Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 132(9): 924-934, 2018 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-30002144

RESUMEN

To date, little is known about the interaction between (pre-)malignant B cells and T cells. We generated transgenic mice that allow B cell-specific induction of the oncogene SV40 large T-antigen (TAg) to analyze the role of oncogene-specific T cells during sporadic B-cell lymphoma development. Constitutive TAg expression in CD19-Cre × LoxP-Tag mice resulted in TAg-tolerant CD8+ T cells and development of B-cell lymphomas. In contrast, CD19-CreERT2 × LoxP-Tag mice retained TAg-competent CD8+ T cells at time of oncogene induction and TAg expression in few B cells of adult mice resulted in exceptionally rare lymphoma formation late in life. Increased lymphoma incidence in the absence of TAg-specific T cells suggested T cell-mediated inhibition of lymphoma progression. However, TAg-initiated B cells were not eliminated by T cells and detected long term. Our results demonstrate a failure of the immune system to eradicate lymphoma-initiating B cells, retaining the risk of lymphoma development.


Asunto(s)
Antígenos Transformadores de Poliomavirus/inmunología , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular , Linfoma de Células B/inmunología , Animales , Antígenos Transformadores de Poliomavirus/genética , Linfocitos B/patología , Linfocitos T CD8-positivos/patología , Linfoma de Células B/genética , Linfoma de Células B/patología , Ratones , Ratones Noqueados
2.
Hum Mol Genet ; 25(6): 1165-75, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26740549

RESUMEN

Most inherited blinding diseases are characterized by compromised retinal function and progressive degeneration of photoreceptors. However, the factors that affect the life span of photoreceptors in such degenerative retinal diseases are rather poorly understood. Here, we explore the role of hyperpolarization-activated cyclic nucleotide-gated channel 1 (HCN1) in this context. HCN1 is known to adjust retinal function under mesopic conditions, and although it is expressed at high levels in rod and cone photoreceptor inner segments, no association with any retinal disorder has yet been found. We investigated the effects of an additional genetic deletion of HCN1 on the function and survival of photoreceptors in a mouse model of CNGB1-linked retinitis pigmentosa (RP). We found that the absence of HCN1 in Cngb1 knockout (KO) mice exacerbated photoreceptor degeneration. The deleterious effect was reduced by expression of HCN1 using a viral vector. Moreover, pharmacological inhibition of HCN1 also enhanced rod degeneration in Cngb1 KO mice. Patch-clamp recordings revealed that the membrane potentials of Cngb1 KO and Cngb1/Hcn1 double-KO rods were both significantly depolarized. We also found evidence for altered calcium homeostasis and increased activation of the protease calpain in Cngb1/Hcn1 double-KO mice. Finally, the deletion of HCN1 also exacerbated degeneration of cone photoreceptors in a mouse model of CNGA3-linked achromatopsia. Our results identify HCN1 as a major modifier of photoreceptor degeneration and suggest that pharmacological inhibition of HCN channels may enhance disease progression in RP and achromatopsia patients.


Asunto(s)
Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Canales de Potasio/genética , Canales de Potasio/metabolismo , Retinitis Pigmentosa/patología , Animales , Defectos de la Visión Cromática/genética , Defectos de la Visión Cromática/metabolismo , Defectos de la Visión Cromática/patología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Potenciales de la Membrana/fisiología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Retina/metabolismo , Retina/patología , Células Fotorreceptoras Retinianas Conos/metabolismo , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Visión Ocular
3.
Hum Mol Genet ; 24(19): 5486-99, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26188004

RESUMEN

Mutations in the PDE6A gene can cause rod photoreceptors degeneration and the blinding disease retinitis pigmentosa (RP). While a number of pathogenic PDE6A mutations have been described, little is known about their impact on compound heterozygous situations and potential interactions of different disease-causing alleles. Here, we used a novel mouse model for the Pde6a R562W mutation in combination with an existing line carrying the V685M mutation to generate compound heterozygous Pde6a V685M/R562W animals, exactly homologous to a case of human RP. We compared the progression of photoreceptor degeneration in these compound heterozygous mice with the homozygous V685M and R562W mutants, and additionally with the D670G line that is known for a relatively mild phenotype. We investigated PDE6A expression, cyclic guanosine mono-phosphate accumulation, calpain and caspase activity, in vivo retinal function and morphology, as well as photoreceptor cell death and survival. This analysis confirms the severity of different Pde6a mutations and indicates that compound heterozygous mutants behave like intermediates of the respective homozygous situations. Specifically, the severity of the four different Pde6a situations may be categorized by the pace of photoreceptor degeneration: V685M (fastest) > V685M/R562W > R562W > D670G (slowest). While calpain activity was strongly increased in all four mutants, caspase activity was not. This points to the execution of non-apoptotic cell death and may lead to the identification of new targets for therapeutic interventions. For individual RP patients, our study may help to predict time-courses for Pde6a-related retinal degeneration and thereby facilitate the definition of a window-of-opportunity for clinical interventions.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Mutación Puntual , Retina/fisiopatología , Retinitis Pigmentosa/patología , Animales , Calpaína/metabolismo , Caspasas/metabolismo , Supervivencia Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Retina/metabolismo , Retina/patología , Células Fotorreceptoras Retinianas Bastones/citología , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/fisiopatología
4.
J Gene Med ; 19(3)2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28095637

RESUMEN

The present review summarizes the current status of achromatopsia (ACHM) gene therapy-related research activities and provides an outlook for their clinical application. ACHM is an inherited eye disease characterized by a congenital absence of cone photoreceptor function. As a consequence, ACHM is associated with strongly impaired daylight vision, photophobia, nystagmus and a lack of color discrimination. Currently, six genes have been linked to ACHM. Up to 80% of the patients carry mutations in the genes CNGA3 and CNGB3 encoding the two subunits of the cone cyclic nucleotide-gated channel. Various animal models of the disease have been established and their characterization has helped to increase our understanding of the pathophysiology associated with ACHM. With the advent of adeno-associated virus vectors as valuable gene delivery tools for retinal photoreceptors, a number of promising gene supplementation therapy programs have been initiated. In recent years, huge progress has been made towards bringing a curative treatment for ACHM into clinics. The first clinical trials are ongoing or will be launched soon and are expected to contribute important data on the safety and efficacy of ACHM gene supplementation therapy.


Asunto(s)
Defectos de la Visión Cromática/genética , Defectos de la Visión Cromática/terapia , Terapia Genética , Animales , Ensayos Clínicos como Asunto , Defectos de la Visión Cromática/diagnóstico , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Dependovirus/genética , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Predisposición Genética a la Enfermedad , Terapia Genética/métodos , Vectores Genéticos/genética , Humanos , Mutación , Transgenes , Resultado del Tratamiento
5.
Mol Ther ; 21(5): 924-33, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23439498

RESUMEN

Insertion of completely complementary microRNA (miR) target sites (miRTS) into a transgene has been shown to be a valuable approach to specifically repress transgene expression in non-targeted tissues. miR-122TS have been successfully used to silence transgene expression in the liver following systemic application of cardiotropic adeno-associated virus (AAV) 9 vectors. For miR-206-mediated skeletal muscle-specific silencing of miR-206TS-bearing AAV9 vectors, however, we found this approach failed due to the expression of another member (miR-1) of the same miR family in heart tissue, the intended target. We introduced single-nucleotide substitutions into the miR-206TS and searched for those which prevented miR-1-mediated cardiac repression. Several mutated miR-206TS (m206TS), in particular m206TS-3G, were resistant to miR-1, but remained fully sensitive to miR-206. All these variants had mismatches in the seed region of the miR/m206TS duplex in common. Furthermore, we found that some m206TS, containing mismatches within the seed region or within the 3' portion of the miR-206, even enhanced the miR-206- mediated transgene repression. In vivo expression of m206TS-3G- and miR-122TS-containing transgene of systemically applied AAV9 vectors was strongly repressed in both skeletal muscle and the liver but remained high in the heart. Thus, site-directed mutagenesis of miRTS provides a new strategy to differentiate transgene de-targeting of related miRs.


Asunto(s)
Dependovirus/fisiología , Regulación de la Expresión Génica , Silenciador del Gen , Vectores Genéticos/genética , MicroARNs/genética , Músculo Esquelético/metabolismo , Transgenes , Emparejamiento Base , Secuencia de Bases , Sitios de Unión , Línea Celular , Orden Génico , Técnicas de Transferencia de Gen , Genes Reporteros , Vectores Genéticos/metabolismo , Humanos , MicroARNs/metabolismo , Mutagénesis Sitio-Dirigida , Miocardio/metabolismo , Especificidad de Órganos/genética , Tropismo Viral
6.
Mol Genet Genomics ; 288(10): 459-67, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23861024

RESUMEN

Visual phototransduction relies on the function of cyclic nucleotide-gated channels in the rod and cone photoreceptor outer segment plasma membranes. The role of these ion channels is to translate light-triggered changes in the second messenger cyclic guanosine 3'-5'-monophosphate levels into an electrical signal that is further processed within the retinal network and then sent to higher visual centers. Rod and cone photoreceptors express distinct CNG channels. The rod photoreceptor CNG channel is composed of one CNGB1 and three CNGA1 subunits, whereas the cone channel is formed by one CNGB3 and three CNGA3 subunits. Mutations in any of these channel subunits result in severe and currently untreatable retinal degenerative diseases like retinitis pigmentosa or achromatopsia. In this review, we provide an overview of the human diseases and relevant animal models of CNG channelopathies. Furthermore, we summarize recent results from preclinical gene therapy studies using adeno-associated viral vectors and discuss the efficacy and translational potential of these gene therapeutic approaches.


Asunto(s)
Canalopatías/genética , Defectos de la Visión Cromática/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Terapia Genética/métodos , Células Fotorreceptoras de Vertebrados/metabolismo , Retinitis Pigmentosa/genética , Animales , Canalopatías/terapia , Defectos de la Visión Cromática/terapia , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Dependovirus , Vectores Genéticos , Humanos , Modelos Animales , Modelos Moleculares , Mutación/genética , Filogenia , Retinitis Pigmentosa/terapia
7.
Bioorg Med Chem ; 21(17): 5139-44, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23859773

RESUMEN

There is a high demand for the development of an imaging agent for neurofibrillary tangles (NFTs) detection in Alzheimer's diagnosis. In the present study, a series of rhodanine-3-acetic acids was synthesized and evaluated for fluorescence imaging of NFTs in brain tissues of AD patients. Five out of seven probes have shown excellent binding affinity to NFTs over amyloid plaques in the Thiazine red R displacement assay. However, the selectivity in this in vitro assay is not confirmed by the histopathological evaluation, which indicates significant differences in the binding sites in the assays. Probe 6 showed binding affinity (IC50=19nM) to tau aggregates which is the highest among this series. Probes 2, 3, 4 and 5 display IC50 values of lower than 100nM to tau aggregates to displace Thiazine red R. Evaluation of the cytotoxicity of these five probes with human liver carcinoma cells revealed that these compounds excert negligible cytotoxicity. The in vivo studies with zebrafish embryos confirmed negligible cytotoxicity at 24 and 72h post fertilization.


Asunto(s)
Acetatos/química , Colorantes Fluorescentes/química , Rodanina/química , Acetatos/síntesis química , Acetatos/toxicidad , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/toxicidad , Humanos , Microscopía Fluorescente , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Pez Cebra/crecimiento & desarrollo , Proteínas tau/química , Proteínas tau/metabolismo
8.
Brain ; 140(6): 1542-1543, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28549130

Asunto(s)
Glaucoma , Retina , Apoptosis , Humanos
9.
Sci Rep ; 13(1): 6745, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37185990

RESUMEN

Enhancers are important cis-regulatory elements controlling cell-type specific expression patterns of genes. Furthermore, combinations of enhancers and minimal promoters are utilized to construct small, artificial promoters for gene delivery vectors. Large-scale functional screening methodology to construct genomic maps of enhancer activities has been successfully established in cultured cell lines, however, not yet applied to terminally differentiated cells and tissues in a living animal. Here, we transposed the Self-Transcribing Active Regulatory Region Sequencing (STARR-seq) technique to the mouse brain using adeno-associated-viruses (AAV) for the delivery of a highly complex screening library tiling entire genomic regions and covering in total 3 Mb of the mouse genome. We identified 483 sequences with enhancer activity, including sequences that were not predicted by DNA accessibility or histone marks. Characterizing the expression patterns of fluorescent reporters controlled by nine candidate sequences, we observed differential expression patterns also in sparse cell types. Together, our study provides an entry point for the unbiased study of enhancer activities in organisms during health and disease.


Asunto(s)
Elementos de Facilitación Genéticos , Genómica , Animales , Ratones , Genómica/métodos , Mapeo Cromosómico/métodos , Regiones Promotoras Genéticas , Encéfalo
10.
Life Sci Alliance ; 4(8)2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34183443

RESUMEN

Age-related macular degeneration (AMD) is the most common cause of blindness among the elderly and can be classified either as dry or as neovascular (or wet). Neovascular AMD is characterized by a strong immune response and the inadequate release of cytokines triggering angiogenesis and induction of photoreceptor death. The pathomechanisms of AMD are only partly understood. Here, we identify the endolysosomal two-pore cation channel TPC2 as a key factor of neovascularization and immune activation in the laser-induced choroidal neovascularization (CNV) mouse model of AMD. Block of TPC2 reduced retinal VEGFA and IL-1ß levels and diminished neovascularization and immune activation. Mechanistically, TPC2 mediates cationic currents in endolysosomal organelles of immune cells and lack of TPC2 leads to reduced IL-1ß levels in areas of choroidal neovascularization due to endolysosomal trapping. Taken together, our study identifies TPC2 as a promising novel therapeutic target for the treatment of AMD.


Asunto(s)
Canales de Calcio/genética , Interleucina-1beta/metabolismo , Rayos Láser/efectos adversos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Degeneración Macular Húmeda/genética , Animales , Línea Celular , Modelos Animales de Enfermedad , Angiografía con Fluoresceína , Humanos , Lisosomas/metabolismo , Ratones , Retina/metabolismo , Degeneración Macular Húmeda/etiología , Degeneración Macular Húmeda/metabolismo
11.
Stem Cell Reports ; 16(9): 2242-2256, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34525384

RESUMEN

Gene therapies using adeno-associated viruses (AAVs) are among the most promising strategies to treat or even cure hereditary and acquired retinal diseases. However, the development of new efficient AAV vectors is slow and costly, largely because of the lack of suitable non-clinical models. By faithfully recreating structure and function of human tissues, human induced pluripotent stem cell (iPSC)-derived retinal organoids could become an essential part of the test cascade addressing translational aspects. Organ-on-chip (OoC) technology further provides the capability to recapitulate microphysiological tissue environments as well as a precise control over structural and temporal parameters. By employing our recently developed retina on chip that merges organoid and OoC technology, we analyzed the efficacy, kinetics, and cell tropism of seven first- and second-generation AAV vectors. The presented data demonstrate the potential of iPSC-based OoC models as the next generation of screening platforms for future gene therapeutic studies.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/genética , Células Madre Pluripotentes Inducidas/citología , Dispositivos Laboratorio en un Chip , Organoides/metabolismo , Retina/metabolismo , Transducción Genética , Biomarcadores , Técnicas de Cultivo de Célula , Técnicas de Cultivo Tridimensional de Células , Diferenciación Celular , Técnica del Anticuerpo Fluorescente , Expresión Génica , Genes Reporteros , Terapia Genética , Humanos , Organoides/citología , Retina/citología , Transgenes
12.
EMBO Mol Med ; 13(4): e13392, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33616280

RESUMEN

Gene therapy using recombinant adeno-associated virus (rAAV) vectors to treat blinding retinal dystrophies has become clinical reality. Therapeutically impactful targeting of photoreceptors still relies on subretinal vector delivery, which detaches the retina and harbours substantial risks of collateral damage, often without achieving widespread photoreceptor transduction. Herein, we report the development of novel engineered rAAV vectors that enable efficient targeting of photoreceptors via less invasive intravitreal administration. A unique in vivo selection procedure was performed, where an AAV2-based peptide-display library was intravenously administered in mice, followed by isolation of vector DNA from target cells after only 24 h. This stringent selection yielded novel vectors, termed AAV2.GL and AAV2.NN, which mediate widespread and high-level retinal transduction after intravitreal injection in mice, dogs and non-human primates. Importantly, both vectors efficiently transduce photoreceptors in human retinal explant cultures. As proof-of-concept, intravitreal Cnga3 delivery using AAV2.GL lead to cone-specific expression of Cnga3 protein and rescued photopic cone responses in the Cnga3-/- mouse model of achromatopsia. These novel rAAV vectors expand the clinical applicability of gene therapy for blinding human retinal dystrophies.


Asunto(s)
Defectos de la Visión Cromática , Dependovirus , Animales , Cápside , Defectos de la Visión Cromática/terapia , Dependovirus/genética , Perros , Terapia Genética , Vectores Genéticos , Ratones , Retina
13.
Methods Mol Biol ; 1834: 383-390, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30324456

RESUMEN

Gene therapy holds promise for treating previously untreatable retinal disorders. The most promising approaches use gene transfer vectors derived from adeno-associated virus (AAV) to supplement a gene function in the affected cell type. One example is gene therapy for achromatopsia which affects daylight vision. In this case, recombinant AAV (rAAV) vectors are being developed to specifically target cone photoreceptors. Development of rAAV vectors could be facilitated by the use of in vitro models. In this chapter we provide a protocol which utilizes mouse 661W cells, an in vitro model of cone photoreceptors for evaluation of the transduction efficacy of rAAV vectors.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/genética , Retina/metabolismo , Animales , Línea Celular , Genes Reporteros , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Humanos , Ratones , Retina/citología , Células Fotorreceptoras Retinianas Conos/metabolismo , Enfermedades de la Retina/genética , Enfermedades de la Retina/terapia , Transducción Genética
14.
Methods Mol Biol ; 1834: 405-412, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30324458

RESUMEN

Gene therapy for inherited eye diseases requires local viral vector delivery by intraocular injection. Since large animal models are lacking for most of these diseases, genetically modified mouse models are commonly used in preclinical proof-of-concept studies. However, because of the relatively small mouse eye, adverse effects of the subretinal delivery procedure itself may interfere with the therapeutic outcome. The method described here aims to provide the details relevant to perform a transscleral pars plana virus-mediated gene transfer to achieve an optimized therapeutic effect in the small mouse eye.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética , Inyecciones Intraoculares , Retina/metabolismo , Animales , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Humanos , Inyecciones Intraoculares/métodos , Ratones , Células Fotorreceptoras/metabolismo , Retina/citología
15.
Methods Mol Biol ; 1715: 33-46, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29188504

RESUMEN

Achromatopsia (ACHM) and retinitis pigmentosa (RP) are inherited disorders caused by mutations in cone and rod photoreceptor-specific genes, respectively. ACHM strongly impairs daylight vision, whereas RP initially affects night vision and daylight vision at later stages. Currently, gene supplementation therapies utilizing recombinant adeno-associated virus (rAAV) vectors are being developed for various forms of ACHM and RP. In this chapter, we describe the procedure of designing and developing specific and efficient rAAV vectors for cone- and rod-specific gene supplementation.


Asunto(s)
Defectos de la Visión Cromática/terapia , Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Retinitis Pigmentosa/terapia , Defectos de la Visión Cromática/genética , Células HEK293 , Humanos , Plásmidos , Retinitis Pigmentosa/genética
16.
J Clin Invest ; 128(1): 190-206, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29202463

RESUMEN

Retinitis pigmentosa (RP) is a major cause of blindness that affects 1.5 million people worldwide. Mutations in cyclic nucleotide-gated channel ß 1 (CNGB1) cause approximately 4% of autosomal recessive RP. Gene augmentation therapy shows promise for treating inherited retinal degenerations; however, relevant animal models and biomarkers of progression in patients with RP are needed to assess therapeutic outcomes. Here, we evaluated RP patients with CNGB1 mutations for potential biomarkers of progression and compared human phenotypes with those of mouse and dog models of the disease. Additionally, we used gene augmentation therapy in a CNGß1-deficient dog model to evaluate potential translation to patients. CNGB1-deficient RP patients and mouse and dog models had a similar phenotype characterized by early loss of rod function and slow rod photoreceptor loss with a secondary decline in cone function. Advanced imaging showed promise for evaluating RP progression in human patients, and gene augmentation using adeno-associated virus vectors robustly sustained the rescue of rod function and preserved retinal structure in the dog model. Together, our results reveal an early loss of rod function in CNGB1-deficient patients and a wide window for therapeutic intervention. Moreover, the identification of potential biomarkers of outcome measures, availability of relevant animal models, and robust functional rescue from gene augmentation therapy support future work to move CNGB1-RP therapies toward clinical trials.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/deficiencia , Mutación , Proteínas del Tejido Nervioso/deficiencia , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Animales , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Dependovirus , Modelos Animales de Enfermedad , Perros , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Retinitis Pigmentosa/patología , Retinitis Pigmentosa/terapia , Transducción Genética
17.
Methods Mol Biol ; 1523: 361-368, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27975264

RESUMEN

Aggregates of hyperphosphorylated tau can be observed in the human brain affected by various neurodegenerative disorders. The development of a noninvasive technique for the visualization of these protein accumulations is a promising task. In the following protocol, we describe a method to image fibrillar tau in the retina of a transgenic mouse model of frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17). This technique can be highly valuable for the preclinical in vivo testing of approaches that target tau aggregation.


Asunto(s)
Diagnóstico por Imagen/métodos , Retina/metabolismo , Retina/patología , Proteínas tau/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Mutación/genética , Tauopatías/genética , Tauopatías/metabolismo , Tauopatías/patología
18.
Hum Gene Ther ; 28(12): 1189-1201, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29212382

RESUMEN

Mutations in the phosphodiesterase 6A gene (PDE6A) result in retinitis pigmentosa (RP) type 43 (RP43) and are responsible for about 4% of autosomal recessive RP. There is currently no treatment for this blinding condition. The aim of this project was to use a large-animal model to test a gene supplementation viral vector designed to be translated for use in a clinical trial for the treatment of RP43. Seven Pde6a-/- puppies were given sub-retinal injections of an adeno-associated viral vector (AAV) serotype 2/8 delivering human PDE6A cDNA under control of a short rhodopsin promoter (AAV8-PDE6A). Three puppies received ∼1 × 1011 vg in one eye and four puppies ∼5 × 1011 vg/per eye, with both eyes being injected in two animals. In vivo outcome measures included vision testing and electroretinography (ERG), as well as fundus and spectral domain-optical coherence tomography imaging. Some puppies were euthanized and their eyes processed for immunohistochemistry. All puppies had improved rod-mediated vision in the treated eye. ERGs showed improved rod-mediated responses in the higher-dose group but in only one of the lower-dose group animals. Receptor+ thickness was preserved and photoreceptor morphology improved in the treated retinal regions in all puppies. Treatment resulted in PDE6A transgene expression, accompanied by much increased levels of Pde6b, in rod outer segments in the injected retinal regions. There were several indications of improved retinal health in the PDE6A-expressing regions, including lack of abnormal cyclic guanosine monophosphate accumulation, appropriate rod opsin localization to the outer segments with a large reduction in mislocalization to other regions of the rod cell, and reduced Müller cell activation. Additionally, cone photoreceptors showed morphological improvement in the treated region, with normal-appearing inner and outer segments. AAV8-PDE6A gene supplementation therapy restored rod vision in Pde6a-/- puppies and preserved retinal morphology. These positive outcomes are an important step toward a human clinical trial to treat PDE6A-RP.

19.
Sci Rep ; 7(1): 2321, 2017 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-28539581

RESUMEN

Mutations in the photoreceptor outer segment (OS) specific peripherin-2 lead to autosomal dominant retinitis pigmentosa (adRP). By contrast, mutations in the peripherin-2 homolog Rom-1 cause digenic RP in combination with certain heterozygous mutations in peripherin-2. The mechanisms underlying the differential role of peripherin-2 and Rom-1 in RP pathophysiology remained elusive so far. Here, focusing on two adRP-linked peripherin-2 mutants, P210L and C214S, we analyzed the binding characteristics, protein assembly, and rod OS targeting of wild type (perWT), mutant peripherin-2 (perMT), or Rom-1 complexes, which can be formed in patients heterozygous for peripherin-2 mutations. Both mutants are misfolded and lead to decreased binding to perWT and Rom-1. Furthermore, both mutants are preferentially forming non-covalent perMT-perMT, perWT-perMT, and Rom-1-perMT dimers. However, only perWT-perMT, but not perMT-perMT or Rom-1-perMT complexes could be targeted to murine rod OS. Our study provides first evidence that non-covalent perWT-perMT dimers can be targeted to rod OS. Finally, our study unravels unexpected opposing roles of perWT and Rom-1 in rod OS targeting of adRP-linked peripherin-2 mutants and suggests a new treatment strategy for the affected individuals.


Asunto(s)
Periferinas/genética , Retinitis Pigmentosa/genética , Segmento Externo de la Célula en Bastón/metabolismo , Tetraspaninas/genética , Animales , Células COS , Chlorocebus aethiops , Proteínas del Ojo , Humanos , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/metabolismo , Ratones , Mutación , Periferinas/metabolismo , Unión Proteica , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Segmento Externo de la Célula en Bastón/patología , Tetraspaninas/metabolismo
20.
Front Neurosci ; 11: 292, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28596720

RESUMEN

Achromatopsia type 2 (ACHM2) is a severe, inherited eye disease caused by mutations in the CNGA3 gene encoding the α subunit of the cone photoreceptor cyclic nucleotide-gated (CNG) channel. Patients suffer from strongly impaired daylight vision, photophobia, nystagmus, and lack of color discrimination. We have previously shown in the Cnga3 knockout (KO) mouse model of ACHM2 that gene supplementation therapy is effective in rescuing cone function and morphology and delaying cone degeneration. In our preclinical approach, we use recombinant adeno-associated virus (AAV) vector-mediated gene transfer to express the murine Cnga3 gene under control of the mouse blue opsin promoter. Here, we provide novel data on the efficiency and permanence of such gene supplementation therapy in Cnga3 KO mice. Specifically, we compare the influence of two different AAV vector capsids, AAV2/5 (Y719F) and AAV2/8 (Y733F), on restoration of cone function, and assess the effect of age at time of treatment on the long-term outcome. The evaluation included in vivo analysis of retinal function using electroretinography (ERG) and immunohistochemical analysis of vector-driven Cnga3 transgene expression. We found that both vector capsid serotypes led to a comparable rescue of cone function over the observation period between 4 weeks and 3 months post treatment. In addition, a clear therapeutic effect was present in mice treated at 2 weeks of age as well as in mice treated at 3 months of age at the first assessment at 4 weeks after treatment. Importantly, the effect extended in both cases over the entire observation period of 12 months post treatment. However, the average ERG amplitude levels differed between the two groups, suggesting a role of the absolute age, or possibly, the associated state of the degeneration, on the achievable outcome. In summary, we found that the therapeutic time window of opportunity for AAV-mediated Cnga3 gene supplementation therapy in the Cnga3 KO mouse model extends at least to an age of 3 months, but is presumably limited by the condition, number and topographical distribution of remaining cones at the time of treatment. No impact of the choice of capsid on the therapeutic success was detected.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA