Citocromo P-450 CYP1B1/metabolismo , Femenino , Frecuencia de los Genes , Genoma Humano , Genómica , Células HEK293 , Haplotipos , Heterocigoto , Células Endoteliales de la Vena Umbilical Humana , Humanos , Desequilibrio de Ligamiento , Masculino , Mutación Missense , Linaje , Receptor TIE-2/metabolismo , Reproducibilidad de los Resultados , Alineación de Secuencia , Análisis de Secuencia de ADN
10.
Proc Natl Acad Sci U S A ; 109(6): 2132-7, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22308428

RESUMEN

Hereditary retinal blindness is caused by mutations in genes expressed in photoreceptors or retinal pigment epithelium. Gene therapy in mouse and dog models of a primary retinal pigment epithelium disease has already been translated to human clinical trials with encouraging results. Treatment for common primary photoreceptor blindness, however, has not yet moved from proof of concept to the clinic. We evaluated gene augmentation therapy in two blinding canine photoreceptor diseases that model the common X-linked form of retinitis pigmentosa caused by mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene, which encodes a photoreceptor ciliary protein, and provide evidence that the therapy is effective. After subretinal injections of adeno-associated virus-2/5-vectored human RPGR with human IRBP or GRK1 promoters, in vivo imaging showed preserved photoreceptor nuclei and inner/outer segments that were limited to treated areas. Both rod and cone photoreceptor function were greater in treated (three of four) than in control eyes. Histopathology indicated normal photoreceptor structure and reversal of opsin mislocalization in treated areas expressing human RPGR protein in rods and cones. Postreceptoral remodeling was also corrected: there was reversal of bipolar cell dendrite retraction evident with bipolar cell markers and preservation of outer plexiform layer thickness. Efficacy of gene therapy in these large animal models of X-linked retinitis pigmentosa provides a path for translation to human treatment.


Asunto(s)
Ceguera/genética , Ceguera/terapia , Enfermedades Genéticas Ligadas al Cromosoma X/terapia , Terapia Genética , Células Fotorreceptoras de Vertebrados/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Animales , Perros , Proteínas del Ojo/genética , Proteínas del Ojo/uso terapéutico , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Genotipo , Humanos , Ratones , Mutación/genética , Sistemas de Lectura Abierta/genética , Opsinas/metabolismo , Fenotipo , Transporte de Proteínas , Células Bipolares de la Retina/patología
11.
Nat Genet ; 38(6): 674-81, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16682973

RESUMEN

The molecular basis of nephronophthisis, the most frequent genetic cause of renal failure in children and young adults, and its association with retinal degeneration and cerebellar vermis aplasia in Joubert syndrome are poorly understood. Using positional cloning, we here identify mutations in the gene CEP290 as causing nephronophthisis. It encodes a protein with several domains also present in CENPF, a protein involved in chromosome segregation. CEP290 (also known as NPHP6) interacts with and modulates the activity of ATF4, a transcription factor implicated in cAMP-dependent renal cyst formation. NPHP6 is found at centrosomes and in the nucleus of renal epithelial cells in a cell cycle-dependent manner and in connecting cilia of photoreceptors. Abrogation of its function in zebrafish recapitulates the renal, retinal and cerebellar phenotypes of Joubert syndrome. Our findings help establish the link between centrosome function, tissue architecture and transcriptional control in the pathogenesis of cystic kidney disease, retinal degeneration, and central nervous system development.


Asunto(s)
Factor de Transcripción Activador 4/genética , Antígenos de Neoplasias/genética , Mutación , Proteínas de Neoplasias/genética , Animales , Proteínas de Ciclo Celular , Proteínas del Citoesqueleto , Femenino , Ligamiento Genético , Humanos , Hibridación in Situ , Masculino , Linaje , Síndrome , Pez Cebra
12.
Hum Mol Genet ; 21(15): 3333-44, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22543976

RESUMEN

Oculocerebral renal syndrome of Lowe (OCRL or Lowe syndrome), a severe X-linked congenital disorder characterized by congenital cataracts and glaucoma, mental retardation and kidney dysfunction, is caused by mutations in the OCRL gene. OCRL is a phosphoinositide 5-phosphatase that interacts with small GTPases and is involved in intracellular trafficking. Despite extensive studies, it is unclear how OCRL mutations result in a myriad of phenotypes found in Lowe syndrome. Our results show that OCRL localizes to the primary cilium of retinal pigment epithelial cells, fibroblasts and kidney tubular cells. Lowe syndrome-associated mutations in OCRL result in shortened cilia and this phenotype can be rescued by the introduction of wild-type OCRL; in vivo, knockdown of ocrl in zebrafish embryos results in defective cilia formation in Kupffer vesicles and cilia-dependent phenotypes. Cumulatively, our data provide evidence for a role of OCRL in cilia maintenance and suggest the involvement of ciliary dysfunction in the manifestation of Lowe syndrome.


Asunto(s)
Cilios/metabolismo , Síndrome Oculocerebrorrenal/metabolismo , Monoéster Fosfórico Hidrolasas/análisis , Monoéster Fosfórico Hidrolasas/genética , Animales , Cilios/química , Embrión no Mamífero/metabolismo , Fibroblastos/metabolismo , Genotipo , Humanos , Inmunohistoquímica , Túbulos Renales/metabolismo , Mutación , Síndrome Oculocerebrorrenal/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Transfección , Pez Cebra/embriología , Pez Cebra/metabolismo
13.
Nat Genet ; 37(3): 282-8, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15723066

RESUMEN

Nephronophthisis (NPHP) is the most frequent genetic cause of chronic renal failure in children. Identification of four genes mutated in NPHP subtypes 1-4 (refs. 4-9) has linked the pathogenesis of NPHP to ciliary functions. Ten percent of affected individuals have retinitis pigmentosa, constituting the renal-retinal Senior-Loken syndrome (SLSN). Here we identify, by positional cloning, mutations in an evolutionarily conserved gene, IQCB1 (also called NPHP5), as the most frequent cause of SLSN. IQCB1 encodes an IQ-domain protein, nephrocystin-5. All individuals with IQCB1 mutations have retinitis pigmentosa. Hence, we examined the interaction of nephrocystin-5 with RPGR (retinitis pigmentosa GTPase regulator), which is expressed in photoreceptor cilia and associated with 10-20% of retinitis pigmentosa. We show that nephrocystin-5, RPGR and calmodulin can be coimmunoprecipitated from retinal extracts, and that these proteins localize to connecting cilia of photoreceptors and to primary cilia of renal epithelial cells. Our studies emphasize the central role of ciliary dysfunction in the pathogenesis of SLSN.


Asunto(s)
Proteínas de Unión a Calmodulina/genética , Proteínas de Unión a Calmodulina/metabolismo , Calmodulina/metabolismo , Proteínas del Ojo/metabolismo , Mutación , Secuencia de Aminoácidos , Northern Blotting , Proteínas de Unión a Calmodulina/química , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Síndrome , Técnicas del Sistema de Dos Híbridos
14.
Hum Mol Genet ; 20(5): 975-87, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21159800

RESUMEN

We recently reported that mutations in the widely expressed nuclear protein TOPORS (topoisomerase I-binding arginine/serine rich) are associated with autosomal dominant retinal degeneration. However, the precise localization and a functional role of TOPORS in the retina remain unknown. Here, we demonstrate that TOPORS is a novel component of the photoreceptor sensory cilium, which is a modified primary cilium involved with polarized trafficking of proteins. In photoreceptors, TOPORS localizes primarily to the basal bodies of connecting cilium and in the centrosomes of cultured cells. Morpholino-mediated silencing of topors in zebrafish embryos demonstrates in another species a comparable retinal problem as seen in humans, resulting in defective retinal development and failure to form outer segments. These defects can be rescued by mRNA encoding human TOPORS. Taken together, our data suggest that TOPORS may play a key role in regulating primary cilia-dependent photoreceptor development and function. Additionally, it is well known that mutations in other ciliary proteins cause retinal degeneration, which may explain why mutations in TOPORS result in the same phenotype.


Asunto(s)
Centrosoma/metabolismo , Cilios/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Degeneración Retiniana/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Células Cultivadas , Cilios/genética , Humanos , Ratones , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Células Fotorreceptoras/metabolismo , Transporte de Proteínas , Retina/metabolismo , Degeneración Retiniana/genética , Ubiquitina-Proteína Ligasas/genética , Pez Cebra
15.
J Biol Chem ; 286(32): 28276-86, 2011 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-21685394

RESUMEN

Primary cilia regulate polarized protein trafficking in photoreceptors, which are dynamic and highly compartmentalized sensory neurons of retina. The ciliary protein Cep290 modulates cilia formation and is frequently mutated in syndromic and non-syndromic photoreceptor degeneration. However, the underlying mechanism of associated retinopathy is unclear. Using the Cep290 mutant mouse rd16 (retinal degeneration 16), we show that Cep290-mediated photoreceptor degeneration is associated with aberrant accumulation of its novel interacting partner Rkip (Raf-1 kinase inhibitory protein). This effect is phenocopied by morpholino-mediated depletion of cep290 in zebrafish. We further demonstrate that ectopic accumulation of Rkip leads to defective cilia formation in zebrafish and cultured cells, an effect mediated by its interaction with the ciliary GTPase Rab8A. Our data suggest that Rkip prevents cilia formation and is associated with Cep290-mediated photoreceptor degeneration. Furthermore, our results indicate that preventing accumulation of Rkip could potentially ameliorate such degeneration.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Trastornos de la Motilidad Ciliar/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Degeneración Retiniana/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Antígenos de Neoplasias/genética , Proteínas de Ciclo Celular , Chlorocebus aethiops , Cilios/genética , Cilios/metabolismo , Cilios/patología , Trastornos de la Motilidad Ciliar/genética , Trastornos de la Motilidad Ciliar/patología , Proteínas del Citoesqueleto , Células HEK293 , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas de Unión a Fosfatidiletanolamina/genética , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
16.
Hum Mol Genet ; 19(18): 3591-8, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20631154

RESUMEN

Defects in biogenesis or function(s) of primary cilia are associated with numerous inherited disorders (called ciliopathies) that may include retinal degeneration phenotype. The cilia-expressed gene RPGR (retinitis pigmentosa GTPase regulator) is mutated in patients with X-linked retinitis pigmentosa (XLRP) and encodes multiple protein isoforms with a common N-terminal domain homologous to regulator of chromosome condensation 1 (RCC1), a guanine nucleotide exchange factor (GEF) for Ran GTPase. RPGR interacts with several ciliopathy proteins, such as RPGRIP1L and CEP290; however, its physiological role in cilia-associated functions has not been delineated. Here, we report that RPGR interacts with the small GTPase RAB8A, which participates in cilia biogenesis and maintenance. We show that RPGR primarily associates with the GDP-bound form of RAB8A and stimulates GDP/GTP nucleotide exchange. Disease-causing mutations in RPGR diminish its interaction with RAB8A and reduce the GEF activity. Depletion of RPGR in hTERT-RPE1 cells interferes with ciliary localization of RAB8A and results in shorter primary cilia. Our data suggest that RPGR modulates intracellular localization and function of RAB8A. We propose that perturbation of RPGR-RAB8A interaction, at least in part, underlies the pathogenesis of photoreceptor degeneration in XLRP caused by RPGR mutations.


Asunto(s)
Cilios/metabolismo , Proteínas del Ojo/metabolismo , Células Fotorreceptoras/metabolismo , Retinitis Pigmentosa/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Línea Celular , Cilios/genética , Perros , Proteínas del Ojo/genética , Humanos , Unión Proteica , Transporte de Proteínas , Retinitis Pigmentosa/genética
17.
Hum Mol Genet ; 19(1): 90-8, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19815619

RESUMEN

Dysfunction of primary cilia is associated with tissue-specific or syndromic disorders. RPGR is a ciliary protein, mutations in which can lead to retinitis pigmentosa (RP), cone-rod degeneration, respiratory infections and hearing disorders. Though RPGR is implicated in ciliary transport, the pathogenicity of RPGR mutations and the mechanism of underlying phenotypic heterogeneity are still unclear. Here we have utilized genetic rescue studies in zebrafish to elucidate the effect of human disease-associated mutations on its function. We show that rpgr is expressed predominantly in the retina, brain and gut of zebrafish. In the retina, RPGR primarily localizes to the sensory cilium of photoreceptors. Antisense morpholino (MO)-mediated knockdown of rpgr function in zebrafish results in reduced length of Kupffer's vesicle (KV) cilia and is associated with ciliary anomalies including shortened body-axis, kinked tail, hydrocephaly and edema but does not affect retinal development. These phenotypes can be rescued by wild-type (WT) human RPGR. Several of the RPGR mutants can also reverse the MO-induced phenotype, suggesting their potential hypomorphic function. Notably, selected RPGR mutations observed in XLRP (T99N, E589X) or syndromic RP (T124fs, K190fs and L280fs) do not completely rescue the rpgr-MO phenotype, indicating a more deleterious effect of the mutation on the function of RPGR. We propose that RPGR is involved in cilia-dependent cascades during development in zebrafish. Our studies provide evidence for a heterogenic effect of the disease-causing mutations on the function of RPGR.


Asunto(s)
Cilios/metabolismo , Proteínas del Ojo/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Animales , Cilios/efectos de los fármacos , Cilios/patología , Embrión no Mamífero/anomalías , Embrión no Mamífero/metabolismo , Proteínas del Ojo/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Mutantes/metabolismo , Mutación/genética , Oligonucleótidos Antisentido/farmacología , Fenotipo , Pez Cebra/genética , Proteínas de Pez Cebra/genética
18.
Hum Mol Genet ; 19(22): 4330-44, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20729296

RESUMEN

Ciliopathies represent a growing group of human genetic diseases whose etiology lies in defects in ciliogenesis or ciliary function. Given the established entity of renal-retinal ciliopathies, we have been examining the role of cilia-localized proteins mutated in retinitis pigmentosa (RP) in regulating renal ciliogenesis or cilia-dependent signaling cascades. Specifically, this study examines the role of the RP2 gene product with an emphasis on renal and vertebrate development. We demonstrate that in renal epithelia, RP2 localizes to the primary cilium through dual acylation of the amino-terminus. We also show that RP2 forms a calcium-sensitive complex with the autosomal dominant polycystic kidney disease protein polycystin 2. Ablation of RP2 by shRNA promotes swelling of the cilia tip that may be a result of aberrant trafficking of polycystin 2 and other ciliary proteins. Morpholino-mediated repression of RP2 expression in zebrafish results in multiple developmental defects that have been previously associated with ciliary dysfunction, such as hydrocephalus, kidney cysts and situs inversus. Finally, we demonstrate that, in addition to our observed physical interaction between RP2 and polycystin 2, dual morpholino-mediated knockdown of polycystin 2 and RP2 results in enhanced situs inversus, indicating that these two genes also regulate a common developmental process. This work suggests that RP2 may be an important regulator of ciliary function through its association with polycystin 2 and provides evidence of a further link between retinal and renal cilia function.


Asunto(s)
Cilios/fisiología , Proteínas del Ojo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Canales Catiónicos TRPP/metabolismo , Acilación , Animales , Cilios/genética , Cilios/metabolismo , Proteínas de Unión al GTP , Técnicas de Silenciamiento del Gen , Humanos , Riñón/metabolismo , Riñón/fisiopatología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Mutación , Vertebrados/genética , Vertebrados/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
19.
Front Genet ; 13: 982127, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36061204

RESUMEN

Ciliopathies are a class of inherited severe human disorders that occur due to defective formation or function of cilia. The RPGRIP1L (retinitis pigmentosa GTPase regulator-interacting protein1-like) gene encodes for a ciliary protein involved in regulating cilia formation and function. Mutations in RPGRIP1L cause ciliopathies associated with severe embryonic defects, such as Meckel-Gruber Syndrome (MKS). Here we report RPGRIP1L mutation analysis in a family diagnosed with MKS. The clinical manifestations of the fetus included thoraco-lumbar open neural tube defect with associated Chiari type II malformation and hydrocephalus, bilateral club feet, and single right kidney/ureter. Analysis of the parental DNA samples revealed that the father carried a previously reported mutation R1236C/+ whereas the mother had a novel splice site mutation IVS6+1 G > A/+ in RPGRIP1L. The splice site mutation resulted in the exclusion of in-frame exon 6 of RPGRIP1L (RPGRIP1L-∆Ex6) but expressed a stable protein in fibroblasts derived from the parents' skin biopsies. The GFP-RPGRIP1L-∆Ex6 mutant protein exhibited relatively reduced ciliary localization in transiently-transfected cultured RPE-1 cells. Taken together, this study identifies a novel RPGRIP1L variant RPGRIP1L-∆Ex6, which in combination with RPGRIP1L-R1236C is associated with MKS. We also suggest that the deletion of exon 6 of RPGRIP1L leads to reduced ciliary localization of RPGRIP1L, indicating a plausible mechanism of associated disease.

20.
Ind Psychiatry J ; 30(2): 353-355, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35017824

RESUMEN

There is minimal literature and case reports on cultural concepts of distress, especially on ghost sickness. The aim of this article is to educate clinicians to culture-bound syndromes/cultural concepts of distress when assessing people from culturally and linguistically diverse backgrounds. This case report describes an elderly male of Papua New Guinea who presented with psychotic symptoms, shaped by the cultural concept of distress from experiencing loss. This paper describes the importance of utilizing of cultural liaison officers to provide culturally-informed care and before diagnosing a patient with the culture concept of distress, every effort should be made to thoroughly investigate to exclude an organic cause for the presentation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA