Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Transl Vis Sci Technol ; 11(11): 8, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36355386

RESUMEN

Purpose: Ocular hypertension is a significant risk factor for vision loss in glaucoma caused by the death of retinal ganglion cells (RGCs). We investigated whether small heat shock proteins (sHsps) expressed in RGCs protect those cells against ocular hypertension in mice. Methods: AAV2 vectors encoding genes for one of the following four human sHsps: HSPB1, HSPB4, HSPB5, or HSPB6 were constructed for RGC-specific expression. Ischemia/reperfusion was induced by elevating the intraocular pressure (IOP) to 120 mm Hg for one hour, followed by a rapid return to normal IOP. Microbeads (MB) were injected into the anterior chamber of mice to induce ocular hypertension. RGC death and glial activation were assessed by immunostaining for Brn3a, RBPMS, Iba1, and glial fibrillary acid protein in retinal flat mounts. RGC axonal defects were evaluated by anterograde transport of intravitreally injected cholera toxin-B. RGC function was assessed by pattern electroretinography. Results: Among the sHsps, HspB1 offered the best protection against RGC death from ischemia/reperfusion injury in the mouse retina. Intravitreal administration of AAV2-HSPB1 either two weeks before or one week after instituting ocular hypertension resulted in significant prevention of RGC loss. The MB-injected mice showed RGC axonal transportation defects, but AAV2-HSPB1 administration significantly inhibited this defect. AAV2-HSPB1 prevented glial activation caused by ocular hypertension. More importantly, a single injection of AAV2-HSPB1 protected RGCs long-term in MB-injected eyes. Conclusions: The administration of AAV2-HSPB1 inhibited RGC death and axonal transport defects and reduced glial activation in a mouse model of ocular hypertension. Translational Relevance: Our results suggested that the intravitreal delivery of AAV2-HSPB1 could be developed as a gene therapy to prevent vision loss on a long-term basis in glaucoma patients.


Asunto(s)
Glaucoma , Hipertensión Ocular , Humanos , Ratones , Animales , Células Ganglionares de la Retina/metabolismo , Transporte Axonal , Hipertensión Ocular/genética , Hipertensión Ocular/metabolismo , Glaucoma/genética , Glaucoma/prevención & control , Presión Intraocular , Modelos Animales de Enfermedad , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo
2.
PLoS One ; 17(5): e0269261, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35639753

RESUMEN

Glaucoma is the leading cause of irreversible blindness worldwide. Therapies for glaucoma are directed toward reducing intraocular pressure (IOP), the leading risk factor and only reliable therapeutic target via topical medications or with procedural intervention including laser or surgery. Though topical therapeutics are typically first line, less than 50% of patients take drops as prescribed. Sustained release technologies that decrease IOP for extended periods of time are being examined for clinical use. We recently identified Stanniocalcin-1, a naturally occurring hormone, as an IOP-lowering agent. Here, we show that a single injection into the anterior chamber of mice with an adeno-associated viral vector containing the transgene of stanniocalcin-1 results in diffuse and sustained expression of the protein and produces IOP reduction for up to 6 months. As the treatment effect begins to wane, IOP-lowering can be rescued with a repeat injection. Aqueous humor dynamic studies revealed an increase in outflow facility as the mechanism of action. This first-in-class therapeutic approach has the potential to improve care and reduce the rates of vision loss in the 80 million people worldwide currently affected by glaucoma.


Asunto(s)
Glaucoma , Hipotensión Ocular , Animales , Glaucoma/tratamiento farmacológico , Glaucoma/genética , Glicoproteínas , Humanos , Presión Intraocular , Ratones , Tonometría Ocular , Transgenes
3.
Dev Biol ; 464(2): 111-123, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32562755

RESUMEN

The transcription factors Prdm1 (Blimp1) and Vsx2 (Chx10) work downstream of Otx2 to regulate photoreceptor and bipolar cell fates in the developing retina. Mice that lack Vsx2 fail to form bipolar cells while Prdm1 mutants form excess bipolars at the direct expense of photoreceptors. Excess bipolars in Prdm1 mutants appear to derive from rods, suggesting that photoreceptor fate remains mutable for some time after cells become specified. Here we tested whether bipolar cell fate is also plastic during development. To do this, we created a system to conditionally misexpress Prdm1 at different stages of bipolar cell development. We found that Prdm1 blocks bipolar cell formation if expressed before the fate choice decision occurred. When we misexpressed Prdm1 just after the decision to become a bipolar cell was made, some cells were reprogrammed into photoreceptors. In contrast, Prdm1 misexpression in mature bipolar cells did not affect cell fate. We also provide evidence that sustained misexpression of Prdm1 was selectively toxic to photoreceptors. Our data show that bipolar fate is malleable, but only for a short temporal window following fate specification. Prdm1 and Vsx2 act by stabilizing photoreceptor and bipolar fates in developing OTX2+ cells of the retina.


Asunto(s)
Reprogramación Celular , Regulación del Desarrollo de la Expresión Génica , Células Fotorreceptoras de Vertebrados/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva/biosíntesis , Animales , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Mutación , Factores de Transcripción Otx/genética , Factores de Transcripción Otx/metabolismo , Células Fotorreceptoras de Vertebrados/citología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Hum Gene Ther ; 30(11): 1361-1370, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31418294

RESUMEN

Autosomal recessive Stargardt disease is the most common inherited macular degeneration in humans. It is caused by mutations in the retina-specific ATP binding cassette transporter A4 (ABCA4) that is essential for the clearance of all-trans-retinal from photoreceptor cells. Loss of this function results in the accumulation of toxic bisretinoids in the outer segment disk membranes and their subsequent transfer into adjacent retinal pigment epithelium (RPE) cells. This ultimately leads to the Stargardt disease phenotype of increased retinal autofluorescence and progressive RPE and photoreceptor cell loss. Adeno-associated virus (AAV) vectors have been widely used in gene therapeutic applications, but their limited cDNA packaging capacity of ∼4.5 kb has impeded their use for transgenes exceeding this limit. AAV dual vectors were developed to overcome this size restriction. In this study, we have evaluated the in vitro expression of ABCA4 using three options: overlap, transplicing, and hybrid ABCA4 dual vector systems. The hybrid system was the most efficient of these dual vector alternatives and used to express the full-length ABCA4 in Abca4-/- mice. The full-length ABCA4 protein correctly localized to photoreceptor outer segments. Moreover, treatment of Abca4-/- mice with this ABCA4 hybrid dual vector system resulted in a reduced accumulation of the lipofuscin/N-retinylidene-N-retinylethanolamine (A2E) autofluorescence in vivo, and retinal A2E quantification supported these findings. These results show that the hybrid AAV dual vector option is both safe and therapeutic in mice, and the delivered ABCA4 transgene is functional and has a significant effect on reducing A2E accumulation in the Abca4-/- mouse model of Stargardt disease.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/uso terapéutico , Dependovirus/genética , Genes Recesivos , Vectores Genéticos/metabolismo , Retina/patología , Enfermedad de Stargardt/genética , Enfermedad de Stargardt/terapia , Animales , Modelos Animales de Enfermedad , Fluorescencia , Fondo de Ojo , Células HEK293 , Humanos , Ratones Endogámicos C57BL , Retina/metabolismo , Retinoides/metabolismo
5.
Mol Ther ; 26(10): 2379-2396, 2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30217731

RESUMEN

Dysregulation of the complement system is implicated in neurodegeneration, including human and animal glaucoma. Optic nerve and retinal damage in glaucoma is preceded by local complement upregulation and activation, but whether targeting this early innate immune response could have therapeutic benefit remains undefined. Because complement signals through three pathways that intersect at complement C3 activation, here we targeted this step to restore complement balance in the glaucomatous retina and to determine its contribution to degeneration onset and/or progression. To achieve this, we combined adeno-associated virus retinal gene therapy with the targeted C3 inhibitor CR2-Crry. We show that intravitreal injection of AAV2.CR2-Crry produced sustained Crry overexpression in the retina and reduced deposition of the activation product complement C3d on retinal ganglion cells and the inner retina of DBA/2J mice. This resulted in neuroprotection of retinal ganglion cell axons and somata despite continued intraocular pressure elevation, suggesting a direct restriction of neurodegeneration onset and progression and significant delay to terminal disease stages. Our study uncovers a damaging effect of complement C3 or downstream complement activation in glaucoma, and it establishes AAV2.CR2-Crry as a viable therapeutic strategy to target pathogenic C3-mediated complement activation in the glaucomatous retina.


Asunto(s)
Complemento C3/genética , Glaucoma/terapia , Degeneración Nerviosa/terapia , Proteínas Recombinantes de Fusión/genética , Animales , Complemento C3/antagonistas & inhibidores , Dependovirus/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Terapia Genética , Glaucoma/genética , Glaucoma/patología , Humanos , Presión Intraocular/efectos de los fármacos , Inyecciones Intravítreas , Ratones , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Proteínas Recombinantes de Fusión/administración & dosificación , Retina/efectos de los fármacos , Retina/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología
6.
Front Mol Neurosci ; 11: 233, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30038560

RESUMEN

Rod and cone phosphodiesterase 6 (PDE6) are key effector enzymes of the vertebrate phototransduction pathway. Rod PDE6 consists of two catalytic subunits PDE6α and PDE6ß and two identical inhibitory PDE6γ subunits, while cone PDE6 is composed of two identical PDE6α' catalytic subunits and two identical cone-specific PDE6γ' inhibitory subunits. Despite their prominent function in regulating cGMP levels and therefore rod and cone light response properties, it is not known how each subunit contributes to the functional differences between rods and cones. In this study, we generated an rd10/cpfl1 mouse model lacking rod PDE6ß and cone PDE6α' subunits. Both rod and cone photoreceptor cells are degenerated with age and all PDE6 subunits degrade in rd10/cpfl1 mice. We expressed cone PDE6α' in both rods and cones of rd10/cpfl1 mice by adeno-associated virus (AAV)-mediated delivery driven by the ubiquitous, constitutive small chicken ß-actin promoter. We show that expression of PDE6α' rescues rod function in rd10/cpfl1 mice, and the restoration of rod light sensitivity is attained through restoration of endogenous rod PDE6γ and formation of a functional PDE6α'γ complex. However, improved photopic cone responses were achieved only after supplementation of both cone PDE6α' and PDE6γ' subunits but not by PDE6α' treatment alone. We observed a two fold increase of PDE6α' levels in the eyes injected with both PDE6α' plus PDE6γ' relative to eyes receiving PDE6α' alone. Despite the presence of both PDE6γ' and PDE6γ, the majority of PDE6α' formed functional complexes with PDE6γ', suggesting that PDE6α' has a higher association affinity for PDE6γ' than for PDE6γ. These results suggest that the presence of PDE6γ' augments cone PDE6 assembly and enhances its stability. Our finding has important implication for gene therapy of PDE6α'-associated achromatopsia.

7.
Proc Natl Acad Sci U S A ; 115(12): E2839-E2848, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29507198

RESUMEN

Mutations in the BEST1 gene cause detachment of the retina and degeneration of photoreceptor (PR) cells due to a primary channelopathy in the neighboring retinal pigment epithelium (RPE) cells. The pathophysiology of the interaction between RPE and PR cells preceding the formation of retinal detachment remains not well-understood. Our studies of molecular pathology in the canine BEST1 disease model revealed retina-wide abnormalities at the RPE-PR interface associated with defects in the RPE microvillar ensheathment and a cone PR-associated insoluble interphotoreceptor matrix. In vivo imaging demonstrated a retina-wide RPE-PR microdetachment, which contracted with dark adaptation and expanded upon exposure to a moderate intensity of light. Subretinal BEST1 gene augmentation therapy using adeno-associated virus 2 reversed not only clinically detectable subretinal lesions but also the diffuse microdetachments. Immunohistochemical analyses showed correction of the structural alterations at the RPE-PR interface in areas with BEST1 transgene expression. Successful treatment effects were demonstrated in three different canine BEST1 genotypes with vector titers in the 0.1-to-5E11 vector genomes per mL range. Patients with biallelic BEST1 mutations exhibited large regions of retinal lamination defects, severe PR sensitivity loss, and slowing of the retinoid cycle. Human translation of canine BEST1 gene therapy success in reversal of macro- and microdetachments through restoration of cytoarchitecture at the RPE-PR interface has promise to result in improved visual function and prevent disease progression in patients affected with bestrophinopathies.


Asunto(s)
Bestrofinas/genética , Enfermedades Hereditarias del Ojo/terapia , Terapia Genética/métodos , Enfermedades de la Retina/terapia , Animales , Enfermedades de los Perros/terapia , Perros , Enfermedades Hereditarias del Ojo/diagnóstico por imagen , Enfermedades Hereditarias del Ojo/patología , Enfermedades Hereditarias del Ojo/veterinaria , Femenino , Vectores Genéticos/farmacología , Humanos , Luz , Masculino , Mutación , Desprendimiento de Retina/diagnóstico por imagen , Desprendimiento de Retina/patología , Desprendimiento de Retina/terapia , Enfermedades de la Retina/diagnóstico por imagen , Enfermedades de la Retina/patología , Enfermedades de la Retina/veterinaria , Epitelio Pigmentado de la Retina/patología , Tomografía de Coherencia Óptica
8.
Mol Vis ; 24: 17-28, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29386880

RESUMEN

Purpose: Blue cone monochromacy (BCM) is an X-linked congenital vision disorder characterized by complete loss or severely reduced L- and M-cone function. Patients with BCM display poor visual acuity, severely impaired color discrimination, myopia, nystagmus, and minimally detectable cone-mediated electroretinogram. Recent studies of patients with BCM with adaptive optics scanning laser ophthalmoscopy (AOSLO) showed that they have a disrupted cone mosaic with reduced numbers of cones in the fovea that is normally dominated by L- and M-cones. The remaining cones in the fovea have significantly shortened outer segments but retain sufficient structural integrity to serve as potential gene therapy targets. In this study, we tested whether exogenously expressed human L- and M-opsins can rescue M-cone function in an M-opsin knockout (Opn1mw-/- ) mouse model for BCM. Methods: Adeno-associated virus type 5 (AAV5) vectors expressing OPN1LW, OPN1MW, or C-terminal tagged OPN1LW-Myc, or OPN1MW-HA driven by a cone-specific promoter were injected subretinally into one eye of Opn1mw-/- mice, while the contralateral eye served as the uninjected control. Expression of cone pigments was determined with western blotting and their cellular localization identified with immunohistochemistry. M-cone function was analyzed with electroretinogram (ERG). Antibodies against cone phototransduction proteins were used to study cone outer segment (OS) morphology in untreated and treated Opn1mw-/- eyes. Results: We showed that cones in the dorsal retina of the Opn1mw-/- mouse do not form outer segments, resembling cones that lack outer segments in the human BCM fovea. We further showed that AAV5-mediated expression of either human M- or L-opsin individually or combined promotes regrowth of cone outer segments and rescues M-cone function in the treated Opn1mw-/- dorsal retina. Conclusions: Exogenously expressed human opsins can regenerate cone outer segments and rescue M-cone function in Opn1mw-/- mice, thus providing a proof-of-concept gene therapy in an animal model of BCM.


Asunto(s)
Defectos de la Visión Cromática/terapia , Fóvea Central/metabolismo , Terapia Genética/métodos , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Opsinas de Bastones/genética , Animales , Defectos de la Visión Cromática/genética , Defectos de la Visión Cromática/metabolismo , Defectos de la Visión Cromática/patología , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Fóvea Central/patología , Expresión Génica , Prueba de Complementación Genética , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Ratones , Ratones Noqueados , Oftalmoscopía , Regiones Promotoras Genéticas , Segmento Externo de las Células Fotorreceptoras Retinianas/patología , Opsinas de Bastones/metabolismo , Transgenes
9.
Diabetes ; 67(4): 769-781, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29362226

RESUMEN

Tight junctions (TJs) involve close apposition of transmembrane proteins between cells. Although TJ proteins have been studied in detail, the role of lipids is largely unknown. We addressed the role of very long-chain (VLC ≥26) ceramides in TJs using diabetes-induced loss of the blood-retinal barrier as a model. VLC fatty acids that incorporate into VLC ceramides are produced by elongase elongation of very long-chain fatty acids protein 4 (ELOVL4). ELOVL4 is significantly reduced in the diabetic retina. Overexpression of ELOVL4 significantly decreased basal permeability, inhibited vascular endothelial growth factor (VEGF)- and interleukin-1ß-induced permeability, and prevented VEGF-induced decrease in occludin expression and border staining of TJ proteins ZO-1 and claudin-5. Intravitreal delivery of AAV2-hELOVL4 reduced diabetes-induced increase in vascular permeability. Ultrastructure and lipidomic analysis revealed that ω-linked acyl-VLC ceramides colocalize with TJ complexes. Overall, normalization of retinal ELOVL4 expression could prevent blood-retinal barrier dysregulation in diabetic retinopathy through an increase in VLC ceramides and stabilization of TJs.


Asunto(s)
Barrera Hematorretinal/metabolismo , Permeabilidad Capilar/genética , Ceramidas/metabolismo , Células Endoteliales/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Vasos Retinianos/metabolismo , Uniones Estrechas/metabolismo , Animales , Bovinos , Claudina-5/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/etiología , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Células Endoteliales/ultraestructura , Humanos , Interleucina-1beta/metabolismo , Ratones , Ocludina/metabolismo , Retina/metabolismo , Vasos Retinianos/ultraestructura , Uniones Estrechas/ultraestructura , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
10.
Mol Vis ; 24: 834-846, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30713422

RESUMEN

Purpose: Recessive mutations in the human IQCB1/NPHP5 gene are associated with Senior-Løken syndrome (SLS), a ciliopathy presenting with nephronophthisis and Leber congenital amaurosis (LCA). Nphp5-knockout mice develop LCA without nephronophthisis. Mutant rods rapidly degenerate while mutant cones survive for months. The purpose of this study was to reinitiate cone ciliogenesis in a Nphp5 -/-; Nrl -/- mouse with viral expression of full-length NPHP5 and rescue function. Methods: Nphp5 -/- mice were mated with Nrl -/- mice to generate Nphp5-/-; Nrl-/- double-knockouts. Nphp5-/-; Nrl-/- mice and Nphp5+/-; Nrl-/- controls were phenotyped with confocal microscopy from postnatal day 10 (P10) until 6 months of age. Nphp5-/-; Nrl-/- mice and Nphp5+/-; Nrl-/- controls were injected at P15 with self-complementary adenoassociated virus 8 (Y733F) (AAV8(Y733F)) expressing GRK1-FL-cNPHP5. Expression of mutant NPHP5 was verified with confocal microscopy and electroretinography (ERG). Results: In the Nphp5 -/- and cone-only Nphp5 -/-; Nrl -/- mice, cone outer segments did not form, but mutant cones continued to express cone pigments in the inner segments without obvious signs of cone cell death. The mutant cone outer nuclear layer (ONL) and the inner segments were stable for more than 6 months in the cone-only Nphp5 -/-; Nrl -/- retinas. Viral expression of NPHP5 initiated after eye opening showed that connecting cilia and RP1-positive axonemes were formed. Furthermore, cone pigments and other cone outer segment proteins (cone transducin and cone PDE6) were present in the nascent mutant cone outer segments, and rescued mutant cones exhibited a significant photopic b-wave (30% of Nphp5 +/-; Nrl -/- controls). Conclusions: Nphp5-/-; Nrl-/- cones persistently express cone pigments in the inner segments without obvious degeneration, providing an extended duration interval for viral gene expression. Viral expression of full-length NPHP5 initiates ciliogenesis between P15 and P60, and mutant cones are, in part, functional, encouraging future retina gene replacement therapy.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Proteínas de Unión a Calmodulina/genética , Proteínas del Ojo/genética , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/terapia , Células Fotorreceptoras Retinianas Conos/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Secuencia de Aminoácidos , Animales , Axonema/metabolismo , Axonema/ultraestructura , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Proteínas de Unión a Calmodulina/deficiencia , Cilios/metabolismo , Cilios/ultraestructura , Cruzamientos Genéticos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Modelos Animales de Enfermedad , Proteínas del Ojo/metabolismo , Femenino , Quinasa 1 del Receptor Acoplado a Proteína-G/genética , Quinasa 1 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Amaurosis Congénita de Leber/metabolismo , Amaurosis Congénita de Leber/patología , Masculino , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Fenotipo , Células Fotorreceptoras Retinianas Conos/patología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducina/genética , Transducina/metabolismo
11.
Hum Gene Ther Clin Dev ; 28(4): 197-207, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29020838

RESUMEN

Achromatopsia is an inherited retinal disorder of cone photoreceptors characterized by markedly reduced visual acuity, extreme light sensitivity, and absence of color discrimination. Approximately 50% of cases are caused by mutations in the cone photoreceptor-specific cyclic nucleotide gated channel beta subunit (CNGB3) gene. Studies in CNGB3-mutant dogs showed that subretinal injection of an AAV vector expressing human CNGB3, which has 76% amino acid identity with canine CNGB3, driven by a 2.1 kb human red cone opsin promoter (PR2.1) and packaged in AAV5 capsids (AAV5-PR2.1-hCNGB3) rescued cone photoreceptor function, but at high doses was associated with an inflammatory response (focal chorioretinitis) consistent with immune-mediated toxicity. AAV vectors containing the PR2.1 promoter packaged in AAV5 capsids and expressing either the native canine CNGB3 (AAV5-PR2.1-cCNGB3) or the human CNGB3 (AAV5-PR2.1-hCNGB3) were evaluated at different dose levels in CNGB3-mutant dogs. The vector expressing canine CNGB3 achieved somewhat better rescue of cone function but unexpectedly was associated with a greater degree of retinal toxicity than the vector expressing human CNGB3. Very low-level T-cell immune responses to some AAV or CNGB3 peptides were observed in animals that received the higher vector dose. There was a more than twofold increase in serum neutralizing antibodies to AAV in one of three animals in the low-dose group and in two of three animals in the high-dose group. No serum anti-hCNGB3 antibodies were detected in any animal. The results of this study do not support the hypothesis that the focal chorioretinitis seen with high doses of AAV5-PR2.1-hCNGB3 in the initial studies was due to an immune response to human CNGB3.


Asunto(s)
Defectos de la Visión Cromática/genética , Defectos de la Visión Cromática/terapia , Canales Catiónicos Regulados por Nucleótidos Cíclicos/uso terapéutico , Terapia Genética , Animales , Coriorretinitis/genética , Coriorretinitis/patología , Coriorretinitis/terapia , Defectos de la Visión Cromática/patología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Dependovirus , Enfermedades de los Perros/genética , Enfermedades de los Perros/patología , Enfermedades de los Perros/terapia , Perros , Vectores Genéticos/uso terapéutico , Humanos , Inmunidad Celular/genética , Opsinas/genética , Parvovirinae/genética , Regiones Promotoras Genéticas/genética , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Fotorreceptoras Retinianas Conos/patología
12.
Am J Pathol ; 186(9): 2486-99, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27423695

RESUMEN

Occludin is a transmembrane tight junction protein that contributes to diverse cellular functions, including control of barrier properties, cell migration, and proliferation. Vascular endothelial growth factor (VEGF) induces phosphorylation of occludin at S490, which is required for VEGF-induced endothelial permeability. Herein, we demonstrate that occludin S490 phosphorylation also regulates VEGF-induced retinal endothelial cell proliferation and neovascularization. Using a specific antibody, phospho-occludin was located in centrosomes in endothelial cell cultures, animal models, and human surgical samples of retinal neovessels. Occludin S490 phosphorylation was found to increase with endothelial tube formation in vitro and in vivo during retinal neovascularization after induction of VEGF expression. More important, expression of occludin mutated at S490 to Ala, completely inhibited angiogenesis in cell culture models and in vivo. Collectively, these data suggest a novel role for occludin in regulation of endothelial proliferation and angiogenesis in a phosphorylation-dependent manner. These findings may lead to methods of regulating pathological neovascularization by specifically targeting endothelial cell proliferation.


Asunto(s)
Ocludina/metabolismo , Neovascularización Retiniana/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Barrera Hematorretinal/metabolismo , Western Blotting , Bovinos , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Fosforilación
13.
PLoS One ; 11(2): e0148874, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26881841

RESUMEN

Usher syndrome type III (USH3A) is an autosomal recessive disorder caused by mutations in clarin-1 (CLRN1) gene, leading to progressive retinal degeneration and sensorineural deafness. Efforts to develop therapies for preventing photoreceptor cell loss are hampered by the lack of a retinal phenotype in the existing USH3 mouse models and by conflicting reports regarding the endogenous retinal localization of clarin-1, a transmembrane protein of unknown function. In this study, we used an AAV-based approach to express CLRN1 in the mouse retina in order to determine the pattern of its subcellular localization in different cell types. We found that all major classes of retinal cells express AAV-delivered CLRN1 driven by the ubiquitous, constitutive small chicken ß-actin promoter, which has important implications for the design of future USH3 gene therapy studies. Within photoreceptor cells, AAV-expressed CLRN1 is mainly localized at the inner segment region and outer plexiform layer, similar to the endogenous expression of other usher proteins. Subretinal delivery using a full strength viral titer led to significant loss of retinal function as evidenced by ERG analysis, suggesting that there is a critical limit for CLRN1 expression in photoreceptor cells. Taken together, these results suggest that CLRN1 expression is potentially supported by a variety of retinal cells, and the right combination of AAV vector dose, promoter, and delivery method needs to be selected to develop safe therapies for USH3 disorder.


Asunto(s)
Terapia Genética , Proteínas de la Membrana/biosíntesis , Degeneración Retiniana/genética , Síndromes de Usher/genética , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Ratones , Retina/metabolismo , Retina/patología , Degeneración Retiniana/patología , Degeneración Retiniana/terapia , Síndromes de Usher/patología , Síndromes de Usher/terapia
14.
Proc Natl Acad Sci U S A ; 112(42): E5689-98, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26438859

RESUMEN

Considerable evidence supports mutations in mitochondrial genes as the cause of maternally inherited diseases affecting tissues that rely primarily on oxidative energy metabolism, usually the nervous system, the heart, and skeletal muscles. Mitochondrial diseases are diverse, and animal models currently are limited. Here we introduced a mutant human mitochondrial gene responsible for Leber hereditary optic neuropathy (LHON) into the mouse germ line using fluorescence imaging for tissue-specific enrichment in the target retinal ganglion cells. A mitochondria-targeted adeno-associated virus (MTS-AAV) containing the mutant human NADH ubiquinone oxidoreductase subunit 4 (ND4) gene followed by mitochondrial-encoded mCherry was microinjected into zygotes. Female founders with mCherry fluorescence on ophthalmoscopy were backcrossed with normal males for eight generations. Mutant human ND4 DNA was 20% of mouse ND4 and did not integrate into the host genome. Translated human ND4 protein assembled into host respiratory complexes, decreasing respiratory chain function and increasing oxidative stress. Swelling of the optic nerve head was followed by progressive demise of ganglion cells and their axons, the hallmarks of human LHON. Early visual loss that began at 3 mo and progressed to blindness 8 mo after birth was reversed by intraocular injection of MTS-AAV expressing wild-type human ND4. The technology of introducing human mitochondrial genes into the mouse germ line has never been described, to our knowledge, and has implications not only for creating animal models recapitulating the counterpart human disorder but more importantly for reversing the adverse effects of the mutant gene using gene therapy to deliver the wild-type allele.


Asunto(s)
ADN Mitocondrial/genética , Técnicas de Transferencia de Gen , Células Germinativas , Mutación , Cigoto , Animales , Axones , Encéfalo/patología , Transporte de Electrón , Humanos , Ratones , Ratones Transgénicos , NADH Deshidrogenasa/genética , Estrés Oxidativo , Degeneración Retiniana/genética
15.
Hum Gene Ther ; 26(9): 593-602, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26076799

RESUMEN

Our collaborative successful gene replacement therapy using AAV vectors expressing a variant of human RPGR-ORF15 in two canine models provided therapeutic proof of concept for translation into human treatment. The ORF15 sequence contained within this AAV vector, however, has ORF15 DNA sequence variations compared to the published sequence that are likely due to its unusual composition of repetitive purine nucleotides. This mutability is a concern for AAV vector production and safety when contemplating a human trial. In this study, we establish the safety profile of AAV-hIRBP-hRPGR and AAV-hGRK1-hRPGR vectors used in the initial canine proof-of-principle experiments by demonstrating hRPGR-ORF15 sequence stability during all phases of manipulation, from plasmid propagation to vector production to its stability in vivo after subretinal administration to animals. We also evaluate potential toxicity in vivo by investigating protein expression, retinal structure and function, and vector biodistribution. Expression of hRPGR is detected in the inner segments and synaptic terminals of photoreceptors and is restricted to the connecting cilium when the vector is further diluted. Treated eyes exhibit no toxicity as assessed by retinal histopathology, immunocytochemistry, optical coherence tomography, fundoscopy, electroretinogram, and vector biodistribution. Therefore, the hRPGR-ORF15 variant in our AAV vectors appears to be a more stable form than the endogenous hRPGR cDNA when propagated in vitro. Its safety profile presented here in combination with its proven efficacy supports future gene therapy clinical trials.


Asunto(s)
Dependovirus/genética , Proteínas del Ojo/genética , Terapia Genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Vectores Genéticos , Humanos , Masculino , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Retina/patología
16.
Hum Mol Genet ; 24(13): 3699-707, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25855802

RESUMEN

The CNGA3(-/-)/Nrl(-/-) mouse is a cone-dominant model with Cnga3 channel deficiency, which partially mimics the all cone foveal structure of human achromatopsia 2 with CNGA3 mutations. Although subretinal (SR) AAV vector administration can transfect retinal cells efficiently, the injection-induced retinal detachment can cause retinal damage, particularly when SR vector bleb includes the fovea. We therefore explored whether cone function-structure could be rescued in CNGA3(-/-)/Nrl(-/-) mice by intravitreal (IVit) delivery of tyrosine to phenylalanine (Y-F) capsid mutant AAV8. We find that AAV-mediated CNGA3 expression can restore cone function and rescue structure following IVit delivery of AAV8 (Y447, 733F) vector. Rescue was assessed by restoration of the cone-mediated electroretinogram (ERG), optomotor responses, and cone opsin immunohistochemistry. Demonstration of gene therapy in a cone-dominant mouse model by IVit delivery provides a potential alternative vector delivery mode for safely transducing foveal cones in achromatopsia patients and in other human retinal diseases affecting foveal function.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Defectos de la Visión Cromática/genética , Defectos de la Visión Cromática/terapia , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Proteínas del Ojo/genética , Terapia Genética , Células Fotorreceptoras Retinianas Conos/fisiología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Defectos de la Visión Cromática/metabolismo , Defectos de la Visión Cromática/fisiopatología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Proteínas del Ojo/metabolismo , Femenino , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Hum Mol Genet ; 24(3): 670-84, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25274777

RESUMEN

Defects in aryl hydrocarbon receptor interacting protein-like1 (AIPL1) are associated with blinding diseases with a wide range of severity in humans. We examined the mechanism behind autosomal dominant cone-rod dystrophy (adCORD) caused by 12 base pair (bp) deletion at proline 351 of hAIPL1 (P351Δ12) mutation in the primate-specific region of human AIPL1. Mutant P351Δ12 human isoform, aryl hydrocarbon receptor interacting protein-like 1 (hAIPL1) mice demonstrated a CORD phenotype with early defects in cone-mediated vision and subsequent photoreceptor degeneration. A dominant CORD phenotype was observed in double transgenic animals expressing both mutant P351Δ12 and normal hAIPL1, but not with co-expression of P351Δ12 hAIPL1 and the mouse isoform, aryl hydrocarbon receptor interacting protein-like 1 (mAipl1). Despite a dominant effect of the mutation, we successfully rescued cone-mediated vision in P351Δ12 hAIPL1 mice following high over-expression of WT hAIPL1 by adeno-associated virus-mediated gene delivery, which was stable up to 6 months after treatment. Our transgenic P351Δ12 hAIPL1 mouse offers a novel model of AIPL1-CORD, with distinct defects from both the Aipl1-null mouse mimicking LCA and the Aipl1-hypomorphic mice mimicking a slow progressing RP.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Retinitis Pigmentosa/terapia , Animales , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Vectores Genéticos/administración & dosificación , Humanos , Ratones , Ratones Transgénicos , Células Fotorreceptoras Retinianas Conos/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Eliminación de Secuencia
18.
Front Cell Neurosci ; 8: 259, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25232303

RESUMEN

Piccolo is the largest known cytomatrix protein at active zones of chemical synapses. A growing number of studies on conventional chemical synapses assign Piccolo a role in the recruitment and integration of molecules relevant for both endo- and exocytosis of synaptic vesicles, the dynamic assembly of presynaptic F-actin, as well as the proteostasis of presynaptic proteins, yet a direct function in the structural organization of the active zone has not been uncovered in part due to the expression of multiple alternatively spliced isoforms. We recently identified Piccolino, a Piccolo splice variant specifically expressed in sensory ribbon synapses of the eye and ear. Here we down regulated Piccolino in vivo via an adeno-associated virus-based RNA interference approach and explored the impact on the presynaptic structure of mouse photoreceptor ribbon synapses. Detailed immunocytochemical light and electron microscopical analysis of Piccolino knockdown in photoreceptors revealed a hitherto undescribed photoreceptor ribbon synaptic phenotype with striking morphological changes of synaptic ribbon ultrastructure.

19.
PLoS One ; 9(4): e94175, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24714389

RESUMEN

The transcription factor p53 mediates the apoptosis of post-mitotic neurons exposed to a wide range of stress stimuli. The apoptotic activity of p53 is tightly regulated by the apoptosis-stimulating proteins of p53 (ASPP) family members: ASPP1, ASPP2 and iASPP. We previously showed that the pro-apoptotic members ASPP1 and ASPP2 contribute to p53-dependent death of retinal ganglion cells (RGCs). However, the role of the p53 inhibitor iASPP in the central nervous system (CNS) remains to be elucidated. To address this, we asked whether iASPP contributes to the survival of RGCs in an in vivo model of acute optic nerve damage. We demonstrate that iASPP is expressed by injured RGCs and that iASPP phosphorylation at serine residues, which increase iASPP affinity towards p53, is significantly reduced following axotomy. We show that short interference RNA (siRNA)-induced iASPP knockdown exacerbates RGC death, whereas adeno-associated virus (AAV)-mediated iASPP expression promotes RGC survival. Importantly, our data also demonstrate that increasing iASPP expression in RGCs downregulates p53 activity and blocks the expression of pro-apoptotic targets PUMA and Fas/CD95. This study demonstrates a novel role for iASPP in the survival of RGCs, and provides further evidence of the importance of the ASPP family in the regulation of neuronal loss after axonal injury.


Asunto(s)
Axones/metabolismo , Supervivencia Celular/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Traumatismos del Nervio Óptico/metabolismo , Nervio Óptico/metabolismo , Proteínas Represoras/metabolismo , Animales , Axotomía , Femenino , Péptidos y Proteínas de Señalización Intracelular/genética , Neuronas/metabolismo , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/genética
20.
Adv Exp Med Biol ; 801: 695-701, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24664760

RESUMEN

Achromatopsia (ACHM) is caused by a progressive loss of cone photoreceptors leading to color blindness and poor visual acuity. Animal studies and human clinical trials have shown that gene replacement therapy with adeno-associate virus (AAV) is a viable treatment option for this disease. Although there have been successful attempts to optimize capsid proteins for increased specificity, it is simpler to restrict expression via the use of cell type-specific promoters. To target cone photoreceptors, a chimeric promoter consisting of an enhancer element of interphotoreceptor retinoid-binding protein promoter and a minimal sequence of the human transducin alpha-subunit promoter (IRBPe/GNAT2) was created. Additionally, a synthetic transducin alpha-subunit promoter (synGNAT2/GNAT2) containing conserved sequence blocks located downstream of the transcriptional start was created. The strength and specificity of these promoters were evaluated in murine retina by immunohistochemistry. The results showed that the chimeric, (IRBPe/GNAT2) promoter is more efficient and specific than the synthetic, synGNAT2/GNAT2 promoter. Additionally, IRBPe/GNAT2-mediated expression was found in all cone subtypes and it was improved over existing promoters currently used for gene therapy of achromatopsia.


Asunto(s)
Defectos de la Visión Cromática/genética , Defectos de la Visión Cromática/terapia , Terapia Genética/métodos , Regiones Promotoras Genéticas/genética , Células Fotorreceptoras Retinianas Conos/fisiología , Transducina/genética , Animales , Defectos de la Visión Cromática/patología , Dependovirus/genética , Perros , Proteínas del Ojo/genética , Regulación de la Expresión Génica , Humanos , Ratones , Ratas , Proteínas Recombinantes de Fusión/genética , Células Fotorreceptoras Retinianas Conos/patología , Proteínas de Unión al Retinol/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...