Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Genes (Basel) ; 11(12)2020 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-33261050

RESUMO

RPE65 isomerase, expressed in the retinal pigmented epithelium (RPE), is an enzymatic component of the retinoid cycle, converting all-trans retinyl ester into 11-cis retinol, and it is essential for vision, because it replenishes the photon capturing 11-cis retinal. To date, almost 200 loss-of-function mutations have been identified within the RPE65 gene causing inherited retinal dystrophies, most notably Leber congenital amaurosis (LCA) and autosomal recessive retinitis pigmentosa (arRP), which are both severe and early onset disease entities. We previously reported a mutation, D477G, co-segregating with the disease in a late-onset form of autosomal dominant RP (adRP) with choroidal involvement; uniquely, it is the only RPE65 variant to be described with a dominant component. Families or individuals with this variant have been encountered in five countries, and a number of subsequent studies have been reported in which the molecular biological and physiological properties of the variant have been studied in further detail, including observations of possible novel functions in addition to reduced RPE65 enzymatic activity. With regard to the latter, a human phase 1b proof-of-concept study has recently been reported in which aspects of remaining vision were improved for up to one year in four of five patients with advanced disease receiving a single one-week oral dose of 9-cis retinaldehyde, which is the first report showing efficacy and safety of an oral therapy for a dominant form of RP. Here, we review data accrued from published studies investigating molecular mechanisms of this unique variant and include hitherto unpublished material on the clinical spectrum of disease encountered in patients with the D477G variant, which, in many cases bears striking similarities to choroideremia.


Assuntos
Substituição de Aminoácidos , Genes Dominantes , Mutação de Sentido Incorreto , Mutação Puntual , Retinose Pigmentar/genética , cis-trans-Isomerases/genética , Idade de Início , Animais , Coroideremia , Ensaios Clínicos Fase I como Assunto , DNA Complementar/administração & dosagem , DNA Complementar/genética , Terapia de Reposição de Enzimas , Feminino , Técnicas de Introdução de Genes , Terapia Genética , Vetores Genéticos/uso terapêutico , Humanos , Amaurose Congênita de Leber/enzimologia , Amaurose Congênita de Leber/genética , Masculino , Camundongos , Linhagem , Estudo de Prova de Conceito , Isoformas de Proteínas/genética , Retinaldeído/uso terapêutico , Retinose Pigmentar/diagnóstico por imagem , Retinose Pigmentar/enzimologia , Retinose Pigmentar/terapia , cis-trans-Isomerases/deficiência , cis-trans-Isomerases/fisiologia , cis-trans-Isomerases/uso terapêutico
2.
Hum Mol Genet ; 26(7): 1230-1246, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28158775

RESUMO

Intraocular pressure (IOP) is maintained as a result of the balance between production of aqueous humour (AH) by the ciliary processes and hydrodynamic resistance to its outflow through the conventional outflow pathway comprising the trabecular meshwork (TM) and Schlemm's canal (SC). Elevated IOP, which can be caused by increased resistance to AH outflow, is a major risk factor for open-angle glaucoma. Matrix metalloproteinases (MMPs) contribute to conventional aqueous outflow homeostasis in their capacity to remodel extracellular matrices, which has a direct impact on aqueous outflow resistance and IOP. We observed decreased MMP-3 activity in human glaucomatous AH compared to age-matched normotensive control AH. Treatment with glaucomatous AH resulted in significantly increased transendothelial resistance of SC endothelial and TM cell monolayers and reduced monolayer permeability when compared to control AH, or supplemented treatment with exogenous MMP-3.Intracameral inoculation of AAV-2/9 containing a CMV-driven MMP-3 gene (AAV-MMP-3) into wild type mice resulted in efficient transduction of corneal endothelium and an increase in aqueous concentration and activity of MMP-3. Most importantly, AAV-mediated expression of MMP-3 increased outflow facility and decreased IOP, and controlled expression using an inducible promoter activated by topical administration of doxycycline achieved the same effect. Ultrastructural analysis of MMP-3 treated matrices by transmission electron microscopy revealed remodelling and degradation of core extracellular matrix components. These results indicate that periodic induction, via use of an eye drop, of AAV-mediated secretion of MMP-3 into AH could have therapeutic potential for those cases of glaucoma that are sub-optimally responsive to conventional pressure-reducing medications.


Assuntos
Dependovirus/genética , Glaucoma/terapia , Pressão Intraocular/genética , Metaloproteinase 3 da Matriz/genética , Animais , Humor Aquoso/metabolismo , Modelos Animais de Doenças , Endotélio Corneano/metabolismo , Endotélio Corneano/patologia , Glaucoma/genética , Glaucoma/patologia , Humanos , Metaloproteinase 3 da Matriz/uso terapêutico , Camundongos , Soluções Oftálmicas/uso terapêutico
3.
Sci Transl Med ; 6(230): 230ra44, 2014 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-24695684

RESUMO

Age-related macular degeneration (AMD) is the most common form of central retinal blindness globally. Distinct processes of the innate immune system, specifically activation of the NLRP3 inflammasome, have been shown to play a central role in the development of both "dry" and neovascular ("wet") forms of the disease. We show that the inflammatory cytokine interleukin-18 (IL-18) can regulate choroidal neovascularization formation in mice. We observed that exogenous administration of mature recombinant IL-18 has no effect on retinal pigment epithelial (RPE) cell viability, but that overexpression of pro-IL-18 or pro-IL-1ß alone can cause RPE cell swelling and subsequent atrophy, a process that can be inhibited by the promotion of autophagy. A direct comparison of local and systemic administration of mature recombinant IL-18 with current anti-VEGF (vascular endothelial growth factor)-based therapeutic strategies shows that IL-18 treatment works effectively alone and more effectively in combination with anti-VEGF therapy and represents a novel therapeutic strategy for the treatment of wet AMD.


Assuntos
Neovascularização de Coroide/tratamento farmacológico , Neovascularização de Coroide/prevenção & controle , Interleucina-18/uso terapêutico , Degeneração Macular/tratamento farmacológico , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Neovascularização de Coroide/complicações , Neovascularização de Coroide/patologia , Hematopoese/efeitos dos fármacos , Humanos , Interleucina-18/farmacologia , Interleucina-1beta/farmacologia , Interleucina-1beta/uso terapêutico , Injeções Intravítreas , Lasers , Degeneração Macular/complicações , Degeneração Macular/patologia , Camundongos , Modelos Biológicos , Permeabilidade/efeitos dos fármacos , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/patologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Nat Commun ; 3: 849, 2012 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-22617289

RESUMO

Traumatic brain injury is the leading cause of death in children and young adults globally. Malignant cerebral oedema has a major role in the pathophysiology that evolves after severe traumatic brain injury. Added to this is the significant morbidity and mortality from cerebral oedema associated with acute stroke, hypoxic ischemic coma, neurological cancers and brain infection. Therapeutic strategies to prevent cerebral oedema are limited and, if brain swelling persists, the risks of permanent brain damage or mortality are greatly exacerbated. Here we show that a temporary and size-selective modulation of the blood-brain barrier allows enhanced movement of water from the brain to the blood and significantly impacts on brain swelling. We also show cognitive improvement in mice with focal cerebral oedema following administration in these animals of short interfering RNA directed against claudin-5. These observations may have profound consequences for early intervention in cases of traumatic brain injury, or indeed any neurological condition where cerebral oedema is the hallmark pathology.


Assuntos
Edema Encefálico/etiologia , Edema Encefálico/terapia , Lesões Encefálicas/complicações , Claudinas/metabolismo , Cognição/fisiologia , Animais , Barreira Hematoencefálica/metabolismo , Edema Encefálico/diagnóstico por imagem , Lesões Encefálicas/diagnóstico por imagem , Criança , Claudina-5 , Claudinas/genética , Humanos , Pressão Intracraniana/fisiologia , Masculino , Camundongos , Interferência de RNA , Tomografia Computadorizada por Raios X
5.
EMBO Mol Med ; 3(4): 235-45, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21374818

RESUMO

We describe a procedure for controlled, periodic, reversible modulation of selected regions of the blood-brain barrier (BBB) or the inner-blood-retina barrier (iBRB) based on incorporation into an AAV-2/9 vector of a doxycycline-inducible gene encoding shRNA targeting claudin-5, one of 30 or so proteins constituting the BBB and iBRB. The vector may be introduced stereotaxically into pre-selected regions of the brain or into the retina, rendering these regions permeable to low-molecular weight compounds up to approximately 1 kDa for the period of time during which the inducing agent, doxycycline, is administered in drinking water, but excluding potentially toxic higher molecular weight materials. We report on the use of barrier modulation in tandem with systemic drug therapy to prevent retinal degeneration and to suppress laser-induced choroidal neovascularization (CNV), the latter being the hallmark pathology associated with the exudative, or wet, form of age-related macular degeneration (AMD). These observations constitute the basis of a minimally invasive systemic therapeutic modality for retinal diseases, including retinitis pigmentosa and AMD, where, in early stage disease, the iBRB is intact and impervious to systemically administered drugs.


Assuntos
Barreira Hematorretiniana/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Terapia Genética , Degeneração Macular/terapia , Animais , Barreira Hematorretiniana/inervação , Barreira Hematorretiniana/metabolismo , Linhagem Celular , Claudina-5 , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Degeneração Macular/genética , Degeneração Macular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Peso Molecular , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/uso terapêutico
6.
Adv Exp Med Biol ; 664: 559-65, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20238059

RESUMO

Recombinant adeno-associated viral (rAAV) vectors have recently been widely used for the delivery of therapeutic transgenes in preclinical and clinical studies for inherited retinal degenerative diseases. Interchanging capsid genes between different AAV serotypes has enabled selective delivery of transgene into specific cell type(s) of the retina. The RP10 form of autosomal dominant retinitis pigmentosa (adRP) is caused by missense mutations within the gene encoding inosine 5'-monophosphate dehydrogenase type 1. Here, we report that the use of rAAV2/5 vectors expressing shRNA targeting mutant IMPDH1 prevents photoreceptor degeneration, and preserves synaptic connectivity in a mouse model of RP10.


Assuntos
Citoproteção , Modelos Animais de Doenças , Células Fotorreceptoras de Vertebrados/patologia , Retinose Pigmentar/patologia , Animais , Dependovirus/genética , Regulação para Baixo/genética , Genes Supressores , Células HeLa , Humanos , IMP Desidrogenase/genética , IMP Desidrogenase/metabolismo , Camundongos , Proteínas Mutantes/metabolismo , Mutação/genética , Células Fotorreceptoras de Vertebrados/enzimologia , Recombinação Genética/genética , Retinose Pigmentar/enzimologia , Supressão Genética , Sinapses/metabolismo
7.
Proc Natl Acad Sci U S A ; 106(42): 17817-22, 2009 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-19822744

RESUMO

Degenerative retinopathies, including age-related macular degeneration, diabetic retinopathy, and hereditary retinal disorders--major causes of world blindness--are potentially treatable by using low-molecular weight neuroprotective, antiapoptotic, or antineovascular drugs. These agents are, however, not in current systemic use owing to, among other factors, their inability to passively diffuse across the microvasculature of the retina because of the presence of the inner blood-retina barrier (iBRB). Moreover, preclinical assessment of the efficacies of new formulations in the treatment of such conditions is similarly compromised. We describe here an experimental process for RNAi-mediated, size-selective, transient, and reversible modulation of the iBRB in mice to molecules up to 800 Da by suppression of transcripts encoding claudin-5, a protein component of the tight junctions of the inner retinal vasculature. MRI produced no evidence indicative of brain or retinal edema, and the process resulted in minimal disturbance of global transcriptional patterns analyzed in neuronal tissue. We show that visual function can be improved in IMPDH1(-/-) mice, a model of autosomal recessive retinitis pigmentosa, and that the rate of photoreceptor cell death can be reduced in a model of light-induced retinal degeneration by systemic drug delivery after reversible barrier opening. These findings provide a platform for high-throughput drug screening in models of retinal degeneration, and they ultimately could result in the development of a novel "humanized" approach to therapy for conditions with little or no current forms of treatment.


Assuntos
Barreira Hematorretiniana/efeitos dos fármacos , Barreira Hematorretiniana/metabolismo , Sistemas de Liberação de Medicamentos , Oligopeptídeos/administração & dosagem , Animais , Calpaína/antagonistas & inibidores , Claudina-5 , Inibidores de Cisteína Proteinase/administração & dosagem , Modelos Animais de Doenças , Eletrorretinografia , Guanosina Trifosfato/administração & dosagem , Guanosina Trifosfato/metabolismo , Humanos , IMP Desidrogenase/deficiência , IMP Desidrogenase/genética , Imageamento por Ressonância Magnética , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Interferência de RNA , RNA Interferente Pequeno/genética , Retina/efeitos dos fármacos , Retina/metabolismo , Retinose Pigmentar/tratamento farmacológico , Retinose Pigmentar/genética , Retinose Pigmentar/metabolismo
8.
J Gene Med ; 10(8): 930-47, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18509865

RESUMO

BACKGROUND: The blood-brain barrier (BBB) contains tight junctions (TJs) which reduce the space between adjacent endothelial cells lining the fine capillaries of the microvasculature of the brain to form a selective and regulatable barrier. METHODS: Using a hydrodynamic approach, we delivered siRNA targeting the TJ protein claudin-5 to the endothelial cells of the BBB in mice. RESULTS: We have shown a significant decrease in claudin-5 mRNA levels 24 and 48 hours post-delivery of siRNA, with levels of protein expression decreasing up to 48 hours post-injection compared to uninjected, phosphate-buffered saline (PBS)-injected and non-targeting siRNA-injected mice. We observed increased permeability at the BBB to molecules up to 742 Da, but not 4400 Da, using tracer molecule perfusion and MRI analysis. To illustrate the functional efficacy of size-selective and transient barrier opening, we have shown that enhanced delivery of the small neuropeptide thyrotropin-releasing hormone (TRH) (MW 360 Da) to the brains of mice 48 hours post-injection of siRNA targeting claudin-5 significantly modifies behavioural output. CONCLUSIONS: These data demonstrate that it is now possible to transiently and size-selectively open the BBB in mice, allowing in principle the delivery of a wide range of agents for the establishment and treatment of experimental mouse models of neurodegenerative, neuropsychiatric and malignant diseases.


Assuntos
Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Proteínas de Membrana/metabolismo , Interferência de RNA , Animais , Barreira Hematoencefálica/diagnóstico por imagem , Permeabilidade Capilar/fisiologia , Claudina-5 , Técnica Indireta de Fluorescência para Anticorpo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Radiografia , Junções Íntimas/metabolismo , Fatores de Tempo
9.
Hum Mol Genet ; 17(14): 2084-100, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18385099

RESUMO

Mutations within the inosine 5'-monophosphate dehydrogenase 1 (IMPDH1) gene cause the RP10 form of autosomal dominant retinitis pigmentosa (adRP), an early-onset retinopathy resulting in extensive visual handicap owing to progressive death of photoreceptors. Apart from the prevalence of RP10, estimated to account for 5-10% of cases of adRP in United States and Europe, two observations render this form of RP an attractive target for gene therapy. First, we show that while recombinant adeno-associated viral (AAV)-mediated expression of mutant human IMPDH1 protein in the mouse retina results in an aggressive retinopathy modelling the human counterpart, expression of a normal human IMPDH1 gene under similar conditions has no observable pathological effect on retinal function, indicating that over-expression of a therapeutic replacement gene may be relatively well tolerated. Secondly, complete absence of IMPDH1 protein in mice with a targeted disruption of the gene results in relatively mild retinal dysfunction, suggesting that significant therapeutic benefit may be derived even from the suppression-only component of an RNAi-based gene therapy. We show that AAV-mediated co-expression in the murine retina of a mutant human IMPDH1 gene together with short hairpin RNAs (shRNA) validated in vitro and in vivo, targeting both human and mouse IMPDH1, substantially suppresses the negative pathological effects of mutant IMPDH1, at a point where, in the absence of shRNA, expression of mutant protein in the RP10 model essentially ablates all photoreceptors in transfected areas of the retina. These data strongly suggest that an RNAi-mediated approach to therapy for RP10 holds considerable promise for human subjects.


Assuntos
Terapia Genética , IMP Desidrogenase/genética , RNA Interferente Pequeno/genética , Retinose Pigmentar/terapia , Animais , Sequência de Bases , Dependovirus/genética , Regulação para Baixo , Genes Dominantes , Vetores Genéticos/genética , Células HeLa , Humanos , IMP Desidrogenase/metabolismo , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Dados de Sequência Molecular , Mutação , Interferência de RNA , RNA Interferente Pequeno/química , Transdução Genética
10.
Am J Hum Genet ; 81(1): 127-35, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17564969

RESUMO

Mutational heterogeneity represents a significant barrier to development of therapies for many dominantly inherited diseases. For example, >100 mutations in the rhodopsin gene (RHO) have been identified in patients with retinitis pigmentosa (RP). The development of therapies for dominant disorders that correct the primary genetic lesion and overcome mutational heterogeneity is challenging. Hence, therapeutics comprising two elements--gene suppression in conjunction with gene replacement--have been investigated. Suppression is targeted to a site independent of the mutation; therefore, both mutant and wild-type alleles are suppressed. In parallel with suppression, a codon-modified replacement gene refractory to suppression is provided. Both in vitro and in vivo validation of suppression and replacement for RHO-linked RP has been undertaken in the current study. RNA interference (RNAi) has been used to achieve ~90% in vivo suppression of RHO in photoreceptors, with use of adeno-associated virus (AAV) for delivery. Demonstration that codon-modifed RHO genes express functional wild-type protein has been explored transgenically, together with in vivo expression of AAV-delivered RHO-replacement genes in the presence of targeting RNAi molecules. Observation of potential therapeutic benefit from AAV-delivered suppression and replacement therapies has been obtained in Pro23His mice. Results provide the first in vivo indication that suppression and replacement can provide a therapeutic solution for dominantly inherited disorders such as RHO-linked RP and can be employed to circumvent mutational heterogeneity.


Assuntos
Terapia Genética/métodos , Interferência de RNA , Retinose Pigmentar/terapia , Rodopsina/genética , Supressão Genética , Adenoviridae/genética , Animais , Sequência de Bases , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , RNA Interferente Pequeno/genética , Retina/química , Retina/metabolismo , Retina/patologia , Retinose Pigmentar/patologia , Rodopsina/análise
11.
Eur J Hum Genet ; 12(10): 864-6, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15241481

RESUMO

Given that mutant COL1A1 is known to cause Osteogenesis Imperfecta (OI), tools to modulate COL1A1 expression are likely to be of significant therapeutic value. In this context, we have evaluated RNA interference (RNAi) as a means to downregulate COL1A1 expression in Cos-7 cells and in human mesenchymal progenitor stem cells (MPCs), the latter cells giving rise to bone and therefore representing a target cell type for collagen-related disorders. In addition, allele-specificity, a key factor to the success of RNAi-based suppression, was explored with a view to developing a mutation-independent RNAi-based therapeutic for OI by targeting an intragenic SNP within transcripts derived from the COL1A1 gene. Preferential suppression of individual polymorphic alleles that differed by a single nucleotide was observed.


Assuntos
Colágeno Tipo I/genética , Células-Tronco Mesenquimais/metabolismo , Osteogênese Imperfeita/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Alelos , Animais , Células COS , Chlorocebus aethiops , Colágeno Tipo I/biossíntese , Cadeia alfa 1 do Colágeno Tipo I , Regulação para Baixo , Terapia Genética/métodos , Humanos , Mutação/genética , Osteogênese Imperfeita/metabolismo , Osteogênese Imperfeita/terapia , Polimorfismo de Nucleotídeo Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA