Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
EMBO Mol Med ; 15(4): e17033, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-36647689

RESUMO

Gyrate atrophy of choroid and retina (GACR) is a chorioretinal degeneration caused by pathogenic variants in the gene encoding ornithine aminotransferase (OAT), an enzyme mainly expressed in liver. Affected patients have increased ornithine concentrations in blood and other body fluids and develop progressive constriction of vision fields leading to blindness. Current therapies are unsatisfactory and better treatments are highly needed. In two mouse models of OAT deficiency that recapitulates biochemical and retinal changes of GACR, we investigated the efficacy of an intravenously injected serotype 8 adeno-associated (AAV8) vector expressing OAT under the control of a hepatocyte-specific promoter. Following injections, OAT-deficient mice showed reductions of ornithine concentrations in blood and eye cups compared with control mice injected with a vector expressing green fluorescent protein. AAV-injected mice showed improved electroretinogram response and partial restoration of retinal structure up to one-year post-injection. In summary, hepatic OAT expression by AAV8 vector was effective at correction of hyperornithinemia and improved function and structure of the retina. In conclusion, this study provides proof-of-concept of efficacy of liver-directed AAV-mediated gene therapy of GACR.


Assuntos
Atrofia Girata , Degeneração Retiniana , Animais , Camundongos , Atrofia Girata/genética , Atrofia Girata/patologia , Ornitina-Oxo-Ácido Transaminase/genética , Ornitina-Oxo-Ácido Transaminase/metabolismo , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Ornitina/genética , Ornitina/metabolismo , Terapia Genética , Fígado/patologia
2.
EMBO Mol Med ; 14(11): e15941, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-36194668

RESUMO

Inherited retinal diseases (IRDs) are a group of diseases whose common landmark is progressive photoreceptor loss. The development of gene-specific therapies for IRDs is hampered by their wide genetic heterogeneity. Mitochondrial dysfunction is proving to constitute one of the key pathogenic events in IRDs; hence, approaches that enhance mitochondrial activities have a promising therapeutic potential for these conditions. We previously reported that miR-181a/b downregulation boosts mitochondrial turnover in models of primary retinal mitochondrial diseases. Here, we show that miR-181a/b silencing has a beneficial effect also in IRDs. In particular, the injection in the subretinal space of an adeno-associated viral vector (AAV) that harbors a miR-181a/b inhibitor (sponge) sequence (AAV2/8-GFP-Sponge-miR-181a/b) improves retinal morphology and visual function both in models of autosomal dominant (RHO-P347S) and of autosomal recessive (rd10) retinitis pigmentosa. Moreover, we demonstrate that miR-181a/b downregulation modulates the level of the mitochondrial fission-related protein Drp1 and rescues the mitochondrial fragmentation in RHO-P347S photoreceptors. Overall, these data support the potential use of miR-181a/b downregulation as an innovative mutation-independent therapeutic strategy for IRDs, which can be effective both to delay disease progression and to aid gene-specific therapeutic approaches.


Assuntos
MicroRNAs , Retinose Pigmentar , Humanos , Regulação para Baixo , Retina/patologia , Retinose Pigmentar/genética , Retinose Pigmentar/terapia , Retinose Pigmentar/metabolismo , Mutação , MicroRNAs/genética , MicroRNAs/metabolismo
3.
Nat Commun ; 13(1): 1963, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35414130

RESUMO

Challenges to the widespread application of gene therapy with adeno-associated viral (AAV) vectors include dominant conditions due to gain-of-function mutations which require allele-specific knockout, as well as long-term transgene expression from proliferating tissues, which is hampered by AAV DNA episomal status. To overcome these challenges, we used CRISPR/Cas9-mediated homology-independent targeted integration (HITI) in retina and liver as paradigmatic target tissues. We show that AAV-HITI targets photoreceptors of both mouse and pig retina, and this results in significant improvements to retinal morphology and function in mice with autosomal dominant retinitis pigmentosa. In addition, we show that neonatal systemic AAV-HITI delivery achieves stable liver transgene expression and phenotypic improvement in a mouse model of a severe lysosomal storage disease. We also show that HITI applications predominantly result in on-target editing. These results lay the groundwork for the application of AAV-HITI for the treatment of diseases affecting various organs.


Assuntos
Dependovirus , Edição de Genes , Animais , Sistemas CRISPR-Cas , Dependovirus/genética , Edição de Genes/métodos , Vetores Genéticos/genética , Fígado , Camundongos , Retina/metabolismo , Suínos
4.
Int J Mol Sci ; 22(11)2021 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-34071252

RESUMO

Gene-expression programs modulated by transcription factors (TFs) mediate key developmental events. Here, we show that the synthetic transcriptional repressor (TR; ZF6-DB), designed to treat Rhodopsin-mediated autosomal dominant retinitis pigmentosa (RHO-adRP), does not perturb murine retinal development, while maintaining its ability to block Rho expression transcriptionally. To express ZF6-DB into the developing retina, we pursued two approaches, (i) the retinal delivery (somatic expression) of ZF6-DB by Adeno-associated virus (AAV) vector (AAV-ZF6-DB) gene transfer during retinogenesis and (ii) the generation of a transgenic mouse (germ-line transmission, TR-ZF6-DB). Somatic and transgenic expression of ZF6-DB during retinogenesis does not affect retinal function of wild-type mice. The P347S mouse model of RHO-adRP, subretinally injected with AAV-ZF6-DB, or crossed with TR-ZF6-DB or shows retinal morphological and functional recovery. We propose the use of developmental transitions as an effective mode to challenge the safety of retinal gene therapies operating at genome, transcriptional, and transcript levels.


Assuntos
Terapia Genética/métodos , Retina/metabolismo , Retinose Pigmentar/genética , Animais , Dependovirus/genética , Modelos Animais de Doenças , Feminino , Expressão Gênica , Genes Dominantes , Vetores Genéticos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Retinose Pigmentar/metabolismo , Rodopsina/genética , Fatores de Transcrição , Transcriptoma , Dedos de Zinco
5.
Am J Hum Genet ; 108(2): 295-308, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33508235

RESUMO

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.


Assuntos
Edição de Genes , Retinose Pigmentar/genética , Retinose Pigmentar/terapia , Rodopsina/genética , Alelos , Animais , Sistemas CRISPR-Cas , Linhagem Celular , Dependovirus/genética , Modelos Animais de Doenças , Eletrorretinografia , Terapia Genética , Humanos , Mutação INDEL , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/metabolismo , Retina/fisiopatologia , Rodopsina/metabolismo
6.
Sci Rep ; 10(1): 9619, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32541823

RESUMO

The presence of α-synuclein aggregates in the retina of Parkinson's disease patients has been associated with vision impairment. In this study we sought to determine the effects of α-synuclein overexpression on the survival and function of dopaminergic amacrine cells (DACs) in the retina. Adult mice were intravitreally injected with an adeno-associated viral (AAV) vector to overexpress human wild-type α-synuclein in the inner retina. Before and after systemic injections of levodopa (L-DOPA), retinal responses and visual acuity-driven behavior were measured by electroretinography (ERG) and a water maze task, respectively. Amacrine cells and ganglion cells were counted at different time points after the injection. α-synuclein overexpression led to an early loss of DACs associated with a decrease of light-adapted ERG responses and visual acuity that could be rescued by systemic injections of L-DOPA. The data show that α-synuclein overexpression affects dopamine neurons in the retina. The approach provides a novel accessible method to model the underlying mechanisms implicated in the pathogenesis of synucleinopathies and for testing novel treatments.


Assuntos
Células Amácrinas/metabolismo , Neurônios Dopaminérgicos/metabolismo , Retina/metabolismo , Degeneração Retiniana/metabolismo , Transtornos da Visão/metabolismo , alfa-Sinucleína/metabolismo , Células Amácrinas/patologia , Animais , Neurônios Dopaminérgicos/patologia , Feminino , Imunofluorescência , Levodopa/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Retina/efeitos dos fármacos , Retina/patologia , Degeneração Retiniana/patologia , Transtornos da Visão/patologia , Acuidade Visual
7.
Brain ; 143(6): 1686-1696, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32413099

RESUMO

Mutations in nuclear-encoded mitochondrial genes are responsible for a broad spectrum of disorders among which Leigh syndrome is the most common in infancy. No effective therapies are available for this severe disease mainly because of the limited capabilities of the standard adeno-associated viral (AAV) vectors to transduce both peripheral organs and the CNS when injected systemically in adults. Here, we used the brain-penetrating AAV-PHP.B vector to reinstate gene expression in the Ndufs4 knockout mouse model of Leigh syndrome. Intravenous delivery of an AAV.PHP.B-Ndufs4 vector in 1-month-old knockout mice restored mitochondrial complex I activity in several organs including the CNS. This gene replacement strategy extended lifespan, rescued metabolic parameters, provided behavioural improvement, and corrected the pathological phenotype in the brain, retina, and heart of Ndufs4 knockout mice. These results provide a robust proof that gene therapy strategies targeting multiple organs can rescue fatal neurometabolic disorders with CNS involvement.


Assuntos
Complexo I de Transporte de Elétrons/genética , Terapia Genética/métodos , Doença de Leigh/genética , Animais , Encéfalo/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/metabolismo , Expressão Gênica/genética , Vetores Genéticos , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Neurônios/metabolismo , Estudo de Prova de Conceito , Transdução Genética/métodos
8.
Mol Ther Nucleic Acids ; 19: 144-156, 2020 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-31837604

RESUMO

Inherited retinal diseases (IRDs) represent a frequent cause of genetic blindness. Their high genetic heterogeneity hinders the application of gene-specific therapies to the vast majority of patients. We recently demonstrated that the microRNA miR-204 is essential for retinal function, although the underlying molecular mechanisms remain poorly understood. Here, we investigated the therapeutic potential of miR-204 in IRDs. We subretinally delivered an adeno-associated viral (AAV) vector carrying the miR-204 precursor to two genetically different IRD mouse models. The administration of AAV-miR-204 preserved retinal function in a mouse model for a dominant form of retinitis pigmentosa (RHO-P347S). This was associated with a reduction of apoptotic photoreceptor cells and with a better preservation of photoreceptor marker expression. Transcriptome analysis showed that miR-204 shifts expression profiles of transgenic retinas toward those of healthy retinas by the downregulation of microglia activation and photoreceptor cell death. Delivery of miR-204 exerted neuroprotective effects also in a mouse model of Leber congenital amaurosis, due to mutations of the Aipl1 gene. Our study highlights the mutation-independent therapeutic potential of AAV-miR204 in slowing down retinal degeneration in IRDs and unveils the previously unreported role of this miRNA in attenuating microglia activation and photoreceptor cell death.

9.
Sci Transl Med ; 11(492)2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31092694

RESUMO

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.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/genética , Inteínas , Retina/virologia , Trans-Splicing/genética , Animais , Vetores Genéticos/administração & dosagem , Vetores Genéticos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Organoides/ultraestrutura , Organoides/virologia , Fenótipo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/virologia , Suínos
10.
JCI Insight ; 2(24)2017 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-29263295

RESUMO

The genome-wide activity of transcription factors (TFs) on multiple regulatory elements precludes their use as gene-specific regulators. Here we show that ectopic expression of a TF in a cell-specific context can be used to silence the expression of a specific gene as a therapeutic approach to regulate gene expression in human disease. We selected the TF Krüppel-like factor 15 (KLF15) based on its putative ability to recognize a specific DNA sequence motif present in the rhodopsin (RHO) promoter and its lack of expression in terminally differentiated rod photoreceptors (the RHO-expressing cells). Adeno-associated virus (AAV) vector-mediated ectopic expression of KLF15 in rod photoreceptors of pigs enables Rho silencing with limited genome-wide transcriptional perturbations. Suppression of a RHO mutant allele by KLF15 corrects the phenotype of a mouse model of retinitis pigmentosa with no observed toxicity. Cell-specific-context conditioning of TF activity may prove a novel mode for somatic gene-targeted manipulation.


Assuntos
Inativação Gênica , Marcação de Genes/métodos , Fatores de Transcrição Kruppel-Like/genética , Proteínas Nucleares/genética , Rodopsina/genética , Animais , Dependovirus/genética , Expressão Ectópica do Gene , Feminino , Terapia Genética/métodos , Vetores Genéticos , Fatores de Transcrição Kruppel-Like/fisiologia , Camundongos Transgênicos , Mutação , Proteínas Nucleares/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Retinose Pigmentar/genética , Retinose Pigmentar/terapia , Rodopsina/metabolismo , Suínos
11.
Elife ; 5: e12242, 2016 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-26974343

RESUMO

Transcription factors (TFs) operate by the combined activity of their DNA-binding domains (DBDs) and effector domains (EDs) enabling the coordination of gene expression on a genomic scale. Here we show that in vivo delivery of an engineered DNA-binding protein uncoupled from the repressor domain can produce efficient and gene-specific transcriptional silencing. To interfere with RHODOPSIN (RHO) gain-of-function mutations we engineered the ZF6-DNA-binding protein (ZF6-DB) that targets 20 base pairs (bp) of a RHOcis-regulatory element (CRE) and demonstrate Rho specific transcriptional silencing upon adeno-associated viral (AAV) vector-mediated expression in photoreceptors. The data show that the 20 bp-long genomic DNA sequence is necessary for RHO expression and that photoreceptor delivery of the corresponding cognate synthetic trans-acting factor ZF6-DB without the intrinsic transcriptional repression properties of the canonical ED blocks Rho expression with negligible genome-wide transcript perturbations. The data support DNA-binding-mediated silencing as a novel mode to treat gain-of-function mutations.


Assuntos
Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Inativação Gênica , Proteínas Recombinantes/metabolismo , Rodopsina/biossíntese , Transcrição Gênica , Adenoviridae/genética , Proteínas de Ligação a DNA/genética , Expressão Gênica , Vetores Genéticos , Ligação Proteica , Proteínas Recombinantes/genética , Transdução Genética
12.
EMBO Mol Med ; 6(2): 194-211, 2014 02.
Artigo em Inglês | MEDLINE | ID: mdl-24150896

RESUMO

Retinal gene therapy with adeno-associated viral (AAV) vectors is safe and effective in humans. However, AAV's limited cargo capacity prevents its application to therapies of inherited retinal diseases due to mutations of genes over 5 kb, like Stargardt's disease (STGD) and Usher syndrome type IB (USH1B). Previous methods based on 'forced' packaging of large genes into AAV capsids may not be easily translated to the clinic due to the generation of genomes of heterogeneous size which raise safety concerns. Taking advantage of AAV's ability to concatemerize, we generated dual AAV vectors which reconstitute a large gene by either splicing (trans-splicing), homologous recombination (overlapping), or a combination of the two (hybrid). We found that dual trans-splicing and hybrid vectors transduce efficiently mouse and pig photoreceptors to levels that, albeit lower than those achieved with a single AAV, resulted in significant improvement of the retinal phenotype of mouse models of STGD and USH1B. Thus, dual AAV trans-splicing or hybrid vectors are an attractive strategy for gene therapy of retinal diseases that require delivery of large genes.


Assuntos
Dependovirus/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos/metabolismo , Retina/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Modelos Animais de Doenças , Vetores Genéticos/administração & dosagem , Células HEK293 , Humanos , Injeções , Lipofuscina/metabolismo , Degeneração Macular/congênito , Degeneração Macular/genética , Degeneração Macular/terapia , Melanossomas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miosina VIIa , Miosinas/metabolismo , Fenótipo , Células Fotorreceptoras de Vertebrados/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Rodopsina/metabolismo , Doença de Stargardt , Sus scrofa , Trans-Splicing/genética , Transdução Genética , Síndromes de Usher/genética , Síndromes de Usher/terapia
13.
Hum Gene Ther ; 24(12): 982-92, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24067103

RESUMO

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.


Assuntos
Terapia Genética , Amaurose Congênita de Leber/terapia , Degeneração Retiniana/terapia , Animais , Dependovirus/genética , Modelos Animais de Doenças , Expressão Gênica , Humanos , Amaurose Congênita de Leber/genética , Camundongos , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Cones/patologia , Degeneração Retiniana/genética , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/patologia , Transdução Genética
14.
PLoS One ; 8(8): e72027, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23991031

RESUMO

Mutations in MYO7A cause autosomal recessive Usher syndrome type IB (USH1B), one of the most frequent conditions that combine severe congenital hearing impairment and retinitis pigmentosa. A promising therapeutic strategy for retinitis pigmentosa is gene therapy, however its pre-clinical development is limited by the mild retinal phenotype of the shaker1 (sh1(-/-)) murine model of USH1B which lacks both retinal functional abnormalities and degeneration. Here we report a significant, early-onset delay of sh1(-/-) photoreceptor ability to recover from light desensitization as well as a progressive reduction of both b-wave electroretinogram amplitude and light sensitivity, in the absence of significant loss of photoreceptors up to 12 months of age. We additionally show that subretinal delivery to the sh1(-/-) retina of AAV vectors encoding the large MYO7A protein results in significant improvement of sh1(-/-) photoreceptor and retinal pigment epithelium ultrastructural anomalies which is associated with improvement of recovery from light desensitization. These findings provide new tools to evaluate the efficacy of experimental therapies for USH1B. In addition, although AAV vectors expressing large genes might have limited clinical applications due to their genome heterogeneity, our data show that AAV-mediated MYO7A gene transfer to the sh1(-/-) retina is effective.


Assuntos
Terapia Genética/métodos , Miosinas/deficiência , Retina/fisiopatologia , Degeneração Retiniana/terapia , Adulto , Animais , Western Blotting , Dependovirus/genética , Modelos Animais de Doenças , Eletrorretinografia , Olho/metabolismo , Olho/fisiopatologia , Olho/ultraestrutura , Feminino , Vetores Genéticos/genética , Células HEK293 , Humanos , Masculino , Melanossomas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Microscopia Eletrônica , Miosina VIIa , Miosinas/genética , Miosinas/metabolismo , Retina/metabolismo , Retina/ultraestrutura , Degeneração Retiniana/genética , Degeneração Retiniana/fisiopatologia , Síndromes de Usher/genética , Síndromes de Usher/fisiopatologia , Síndromes de Usher/terapia
15.
PLoS One ; 8(3): e59025, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23520549

RESUMO

Recombinant adeno-associated viral (AAV) vectors are known to safely and efficiently transduce the retina. Among the various AAV serotypes available, AAV2/5 and 2/8 are the most effective for gene transfer to photoreceptors (PR), which are the most relevant targets for gene therapy of inherited retinal degenerations. However, the search for novel AAV serotypes with improved PR transduction is ongoing. In this work we tested vectors derived from five AAV serotypes isolated from porcine tissues (referred to as porcine AAVs, four of which are newly identified) for their ability to transduce both the murine and the cone-enriched pig retina. Porcine AAV vectors expressing EGFP under the control of the CMV promoter were injected subretinally either in C57BL/6 mice or Large White pigs. The resulting retinal tropism was analyzed one month later on histological sections, while levels of PR transduction were assessed by Western blot. Our results show that all porcine AAV transduce murine and porcine retinal pigment epithelium and PR upon subretinal administration. AAV2/po1 and 2/po5 are the most efficient porcine AAVs for murine PR transduction and exhibit the strongest tropism for pig cone PR. The levels of PR transduction obtained with AAV2/po1 and 2/po5 are similar, albeit not superior, to those obtained with AAV2/5 and AAV2/8, which evinces AAV2/po1 and 2/po5 to be promising vectors for retinal gene therapy.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Retina , Transdução Genética/métodos , Animais , Linhagem Celular , Camundongos , Suínos
16.
PLoS One ; 6(7): e22166, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21818300

RESUMO

BACKGROUND: Gene transfer using adeno-associated viral (AAV) vectors has been successfully applied in the retina for the treatment of inherited retinal dystrophies. Recently, microRNAs have been exploited to fine-tune transgene expression improving therapeutic outcomes. Here we evaluated the ability of retinal-expressed microRNAs to restrict AAV-mediated transgene expression to specific retinal cell types that represent the main targets of common inherited blinding conditions. METHODOLOGY/PRINCIPAL FINDINGS: To this end, we generated AAV2/5 vectors expressing EGFP and containing four tandem copies of miR-124 or miR-204 complementary sequences in the 3'UTR of the transgene expression cassette. These vectors were administered subretinally to adult C57BL/6 mice and Large White pigs. Our results demonstrate that miR-124 and miR-204 target sequences can efficiently restrict AAV2/5-mediated transgene expression to retinal pigment epithelium and photoreceptors, respectively, in mice and pigs. Interestingly, transgene restriction was observed at low vector doses relevant to therapy. CONCLUSIONS: We conclude that microRNA-mediated regulation of transgene expression can be applied in the retina to either restrict to a specific cell type the robust expression obtained using ubiquitous promoters or to provide an additional layer of gene expression regulation when using cell-specific promoters.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/metabolismo , Retina/metabolismo , Transgenes/genética , Animais , Sequência de Bases , Dependovirus/genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Dados de Sequência Molecular , Especificidade de Órgãos/genética , Células Fotorreceptoras de Vertebrados/citologia , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/citologia , Epitélio Pigmentado da Retina/metabolismo , Sus scrofa , Transdução Genética
17.
Invest Ophthalmol Vis Sci ; 52(8): 5618-24, 2011 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-21474771

RESUMO

PURPOSE: To evaluate the suitability of gene delivery-based approaches as potential treatment of Leber congenital amaurosis 4 (LCA4) due to AIPL1 mutations. METHODS: Genomic DNA from patients was analyzed using a microarray chip and direct sequencing. A detailed clinical evaluation including fundus autofluorescence (FAF) and optical coherence tomography (OCT) was performed in patients with AIPL1 mutations. Aipl1 null mice and porcine eyes were subretinally injected with adeno-associated viral (AAV) vectors harboring the human AIPL1 coding sequence. RESULTS: We identified 10 LCA4 patients with mutations in AIPL1. The p.W278X sequence variation was the one most frequently found. Clinical assessment revealed common features including diffuse retinal dystrophies and maculopathy. However, optical coherence tomography showed partially retained photoreceptors in extramacular regions at all ages. The fundus autofluorescence was elicitable at the posterior pole and absent in the fovea. AAV-mediated gene transfer in Aipl1 -/- mice was associated with restoration of AIPL1 and ßPDE expression in photoreceptors and protection from degeneration. Administration of a clinically relevant dose of AAV2/8-AIPL1 to the preclinical large porcine retina resulted in high level of AIPL1 photoreceptor expression in the absence of toxicity. CONCLUSIONS: Using advanced imaging diagnostics we showed that maculopathy is a main feature of LCA4. We identified retinal areas at the posterior pole with surviving photoreceptors present even in adult LCA4 patients, which could be the target of gene therapy. The possible use of gene therapy for LCA4 is additionally supported by the protection from photoreceptor degeneration observed in Aipl 1-/- mice and by the high levels of photoreceptor transduction in the absence of toxicity observed after AAV2/8 delivery to the large porcine retina.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Transporte/genética , Proteínas do Olho/genética , Terapia Genética/métodos , Amaurose Congênita de Leber , Adolescente , Adulto , Idoso , Animais , Criança , Pré-Escolar , Modelos Animais de Doenças , Eletrorretinografia , Vetores Genéticos , Humanos , Itália , Amaurose Congênita de Leber/genética , Amaurose Congênita de Leber/patologia , Amaurose Congênita de Leber/terapia , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Sus scrofa , Tomografia de Coerência Óptica , Adulto Jovem
18.
Mol Ther ; 17(8): 1347-54, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19436266

RESUMO

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.


Assuntos
Albinismo Oculocutâneo/metabolismo , Albinismo Oculocutâneo/terapia , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Melaninas/metabolismo , Monofenol Mono-Oxigenase/fisiologia , Retina/metabolismo , Albinismo Oculocutâneo/patologia , Albinismo Oculocutâneo/ultraestrutura , Animais , Eletrofisiologia , Humanos , Iris/metabolismo , Iris/patologia , Iris/ultraestrutura , Melanócitos/metabolismo , Melanócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Monofenol Mono-Oxigenase/genética , Retina/patologia , Retina/ultraestrutura , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA