Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Mol Ther Methods Clin Dev ; 23: 448-459, 2021 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-34786437

RESUMEN

Split intein-mediated protein trans-splicing expands AAV transfer capacity, thus overcoming the limited AAV cargo. However, non-mammalian inteins persist as trans-splicing by-products, and this could raise safety concerns for AAV intein clinical applications. In this study, we tested the ability of several degrons to selectively decrease levels of inteins after protein trans-splicing and found that a version of E. coli dihydrofolate reductase, which we have shortened to better fit into the AAV vector, is the most effective. We show that subretinal administration of AAV intein armed with this short degron is both safe and effective in a mouse model of Stargardt disease (STGD1), which is the most common form of inherited macular degeneration in humans. This supports the use of optimized AAV intein for gene therapy of both STGD1 and other conditions that require transfer of large genes.

2.
Am J Hum Genet ; 108(2): 295-308, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33508235

RESUMEN

Retinitis pigmentosa (RP) is a group of progressive retinal degenerations of mostly monogenic inheritance, which cause blindness in about 1:3,500 individuals worldwide. Heterozygous variants in the rhodopsin (RHO) gene are the most common cause of autosomal dominant RP (adRP). Among these, missense variants at C-terminal proline 347, such as p.Pro347Ser, cause severe adRP recurrently in European affected individuals. Here, for the first time, we use CRISPR/Cas9 to selectively target the p.Pro347Ser variant while preserving the wild-type RHO allele in vitro and in a mouse model of adRP. Detailed in vitro, genomic, and biochemical characterization of the rhodopsin C-terminal editing demonstrates a safe downregulation of p.Pro347Ser expression leading to partial recovery of photoreceptor function in a transgenic mouse model treated with adeno-associated viral vectors. This study supports the safety and efficacy of CRISPR/Cas9-mediated allele-specific editing and paves the way for a permanent and precise correction of heterozygous variants in dominantly inherited retinal diseases.


Asunto(s)
Edición Génica , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Rodopsina/genética , Alelos , Animales , Sistemas CRISPR-Cas , Línea Celular , Dependovirus/genética , Modelos Animales de Enfermedad , Electrorretinografía , Terapia Genética , Humanos , Mutación INDEL , Ratones , Ratones Transgénicos , Mutación Missense , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/metabolismo , Retina/fisiopatología , Rodopsina/metabolismo
3.
Sci Transl Med ; 11(492)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092694

RESUMEN

Retinal gene therapy with adeno-associated viral (AAV) vectors holds promises for treating inherited and noninherited diseases of the eye. Although clinical data suggest that retinal gene therapy is safe and effective, delivery of large genes is hindered by the limited AAV cargo capacity. Protein trans-splicing mediated by split inteins is used by single-cell organisms to reconstitute proteins. Here, we show that delivery of multiple AAV vectors each encoding one of the fragments of target proteins flanked by short split inteins results in protein trans-splicing and full-length protein reconstitution in the retina of mice and pigs and in human retinal organoids. The reconstitution of large therapeutic proteins using this approach improved the phenotype of two mouse models of inherited retinal diseases. Our data support the use of split intein-mediated protein trans-splicing in combination with AAV subretinal delivery for gene therapy of inherited blindness due to mutations in large genes.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Inteínas , Retina/virología , Trans-Empalme/genética , Animales , Vectores Genéticos/administración & dosificación , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Organoides/ultraestructura , Organoides/virología , Fenotipo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/virología , Porcinos
4.
EMBO Mol Med ; 11(5)2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30979712

RESUMEN

Mitochondrial diseases (MDs) are a heterogeneous group of devastating and often fatal disorders due to defective oxidative phosphorylation. Despite the recent advances in mitochondrial medicine, effective therapies are still not available for these conditions. Here, we demonstrate that the microRNAs miR-181a and miR-181b (miR-181a/b) regulate key genes involved in mitochondrial biogenesis and function and that downregulation of these miRNAs enhances mitochondrial turnover in the retina through the coordinated activation of mitochondrial biogenesis and mitophagy. We thus tested the effect of miR-181a/b inactivation in different animal models of MDs, such as microphthalmia with linear skin lesions and Leber's hereditary optic neuropathy. We found that miR-181a/b downregulation strongly protects retinal neurons from cell death and significantly ameliorates the disease phenotype in all tested models. Altogether, our results demonstrate that miR-181a/b regulate mitochondrial homeostasis and that these miRNAs may be effective gene-independent therapeutic targets for MDs characterized by neuronal degeneration.


Asunto(s)
Regulación hacia Abajo/genética , MicroARNs/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/genética , Animales , Autofagia/genética , Muerte Celular , Línea Celular , Citoprotección , Modelos Animales de Enfermedad , Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/metabolismo , Femenino , Humanos , Masculino , Ratones , MicroARNs/genética , Mitocondrias/ultraestructura , Enfermedades Mitocondriales/patología , Dinámicas Mitocondriales/genética , Modelos Biológicos , Biogénesis de Organelos , Oryzias , Fenotipo , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología
6.
Hum Gene Ther ; 26(4): 193-200, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25762209

RESUMEN

Because of its favorable anatomical and immunological characteristics, the eye has been at the forefront of translational gene therapy. Dozens of promising proofs of concept have been obtained in animal models of inherited retinal degenerations (IRDs), and some of them have been relayed to the clinic. The results from the first clinical trials for a congenital form of blindness have generated great interest and have demonstrated the safety and efficacy of intraocular administrations of viral vectors in humans. However, this progress has also generated new questions and posed challenges that need to be addressed to further expand the applicability of gene therapy in the eye, including safe delivery of viral vectors to the outer retina, treatment of dominant IRDs as well as of IRDs caused by mutations in large genes, and, finally, selection of the appropriate IRDs and patients to maximize the efficacy of gene transfer. This review summarizes the strategies that are currently being exploited to overcome these challenges and drive the clinical development of retinal gene therapy.


Asunto(s)
Terapia Genética , Degeneración Retiniana/terapia , Animales , Ensayos Clínicos como Asunto , Vectores Genéticos , Humanos , Retina/metabolismo , Retina/patología , Degeneración Retiniana/genética , Rodopsina/genética , Rodopsina/metabolismo , Transducción Genética
7.
Lab Anim ; 48(4): 345-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24968696

RESUMEN

The collection of cerebrospinal fluid is necessary in order to determine its composition. It can then be used to diagnose various diseases. The aim of the study was to develop and optimize a technique for performing safe centesis for the collection of cerebrospinal fluid in piglets and its injection through the cisterna magna. The study was divided into three phases: (1) anatomical study of cadavers, (2) in vivo application of the technique and (3) observation of recovery time. The proposed technique resulted in a safe puncture of the cisterna magna. The authors identified and confirmed the correspondence of the crista occipitalis and the wings of the atlas with the external landmarks on the cadaver by means of direct radiological visualization. The punctures were performed successfully at the first attempt in 11 out of 12 anaesthetized piglets. The technique herein described provides a reproducible safe and easy route for approaching the cisterna magna for cerebrospinal fluid collection, drug administration and gene delivery.


Asunto(s)
Líquido Cefalorraquídeo , Cisterna Magna , Ventriculostomía/veterinaria , Animales , Femenino , Masculino , Punciones , Sus scrofa , Ventriculostomía/instrumentación
8.
Hum Gene Ther ; 24(12): 982-92, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24067103

RESUMEN

Gene transfer to both cone and rod photoreceptors (PRs) is essential for gene therapy of inherited retinal degenerations that are caused by mutations in genes expressed in both PR types. Vectors based on the adeno-associated virus (AAV) efficiently transduce PRs of different species. However, these are predominantly rods and little is known about the ability of the AAV to transduce cones in combination with rods. Here we show that AAV2/8 transduces pig cones to levels that are similar to AAV2/9, and the outer nuclear layer (mainly rods) to levels that are on average higher, although not statistically significant, than both AAV2/5 and AAV2/9. We additionally found that the ubiquitous cytomegalovirus (CMV), but not the PR-specific GRK1 promoter, transduced pig cones efficiently, presumably because GRK1 is not expressed in pig cones as observed in mice and humans. Indeed, the GRK1 and CMV promoters transduce a similar percentage of murine cones with the CMV reaching the highest expression levels. Consistent with this, the AAV2/8 vectors with either the CMV or the GRK1 promoter restore cone function in a mouse model of Leber congenital amaurosis type 1 (LCA1), supporting the use of AAV2/8 for gene therapy of LCA1 as well as of other retinal diseases requiring gene transfer to both PR types.


Asunto(s)
Terapia Genética , Amaurosis Congénita de Leber/terapia , Degeneración Retiniana/terapia , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Amaurosis Congénita de Leber/genética , Ratones , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Degeneración Retiniana/genética , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Transducción Genética
9.
Ophthalmology ; 120(6): 1283-91, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23474247

RESUMEN

OBJECTIVE: The aim of this study was to show the clinical data of long-term (3-year) follow-up of 5 patients affected by Leber congenital amaurosis type 2 (LCA2) treated with a single unilateral injection of adeno-associated virus AAV2-hRPE65v2. DESIGN: Clinical trial. PARTICIPANTS: Five LCA2 patients with RPE65 gene mutations. METHODS: After informed consent and confirmation of trial eligibility criteria, the eye with worse visual function was selected for subretinal delivery of adeno-associated virus (AAV2-hRPE65v2). Subjects were evaluated before and after surgery at designated follow-up visits (1, 2, 3, 14, 30, 60, 90, 180, 270, and 365 days, 1.5 years, and 3 years) by complete ophthalmic examination. Efficacy for each subject was monitored with best-corrected visual acuity, kinetic visual field, nystagmus testing, and pupillary light reflex. MAIN OUTCOME MEASURES: Best-corrected visual acuity, kinetic visual field, nystagmus testing, and pupillary light reflex. RESULTS: The data showed a statistically significant improvement of best-corrected visual acuity between baseline and 3 years after treatment in the treated eye (P<0.001). In all patients, an enlargement of the area of visual field was observed that remained stable until 3 years after injection (average values: baseline, 1058 deg(2) vs. 3 years after treatment, 4630 deg(2)) and a reduction of the nystagmus frequency compared with baseline at the 3-year time point. Furthermore, a statistically significant difference was observed in the pupillary constriction of the treated eye (P<0.05) compared with the untreated eye in 3 patients at 1- and 3-year time points. No patients experienced serious adverse events related to the vector in the 3-year postinjection period. CONCLUSIONS: The long-term follow-up data (3 years) on the 5-patient Italian cohort involved in the LCA2 gene therapy clinical trial clearly showed a stability of improvement in visual and retinal function that had been achieved a few months after treatment. Longitudinal data analysis showed that the maximum improvement was achieved within 6 months after treatment, and the visual improvement was stable up to the last observed time point. FINANCIAL DISCLOSURE(S): Proprietary or commercial disclosure may be found after the references.


Asunto(s)
Dependovirus/genética , Terapia Genética , Amaurosis Congénita de Leber/terapia , cis-trans-Isomerasas/genética , Adolescente , Adulto , Niño , Femenino , Estudios de Seguimiento , Vectores Genéticos , Humanos , Inyecciones Intraoculares , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/fisiopatología , Masculino , Mutación , Reflejo Pupilar/fisiología , Tomografía de Coherencia Óptica , Transfección , Agudeza Visual/fisiología , Campos Visuales/fisiología , Adulto Joven
10.
Hum Mol Genet ; 20(11): 2251-62, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21421996

RESUMEN

Given the high genetic heterogeneity of inherited retinal degenerations (IRDs), a wide applicable treatment would be desirable to halt/slow progressive photoreceptor (PR) cell loss in a mutation-independent manner. In addition to its erythropoietic activity, erythropoietin (EPO) presents neurotrophic characteristics. We have previously shown that adeno-associated viral (AAV) vector-mediated systemic EPO delivery protects from PR degeneration. However, this is associated with an undesired hematocrit increase that could contribute to PR protection. Non-erythropoietic EPO derivatives (EPO-D) are available which allow us to dissect erythropoiesis's role in PR preservation and may be more versatile and safe than EPO as anti-apoptotic agents. We delivered in animal models of light-induced or genetic retinal degeneration either intramuscularly or subretinally AAV vectors encoding EPO or one of the three selected EPO-D: the mutant S100E, the helix A- and B-derived EPO-mimetic peptides. We observed that (i) systemic expression of S100E induces a significantly lower hematocrit increase than EPO and provides similar protection from PR degeneration, and (ii) intraocular expression of EPO-D protects PR from degeneration in the absence of significant hematocrit increase. On the basis of this, we conclude that erythropoiesis is not required for EPO-mediated PR protection. However, the lower efficacy observed when EPO or S100E is expressed intraocularly rather than systemically suggests that hormone systemic effects contribute to PR protection. Unlike S100E, EPO-mimetic peptides preserve PR only when given locally, suggesting that different EPO-D have a different potency or mode of action. In conclusion, our data show that subretinal delivery of AAV vectors encoding EPO-D protects from light-induced and genetic PR degeneration.


Asunto(s)
Eritropoyetina/farmacología , Luz/efectos adversos , Células Fotorreceptoras de Vertebrados/patología , Degeneración Retiniana/terapia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Dependovirus , Eritropoyesis , Eritropoyetina/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Hematócrito , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Modelos Animales , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Periferinas , Ratas , Ratas Endogámicas Lew , Degeneración Retiniana/genética
11.
Mol Ther ; 19(5): 860-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21326216

RESUMEN

Multiple sulfatase deficiency (MSD), a severe autosomal recessive disease is caused by mutations in the sulfatase modifying factor 1 gene (Sumf1). We have previously shown that in the Sumf1 knockout mouse model (Sumf1(-/-)) sulfatase activities are completely absent and, similarly to MSD patients, this mouse model displays growth retardation and early mortality. The severity of the phenotype makes MSD unsuitable to be treated by enzyme replacement or bone marrow transplantation, hence the importance of testing the efficacy of novel treatment strategies. Here we show that recombinant adeno-associated virus serotype 9 (rAAV9) vector injected into the cerebral ventricles of neonatal mice resulted in efficient and widespread transduction of the brain parenchyma. In addition, we compared a combined, intracerebral ventricles and systemic, administration of an rAAV9 vector encoding SUMF1 gene to the single administrations-either directly in brain, or systemic alone -in MSD mice. The combined treatment resulted in the global activation of sulfatases, near-complete clearance of glycosaminoglycans (GAGs) and decrease of inflammation in both the central nervous system (CNS) and visceral organs. Furthermore, behavioral abilities were improved by the combined treatment. These results underscore that the "combined" mode of rAAV9 vector administration is an efficient option for the treatment of severe whole-body disorders.


Asunto(s)
Terapia Genética , Enfermedad por Deficiencia de Múltiples Sulfatasas/genética , Enfermedad por Deficiencia de Múltiples Sulfatasas/terapia , Sulfatasas/metabolismo , Animales , Western Blotting , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Ventrículos Cerebrales/virología , Dependovirus/genética , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Técnicas de Transferencia de Gen , Genes Transgénicos Suicidas , Vectores Genéticos , Glicosaminoglicanos/metabolismo , Inflamación/terapia , Ratones , Ratones Endogámicos C57BL , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro , Sulfatasas/deficiencia
12.
Mol Ther ; 18(3): 643-50, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19953081

RESUMEN

The safety and efficacy of gene therapy for inherited retinal diseases is being tested in humans affected with Leber's congenital amaurosis (LCA), an autosomal recessive blinding disease. Three independent studies have provided evidence that the subretinal administration of adeno-associated viral (AAV) vectors encoding RPE65 in patients affected with LCA2 due to mutations in the RPE65 gene, is safe and, in some cases, results in efficacy. We evaluated the long-term safety and efficacy (global effects on retinal/visual function) resulting from subretinal administration of AAV2-hRPE65v2. Both the safety and the efficacy noted at early timepoints persist through at least 1.5 years after injection in the three LCA2 patients enrolled in the low dose cohort of our trial. A transient rise in neutralizing antibodies to AAV capsid was observed but there was no humoral response to RPE65 protein. The persistence of functional amelioration suggests that AAV-mediated gene transfer to the human retina does not elicit immunological responses which cause significant loss of transduced cells. The persistence of physiologic effect supports the possibility that gene therapy may influence LCA2 disease progression. The safety of the intervention and the stability of the improvement in visual and retinal function in these subjects support the use of AAV-mediated gene augmentation therapy for treatment of inherited retinal diseases.


Asunto(s)
Proteínas Portadoras/genética , Proteínas del Ojo/genética , Terapia Genética/métodos , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/terapia , Adulto , Dependovirus/genética , Progresión de la Enfermedad , Estudios de Seguimiento , Vectores Genéticos , Humanos , Modelos Genéticos , Retina/metabolismo , Factores de Tiempo , Transgenes , Resultado del Tratamiento , Visión Ocular , cis-trans-Isomerasas
13.
Lancet ; 374(9701): 1597-605, 2009 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-19854499

RESUMEN

BACKGROUND: Gene therapy has the potential to reverse disease or prevent further deterioration of vision in patients with incurable inherited retinal degeneration. We therefore did a phase 1 trial to assess the effect of gene therapy on retinal and visual function in children and adults with Leber's congenital amaurosis. METHODS: We assessed the retinal and visual function in 12 patients (aged 8-44 years) with RPE65-associated Leber's congenital amaurosis given one subretinal injection of adeno-associated virus (AAV) containing a gene encoding a protein needed for the isomerohydrolase activity of the retinal pigment epithelium (AAV2-hRPE65v2) in the worst eye at low (1.5 x 10(10) vector genomes), medium (4.8 x 10(10) vector genomes), or high dose (1.5 x 10(11) vector genomes) for up to 2 years. FINDINGS: AAV2-hRPE65v2 was well tolerated and all patients showed sustained improvement in subjective and objective measurements of vision (ie, dark adaptometry, pupillometry, electroretinography, nystagmus, and ambulatory behaviour). Patients had at least a 2 log unit increase in pupillary light responses, and an 8-year-old child had nearly the same level of light sensitivity as that in age-matched normal-sighted individuals. The greatest improvement was noted in children, all of whom gained ambulatory vision. The study is registered with ClinicalTrials.gov, number NCT00516477. INTERPRETATION: The safety, extent, and stability of improvement in vision in all patients support the use of AAV-mediated gene therapy for treatment of inherited retinal diseases, with early intervention resulting in the best potential gain. FUNDING: Center for Cellular and Molecular Therapeutics at the Children's Hospital of Philadelphia, Foundation Fighting Blindness, Telethon, Research to Prevent Blindness, F M Kirby Foundation, Mackall Foundation Trust, Regione Campania Convenzione, European Union, Associazione Italiana Amaurosi Congenita di Leber, Fund for Scientific Research, Fund for Research in Ophthalmology, and National Center for Research Resources.


Asunto(s)
Proteínas Portadoras/genética , Proteínas del Ojo/genética , Terapia Genética/métodos , Atrofia Óptica Hereditaria de Leber/terapia , Adolescente , Adulto , Factores de Edad , Ceguera/congénito , Ceguera/genética , Niño , Adaptación a la Oscuridad , Dependovirus/genética , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Electrorretinografía , Femenino , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Humanos , Inyecciones , Masculino , Mutación/genética , Nistagmo Fisiológico , Atrofia Óptica Hereditaria de Leber/diagnóstico , Atrofia Óptica Hereditaria de Leber/genética , Seguridad , Resultado del Tratamiento , Agudeza Visual , Adulto Joven , cis-trans-Isomerasas
14.
Hum Mol Genet ; 18(23): 4530-45, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19717472

RESUMEN

OA1 (GPR143; GPCR, G-protein-coupled receptor), the protein product of the ocular albinism type 1 gene, encodes a pigment-cell-specific GPCR that localizes intracellularly to melanosomes. OA1 mutations result in ocular albinism due to alterations in melanosome formation, suggesting that OA1 is a key player in the biogenesis of melanosomes. To address the function of OA1 in melanosome biogenesis, we have used siRNA inactivation and combined morphological and biochemical methods to investigate melanosome ultrastructure, melanosomal protein localization and expression in human pigmented melanocytic cells. OA1 loss of function leads to decreased pigmentation and causes formation of enlarged aberrant premelanosomes harboring disorganized fibrillar structures and displaying proteins of mature melanosomes and lysosomes at their membrane. Moreover, we show that OA1 interacts biochemically with the premelanosomal protein MART-1. Inactivation of MART-1 by siRNA leads to a decreased stability of OA1 and is accompanied by similar defects in premelanosome biogenesis and composition. These data show for the first time that melanosome composition and identity are regulated at early stages by OA1 and that MART-1 likely acts as an escort protein for this GPCR.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteínas del Ojo/metabolismo , Melanocitos/metabolismo , Melanosomas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Retina/crecimiento & desarrollo , Animales , Antígenos de Neoplasias/genética , Línea Celular Tumoral , Células Cultivadas , Proteínas del Ojo/genética , Humanos , Antígeno MART-1 , Melanosomas/genética , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Unión Proteica , Transporte de Proteínas , Receptores Acoplados a Proteínas G/genética , Retina/metabolismo
15.
Mol Ther ; 17(8): 1347-54, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19436266

RESUMEN

Oculo-cutaneous albinism type 1 (OCA1) is characterized by congenital hypopigmentation and is due to mutations in the TYROSINASE gene (TYR). In this study, we have characterized the morpho-functional consequences of the lack of tyrosinase activity in the spontaneous null mouse model of OCA1 (Tyr(c-2j)). Here, we show that adult Tyr(c-2j) mice have several retinal functional anomalies associated with photoreceptor loss. To test whether these anomalies are reversible upon TYR complementation, we performed intraocular administration of an adeno-associated virus (AAV)-based vector, encoding the human TYR gene, in adult Tyr(c-2j) mice. This resulted in melanosome biogenesis and ex novo synthesis of melanin in both neuroectodermally derived retinal pigment epithelium (RPE) and in neural crest-derived choroid and iris melanocytes. Ocular melanin accumulation prevented progressive photoreceptor degeneration and resulted in restoration of retinal function. Our results reveal novel properties of pigment cells and show that the developmental anomalies of albino mice are associated with defects occurring in postnatal life, adding novel insights on OCA1 disease pathogenesis. In addition, we provide proof-of-principle of an effective gene-based strategy relevant for future application in albino patients.


Asunto(s)
Albinismo Oculocutáneo/metabolismo , Albinismo Oculocutáneo/terapia , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Melaninas/metabolismo , Monofenol Monooxigenasa/fisiología , Retina/metabolismo , Albinismo Oculocutáneo/patología , Albinismo Oculocutáneo/ultraestructura , Animales , Electrofisiología , Humanos , Iris/metabolismo , Iris/patología , Iris/ultraestructura , Melanocitos/metabolismo , Melanocitos/patología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Monofenol Monooxigenasa/genética , Retina/patología , Retina/ultraestructura , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología
16.
Hum Mol Genet ; 17(22): 3487-501, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18697795

RESUMEN

The protein product of the ocular albinism type 1 gene, named OA1, is a pigment cell-specific G protein-coupled receptor exclusively localized to intracellular organelles, namely lysosomes and melanosomes. Loss of OA1 function leads to the formation of macromelanosomes, suggesting that this receptor is implicated in organelle biogenesis, however the mechanism involved in the pathogenesis of the disease remains obscure. We report here the identification of an unexpected abnormality in melanosome distribution both in retinal pigment epithelium (RPE) and skin melanocytes of Oa1-knock-out (KO) mice, consisting in a displacement of the organelles from the central cytoplasm towards the cell periphery. Despite their depletion from the microtubule (MT)-enriched perinuclear region, Oa1-KO melanosomes were able to aggregate at the centrosome upon disruption of the actin cytoskeleton or expression of a dominant-negative construct of myosin Va. Consistently, quantification of organelle transport in living cells revealed that Oa1-KO melanosomes displayed a severe reduction in MT-based motility; however, this defect was rescued to normal following inhibition of actin-dependent capture at the cell periphery. Together, these data point to a defective regulation of organelle transport in the absence of OA1 and imply that the cytoskeleton might represent a downstream effector of this receptor. Furthermore, our results enlighten a novel function for OA1 in pigment cells and suggest that ocular albinism type 1 might result from a different pathogenetic mechanism than previously thought, based on an organelle-autonomous signalling pathway implicated in the regulation of both membrane traffic and transport.


Asunto(s)
Proteínas del Ojo/metabolismo , Melanocitos/metabolismo , Melanosomas/metabolismo , Glicoproteínas de Membrana/metabolismo , Epitelio Pigmentado Ocular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Albinismo Ocular/genética , Albinismo Ocular/metabolismo , Animales , Citoesqueleto/fisiología , Proteínas del Ojo/genética , Humanos , Melanocitos/patología , Melanocitos/ultraestructura , Melanosomas/genética , Melanosomas/ultraestructura , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Microscopía Electrónica , Epitelio Pigmentado Ocular/citología , Receptores Acoplados a Proteínas G/genética
17.
N Engl J Med ; 358(21): 2240-8, 2008 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-18441370

RESUMEN

Leber's congenital amaurosis (LCA) is a group of inherited blinding diseases with onset during childhood. One form of the disease, LCA2, is caused by mutations in the retinal pigment epithelium-specific 65-kDa protein gene (RPE65). We investigated the safety of subretinal delivery of a recombinant adeno-associated virus (AAV) carrying RPE65 complementary DNA (cDNA) (ClinicalTrials.gov number, NCT00516477 [ClinicalTrials.gov]). Three patients with LCA2 had an acceptable local and systemic adverse-event profile after delivery of AAV2.hRPE65v2. Each patient had a modest improvement in measures of retinal function on subjective tests of visual acuity. In one patient, an asymptomatic macular hole developed, and although the occurrence was considered to be an adverse event, the patient had some return of retinal function. Although the follow-up was very short and normal vision was not achieved, this study provides the basis for further gene therapy studies in patients with LCA.


Asunto(s)
Ceguera/terapia , Proteínas Portadoras/genética , Proteínas del Ojo/genética , Terapia Genética , Vectores Genéticos , Degeneración Retiniana/terapia , Adulto , Ceguera/congénito , Ceguera/genética , Ceguera/patología , ADN Complementario , Dependovirus/genética , Técnicas de Transferencia de Gen , Humanos , Inyecciones , Mutación , Regiones Promotoras Genéticas , Reflejo Pupilar , Retina/patología , Degeneración Retiniana/congénito , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Agudeza Visual , cis-trans-Isomerasas
18.
Vision Res ; 48(3): 353-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17923143

RESUMEN

Gene therapy represents a promising therapeutic option for many inherited and acquired retinal diseases. Recombinant adeno-associated viral vectors (AAV) are the most efficient tools to transfer genes in vivo to the retina. The recent identification of dozens of novel AAV serotypes enormously expands on the versatility of AAV as vector system for in vivo somatic gene transfer. The results from the forthcoming trials with AAV in the retina of patients with Leber Congenital Amaurosis will be critical for the rapid development of AAV-based therapeutics for retinal diseases.


Asunto(s)
Dependovirus/genética , Enfermedades Hereditarias del Ojo/terapia , Técnicas de Transferencia de Gen , Vectores Genéticos , Enfermedades de la Retina/terapia , Animales , Terapia Genética/métodos , Humanos , Transducción Genética
19.
J Virol ; 81(20): 11372-80, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17699581

RESUMEN

Severe inherited retinal diseases, such as retinitis pigmentosa and Leber congenital amaurosis, are caused by mutations in genes preferentially expressed in photoreceptors. While adeno-associated virus (AAV)-mediated gene transfer can correct retinal pigment epithelium (RPE) defects in animal models, approaches for the correction of photoreceptor-specific diseases are less efficient. We evaluated the ability of novel AAV serotypes (AAV2/7, AAV2/8, AAV2/9, AAV2rh.43, AAV2rh.64R1, and AAV2hu.29R) in combination with constitutive or photoreceptor-specific promoters to improve photoreceptor transduction, a limiting step in photoreceptor rescue. Based on a qualitative analysis, all AAV serotypes tested efficiently transduce the RPE as well as rod and cone photoreceptors after subretinal administration in mice. Interestingly, AAV2/9 efficiently transduces Müller cells. To compare photoreceptor transduction from different AAVs and promoters in both a qualitative and quantitative manner, we designed a strategy based on the use of a bicistronic construct expressing both enhanced green fluorescent protein and luciferase. We found that AAV2/8 and AAV2/7 mediate six- to eightfold higher levels of in vivo photoreceptor transduction than AAV2/5, considered so far the most efficient AAV serotype for photoreceptor targeting. In addition, following subretinal administration of AAV, the rhodopsin promoter allows significantly higher levels of photoreceptor expression than the other ubiquitous or photoreceptor-specific promoters tested. Finally, we show that AAV2/7, AAV2/8, and AAV2/9 outperform AAV2/5 following ex vivo transduction of retinal progenitor cells differentiated into photoreceptors. We conclude that AAV2/7 or AAV2/8 and the rhodopsin promoter provide the highest levels of photoreceptor transduction both in and ex vivo and that this may overcome the limitation to therapeutic success observed so far in models of inherited severe photoreceptor diseases.


Asunto(s)
Dependovirus/genética , Células Fotorreceptoras/metabolismo , Transducción Genética/métodos , Animales , Vectores Genéticos , Ratones , Regiones Promotoras Genéticas , Retina/citología , Rodopsina/genética , Serotipificación , Transducción Genética/normas
20.
Hum Mutat ; 27(5): 420-6, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16550551

RESUMEN

An intronic point mutation was identified in the ocular albinism type 1 (OA1) gene (HUGO symbol, GPR143) in a family with the X-linked form of ocular albinism. Interestingly, the mutation creates a new acceptor splice site in intron 7 of the OA1 gene. In addition to low levels of normally spliced mRNA product of the OA1 gene, the patient samples contained also an aberrantly spliced mRNA with a 165 bp fragment of intron 7 (from position +750 to +914) inserted between exons 7 and 8. The abnormal transcript contained a premature stop codon and was unstable, as revealed by Northern blot analysis. We defined that mutation NC_000023.8:g.25288G>A generated a consensus binding motif for the splicing factor enhancer ASF/SF2, which most likely favored transcription of the aberrant mRNA. Furthermore, it activated a cryptic donor-splice site causing the inclusion between exons 7 and 8 of the 165 bp intronic fragment. Thus, the aberrant splicing is most likely explained by the generation of a de novo splicing enhancer motif. Finally, to rescue OA1 expression in the patient's melanocytes, we designed an antisense morpholino modified oligonucleotide complementary to the mutant sequence. The morpholino oligonucleotide (MO) was able to rescue OA1 expression and restore the OA1 protein level in the patient's melanocytes through skipping of the aberrant inclusion. The use of MO demonstrated that the lack of OA1 was caused by the generation of a new splice site. Furthermore, this technique will lead to new approaches to correct splice site mutations that cause human diseases.


Asunto(s)
Albinismo Ocular/genética , Proteínas del Ojo/genética , Glicoproteínas de Membrana/genética , Oligonucleótidos Antisentido/farmacología , Mutación Puntual , Sitios de Empalme de ARN/genética , Secuencia de Bases , Análisis Mutacional de ADN , Proteínas del Ojo/efectos de los fármacos , Humanos , Melanocitos/citología , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Glicoproteínas de Membrana/efectos de los fármacos , Datos de Secuencia Molecular , Morfolinas/química , Empalme del ARN , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...