Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Exp Eye Res ; 165: 175-181, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28974356

RESUMO

Retinal degenerations, including age-related macular degeneration and the retinitis pigmentosa family of diseases, are among the leading causes of legal blindness in the United States. We previously found that Stanniocalcin-1 (STC-1) reduced photoreceptor loss in the S334ter-3 and Royal College of Surgeons rat models of retinal degeneration. The results were attributed in part to a reduction in oxidative stress. Herein, we tested the hypothesis that long-term delivery of STC-1 would provide therapeutic rescue in more chronic models of retinal degeneration. To achieve sustained delivery, we produced an adeno-associated virus (AAV) construct to express STC-1 (AAV-STC-1) under the control of a retinal ganglion cell targeting promoter human synapsin 1 (hSYN1). AAV-STC-1 was injected intravitreally into the P23H-1 and S334ter-4 rhodopsin transgenic rats at postnatal day 10. Tissues were collected at postnatal day 120 for confirmation of STC-1 overexpression and histologic and molecular analysis. Electroretinography (ERG) was performed in a cohort of animals at that time. Overexpression of STC-1 resulted in a significant preservation of photoreceptors as assessed by outer nuclear thickness in the P23H-1 (P < 0.05) and the S334ter-4 (P < 0.005) models compared to controls. Additionally, retinal function was significantly improved in the P23H-1 model with overexpressed STC-1 as assessed by ERG analysis (scotopic b-wave P < 0.005 and photopic b-wave P < 0.05). Microarray analysis identified common downstream gene expression changes that occurred in both models. Genes of interest based on their function were selected for validation by quantitative real-time PCR and were significantly increased in the S334ter-4 model.


Assuntos
Dependovirus , Glicoproteínas/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Retinose Pigmentar/tratamento farmacológico , Animais , Modelos Animais de Doenças , Eletrorretinografia , Glicoproteínas/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Células Fotorreceptoras de Vertebrados/patologia , Ratos , Ratos Transgênicos , Retinose Pigmentar/patologia
2.
Hum Genet ; 135(3): 327-43, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26825853

RESUMO

MERTK is an essential component of the signaling network that controls phagocytosis in retinal pigment epithelium (RPE), the loss of which results in photoreceptor degeneration. Previous proof-of-concept studies have demonstrated the efficacy of gene therapy using human MERTK (hMERTK) packaged into adeno-associated virus (AAV2) in treating RCS rats and mice with MERTK deficiency. The purpose of this study was to assess the safety of gene transfer via subretinal administration of rAAV2-VMD2-hMERTK in subjects with MERTK-associated retinitis pigmentosa (RP). After a preclinical phase confirming the safety of the study vector in monkeys, six patients (aged 14 to 54, mean 33.3 years) with MERTK-related RP and baseline visual acuity (VA) ranging from 20/50 to <20/6400 were entered in a phase I open-label, dose-escalation trial. One eye of each patient (the worse-seeing eye in five subjects) received a submacular injection of the viral vector, first at a dose of 150 µl (5.96 × 10(10)vg; 2 patients) and then 450 µl (17.88 × 10(10)vg; 4 patients). Patients were followed daily for 10 days at 30, 60, 90, 180, 270, 365, 540, and 730 days post-injection. Collected data included (1) full ophthalmologic examination including best-corrected VA, intraocular pressure, color fundus photographs, macular spectral domain optical coherence tomography and full-field stimulus threshold test (FST) in both the study and fellow eyes; (2) systemic safety data including CBC, liver and kidney function tests, coagulation profiles, urine analysis, AAV antibody titers, peripheral blood PCR and ASR measurement; and (3) listing of ophthalmological or systemic adverse effects. All patients completed the 2-year follow-up. Subretinal injection of rAAV2-VMD2-hMERTK was associated with acceptable ocular and systemic safety profiles based on 2-year follow-up. None of the patients developed complications that could be attributed to the gene vector with certainty. Postoperatively, one patient developed filamentary keratitis, and two patients developed progressive cataract. Of these two patients, one also developed transient subfoveal fluid after the injection as well as monocular oscillopsia. Two patients developed a rise in AAV antibodies, but neither patient was positive for rAAV vector genomes via PCR. Three patients also displayed measurable improved visual acuity in the treated eye following surgery, although the improvement was lost by 2 years in two of these patients. Gene therapy for MERTK-related RP using careful subretinal injection of rAAV2-VMD2-hMERTK is not associated with major side effects and may result in clinical improvement in a subset of patients.


Assuntos
Terapia Genética/métodos , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Retinose Pigmentar/genética , Retinose Pigmentar/terapia , Adolescente , Adulto , Animais , Dependovirus/genética , Modelos Animais de Doenças , Determinação de Ponto Final , Feminino , Seguimentos , Vetores Genéticos , Humanos , Macaca , Masculino , Pessoa de Meia-Idade , Mutação , Complicações Pós-Operatórias/terapia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Líquido Sub-Retiniano , Tomografia de Coerência Óptica , Resultado do Tratamento , Acuidade Visual , Adulto Jovem , c-Mer Tirosina Quinase
3.
Adv Exp Med Biol ; 854: 487-93, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26427450

RESUMO

MERTK-associated retinal degenerations are thought to have defects in phagocytosis of shed outer segment membranes by the retinal pigment epithelium (RPE), as do the rodent models of these diseases. We have subretinally injected an RPE-specific AAV2 vector, AAV2-VMD2-hMERTK, to determine whether this would provide long-term photoreceptor rescue in the RCS rat, which it did for up to 6.5 months, the longest time point examined. Moreover, we found phagosomes in the RPE in the rescued regions of RCS retinas soon after the onset of light. The same vector also had a major protective effect in Mertk-null mice, with a concomitant increase in ERG response amplitudes in the vector-injected eyes. These findings suggest that planned clinical trials with this vector will have a favorable outcome.


Assuntos
Terapia Genética/métodos , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Degeneração Retiniana/genética , Degeneração Retiniana/terapia , Animais , Bestrofinas , Canais de Cloreto/genética , Dependovirus/genética , Modelos Animais de Doenças , Eletrorretinografia , Proteínas do Olho/genética , Vetores Genéticos/genética , Humanos , Camundongos Knockout , Fagocitose/genética , Fagocitose/fisiologia , Fagossomos/metabolismo , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/metabolismo , Ratos Mutantes , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases/deficiência , Receptores Proteína Tirosina Quinases/metabolismo , Degeneração Retiniana/fisiopatologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/fisiopatologia , Resultado do Tratamento , c-Mer Tirosina Quinase
4.
PLoS Genet ; 11(12): e1005723, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26656104

RESUMO

Inherited photoreceptor degenerations (IPDs) are the most genetically heterogeneous of Mendelian diseases. Many IPDs exhibit substantial phenotypic variability, but the basis is usually unknown. Mutations in MERTK cause recessive IPD phenotypes associated with the RP38 locus. We have identified a murine genetic modifier of Mertk-associated photoreceptor degeneration, the C57BL/6 (B6) allele of which acts as a suppressor. Photoreceptors degenerate rapidly in Mertk-deficient animals homozygous for the 129P2/Ola (129) modifier allele, whereas animals heterozygous for B6 and 129 modifier alleles exhibit an unusual intermixing of degenerating and preserved retinal regions, with females more severely affected than males. Mertk-deficient mice homozygous for the B6 modifier allele display degeneration only in the far periphery, even at 8 months of age, and have improved retinal function compared to animals homozygous for the 129 allele. We genetically mapped the modifier to an approximately 2-megabase critical interval that includes Tyro3, a paralog of Mertk. Tyro3 expression in the outer retina varies with modifier genotype in a manner characteristic of a cis-acting expression quantitative trait locus (eQTL), with the B6 allele conferring an approximately three-fold higher expression level. Loss of Tyro3 function accelerates the pace of photoreceptor degeneration in Mertk knockout mice, and TYRO3 protein is more abundant in the retinal pigment epithelium (RPE) adjacent to preserved central retinal regions of Mertk knockout mice homozygous for the B6 modifier allele. Endogenous human TYRO3 protein co-localizes with nascent photoreceptor outer segment (POS) phagosomes in a primary RPE cell culture assay, and expression of murine Tyro3 in cultured cells stimulates phagocytic ingestion of POS. Our findings demonstrate that Tyro3 gene dosage modulates Mertk-associated retinal degeneration, provide strong evidence for a direct role for TYRO3 in RPE phagocytosis, and suggest that an eQTL can modify a recessive IPD.


Assuntos
Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Degeneração Retiniana/genética , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Fagocitose , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patologia , Proteínas Proto-Oncogênicas/biossíntese , Receptores Proteína Tirosina Quinases/biossíntese , Retina/metabolismo , Retina/patologia , Degeneração Retiniana/patologia , Epitélio Pigmentado da Retina/patologia , c-Mer Tirosina Quinase
5.
Mol Biol Cell ; 25(9): 1411-20, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24623724

RESUMO

Endoplasmic reticulum (ER) protein misfolding activates the unfolded protein response (UPR) to help cells cope with ER stress. If ER homeostasis is not restored, UPR promotes cell death. The mechanisms of UPR-mediated cell death are poorly understood. The PKR-like endoplasmic reticulum kinase (PERK) arm of the UPR is implicated in ER stress-induced cell death, in part through up-regulation of proapoptotic CCAAT/enhancer binding protein homologous protein (CHOP). Chop((-)/(-)) cells are partially resistant to ER stress-induced cell death, and CHOP overexpression alone does not induce cell death. These findings suggest that additional mechanisms regulate cell death downstream of PERK. Here we find dramatic suppression of antiapoptosis XIAP proteins in response to chronic ER stress. We find that PERK down-regulates XIAP synthesis through eIF2α and promotes XIAP degradation through ATF4. Of interest, PERK's down-regulation of XIAP occurs independently of CHOP activity. Loss of XIAP leads to increased cell death, whereas XIAP overexpression significantly enhances resistance to ER stress-induced cell death, even in the absence of CHOP. Our findings define a novel signaling circuit between PERK and XIAP that operates in parallel with PERK to CHOP induction to influence cell survival during ER stress. We propose a "two-hit" model of ER stress-induced cell death involving concomitant CHOP up-regulation and XIAP down-regulation both induced by PERK.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Apoptose , Estresse do Retículo Endoplasmático , Fator de Iniciação 2 em Eucariotos/metabolismo , Resposta a Proteínas não Dobradas , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Regulação para Baixo , Células HEK293 , Humanos , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional , Proteólise , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição CHOP/metabolismo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , eIF-2 Quinase/metabolismo
6.
PLoS One ; 8(12): e83974, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24391855

RESUMO

Melanopsin-expressing retinal ganglion cells (mRGCs) in the eye play an important role in many light-activated non-image-forming functions including neonatal photoaversion and the adult pupillary light reflex (PLR). MRGCs rely on glutamate and possibly PACAP (pituitary adenylate cyclase-activating polypeptide) to relay visual signals to the brain. However, the role of these neurotransmitters for individual non-image-forming responses remains poorly understood. To clarify the role of glutamatergic signaling from mRGCs in neonatal aversion to light and in adult PLR, we conditionally deleted vesicular glutamate transporter (VGLUT2) selectively from mRGCs in mice. We found that deletion of VGLUT2 in mRGCs abolished negative phototaxis and light-induced distress vocalizations in neonatal mice, underscoring a necessary role for glutamatergic signaling. In adult mice, loss of VGLUT2 in mRGCs resulted in a slow and an incomplete PLR. We conclude that glutamatergic neurotransmission from mRGCs is required for neonatal photoaversion but is complemented by another non-glutamatergic signaling mechanism for the pupillary light reflex in adult mice. We speculate that this complementary signaling might be due to PACAP neurotransmission from mRGCs.


Assuntos
Luz , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Reflexo Pupilar/fisiologia , Células Ganglionares da Retina/metabolismo , Opsinas de Bastonetes/fisiologia , Transmissão Sináptica/fisiologia , Proteína Vesicular 2 de Transporte de Glutamato/fisiologia , Animais , Animais Recém-Nascidos , Comportamento Animal , Feminino , Técnicas Imunoenzimáticas , Integrases/metabolismo , Transdução de Sinal Luminoso , Masculino , Camundongos , Camundongos Knockout , Neurotransmissores/metabolismo , Estimulação Luminosa , Reflexo Pupilar/efeitos da radiação , Células Ganglionares da Retina/efeitos da radiação , Transtornos da Visão , Visão Ocular/fisiologia , Visão Ocular/efeitos da radiação
7.
PLoS One ; 7(3): e33266, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22432009

RESUMO

The S334ter rhodopsin (Rho) rat (line 4) bears the rhodopsin gene with an early termination codon at residue 334 that is a model for several such mutations found in human patients with autosomal dominant retinitis pigmentosa (ADRP). The Unfolded Protein Response (UPR) is implicated in the pathophysiology of several retinal disorders including ADRP in P23H Rho rats. The aim of this study was to examine the onset of UPR gene expression in S334ter Rho retinas to determine if UPR is activated in ADRP animal models and to investigate how the activation of UPR molecules leads to the final demise of S334ter Rho photoreceptors. RT-PCR was performed to evaluate the gene expression profiles for the P10, P12, P15, and P21 stages of the development and progression of ADRP in S334ter Rho photoreceptors. We determined that during the P12-P15 period, ER stress-related genes are strongly upregulated in transgenic retinas, resulting in the activation of the UPR that was confirmed using western blot analysis and RT-PCR. The activation of UPR was associated with the increased expression of JNK, Bik, Bim, Bid, Noxa, and Puma genes and cleavage of caspase-12 that together with activated calpains presumably compromise the integrity of the mitochondrial MPTP, leading to the release of pro-apoptotic AIF1 into the cytosol of S334ter Rho photoreceptor cells. Therefore, two major cross-talking pathways, the UPR and mitochondrial MPTP occur in S334ter-4 Rho retina concomitantly and eventually promote the death of the photoreceptor cells.


Assuntos
Estresse do Retículo Endoplasmático , Mutação/genética , Degeneração Retiniana/patologia , Rodopsina/genética , Envelhecimento/genética , Envelhecimento/patologia , Animais , Apoptose/genética , Fator Apoptótico 1 Ativador de Proteases/metabolismo , Autofagia , Biomarcadores/metabolismo , Calpaína/metabolismo , Estresse do Retículo Endoplasmático/genética , Degradação Associada com o Retículo Endoplasmático/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo/genética , PTEN Fosfo-Hidrolase/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Retina/enzimologia , Retina/crescimento & desenvolvimento , Retina/patologia , Degeneração Retiniana/enzimologia , Degeneração Retiniana/genética , Fatores de Tempo , Fatores de Transcrição/metabolismo
8.
Invest Ophthalmol Vis Sci ; 53(4): 1895-904, 2012 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-22408006

RESUMO

PURPOSE: The absence of Mertk in RCS rats results in defective RPE phagocytosis, accumulation of outer segment (OS) debris in the subretinal space, and subsequent death of photoreceptors. Previous research utilizing Mertk gene replacement therapy in RCS rats provided proof of concept for treatment of this form of recessive retinitis pigmentosa (RP); however, the beneficial effects on retinal function were transient. In the present study, we evaluated whether delivery of a MERTK transgene using a tyrosine-mutant AAV8 capsid could lead to more robust and longer-term therapeutic outcomes than previously reported. METHODS: An AAV8 Y733F vector expressing a human MERTK cDNA driven by a RPE-selective promoter was administrated subretinally at postnatal day 2. Functional and morphological analyses were performed at 4 months and 8 months post-treatment. Retinal vasculature and Müller cell activation were analyzed by quantifying acellular capillaries and glial fibrillary acidic protein immunostaining, respectively. RESULTS: Electroretinographic responses from treated eyes were more than one-third of wild-type levels and OS were well preserved in the injection area even at 8 months. Rescue of RPE phagocytosis, prevention of retinal vasculature degeneration, and inhibition of Müller cell activation were demonstrated in the treated eyes for at least 8 months. CONCLUSIONS: This research describes a longer and much more robust functional and morphological rescue than previous studies. We also demonstrate for the first time that an AAV8 mutant capsid serotype vector has a substantial therapeutic potential for RPE-specific gene delivery. These results suggest that tyrosine-mutant AAV8 vectors hold promise for the treatment of individuals with MERTK-associated RP.


Assuntos
Terapia Genética/métodos , Proteínas Proto-Oncogênicas/administração & dosagem , Receptores Proteína Tirosina Quinases/administração & dosagem , Retinose Pigmentar/terapia , Animais , Animais Recém-Nascidos , Western Blotting , Modelos Animais de Doenças , Eletrorretinografia , Seguimentos , Vetores Genéticos , Humanos , Imuno-Histoquímica , Injeções , Microscopia Eletrônica de Transmissão , Mutação , Plasmídeos , Proteínas Proto-Oncogênicas/uso terapêutico , RNA/genética , Ratos , Ratos Mutantes , Receptores Proteína Tirosina Quinases/uso terapêutico , Retina , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/ultraestrutura , Retinose Pigmentar/genética , Retinose Pigmentar/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Tomografia de Coerência Óptica , Transgenes , Tirosina/genética , c-Mer Tirosina Quinase
11.
Invest Ophthalmol Vis Sci ; 52(7): 4703-9, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21436282

RESUMO

PURPOSE: To determine the basis and to characterize the phenotype of a chemically induced mutation in a mouse model of retinal degeneration. METHODS: Screening by indirect ophthalmoscopy identified a line of N-ethyl-N-nitrosourea (ENU) mutagenized mice demonstrating retinal patches. Longitudinal studies of retinal histologic sections showed photoreceptors in the peripheral retina undergoing slow, progressive degeneration. The mutation was named neuroscience mutagenesis facility 12 (nmf12), and mapping localized the critical region to Chromosome 2. RESULTS: Sequencing of nmf12 DNA revealed a point mutation in the c-mer tyrosine kinase gene, designated Mertk(nmf12). We detected elevated levels of tumor necrosis factor (Tnf, previously Tnfa) in retinas of Mertk(nmf12) homozygotes relative to wild-type controls and investigated whether the increase of TNF, an inflammatory cytokine produced by macrophages/monocytes that signals intracellularly to cause necrosis or apoptosis, could underlie the retinal degeneration observed in Mertk(nmf12) homozygotes. Mertk(nmf12) homozygous mice were mated to mice lacking the entire Tnf gene and partial coding sequences of the Lta (Tnfb) and Ltb (Tnfc) genes.(2) B6.129P2-Ltb/Tnf/Lta(tm1Dvk)/J homozygotes did not exhibit a retinal degeneration phenotype and will, hereafter, be referred to as Tnfabc(-/-) mice. Surprisingly, mice homozygous for both the Mertk(nmf12) and the Ltb/Tnf/Lta(tm1Dvk) allele (Tnfabc(-/-)) demonstrated an increase in the rate of retinal degeneration. CONCLUSIONS: These findings illustrate that a mutation in the Mertk gene leads to a significantly slower progressive retinal degeneration compared with other alleles of Mertk. These results demonstrate that TNF family members play a role in protecting photoreceptors of Mertk(nmf12) homozygotes from cell death.


Assuntos
DNA/genética , Etilnitrosoureia/toxicidade , Mutação/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Retina/metabolismo , Degeneração Retiniana/genética , Animais , Western Blotting , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Modelos Animais de Doenças , Progressão da Doença , Eletrorretinografia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Oftalmoscopia , Fenótipo , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Proteínas Proto-Oncogênicas/biossíntese , Receptores Proteína Tirosina Quinases/biossíntese , Retina/efeitos dos fármacos , Retina/patologia , Degeneração Retiniana/patologia , Degeneração Retiniana/fisiopatologia , Fator de Necrose Tumoral alfa/biossíntese , c-Mer Tirosina Quinase
12.
J Clin Invest ; 121(1): 369-83, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21135502

RESUMO

Retinal pigment epithelial (RPE) cell dysfunction plays a central role in various retinal degenerative diseases, but knowledge is limited regarding the pathways responsible for adult RPE stress responses in vivo. RPE mitochondrial dysfunction has been implicated in the pathogenesis of several forms of retinal degeneration. Here we have shown that postnatal ablation of RPE mitochondrial oxidative phosphorylation in mice triggers gradual epithelium dedifferentiation, typified by reduction of RPE-characteristic proteins and cellular hypertrophy. The electrical response of the retina to light decreased and photoreceptors eventually degenerated. Abnormal RPE cell behavior was associated with increased glycolysis and activation of, and dependence upon, the hepatocyte growth factor/met proto-oncogene pathway. RPE dedifferentiation and hypertrophy arose through stimulation of the AKT/mammalian target of rapamycin (AKT/mTOR) pathway. Administration of an oxidant to wild-type mice also caused RPE dedifferentiation and mTOR activation. Importantly, treatment with the mTOR inhibitor rapamycin blunted key aspects of dedifferentiation and preserved photoreceptor function for both insults. These results reveal an in vivo response of the mature RPE to diverse stressors that prolongs RPE cell survival at the expense of epithelial attributes and photoreceptor function. Our findings provide a rationale for mTOR pathway inhibition as a therapeutic strategy for retinal degenerative diseases involving RPE stress.


Assuntos
Degeneração Retiniana/etiologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Autofagia , Morte Celular , Desdiferenciação Celular/efeitos dos fármacos , Desdiferenciação Celular/fisiologia , Movimento Celular , Sobrevivência Celular , Feminino , Glicólise , Fator de Crescimento de Hepatócito/metabolismo , Hipertrofia , Masculino , Camundongos , Camundongos Transgênicos , Fosforilação Oxidativa , Células Fotorreceptoras de Vertebrados/efeitos dos fármacos , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Degeneração Retiniana/prevenção & controle , Epitélio Pigmentado da Retina/efeitos dos fármacos , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores
13.
Mol Vis ; 16: 1004-18, 2010 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-20577653

RESUMO

PURPOSE: In a previous study, several quantitative trait loci (QTL) that influence age-related degeneration (ageRD) were identified in a cross between the albino strains B6(Cg)-Tyr(c-2J)/J (B6a) and BALB/cByJ (C). The Chromosome (Chr) 6 and Chr 10 QTL were the strongest and most highly significant loci and both involved B6a protective alleles. The QTL were responsible for 21% and 9% of the variance in phenotypes, respectively. We focused on these two QTL to identify candidate genes. METHODS: DNA microarrays were used for the two mouse strains at four and eight months of age to identify genes that are differentially regulated and map to either QTL. Gene Ontology (GO) analysis of the differentially expressed genes was performed to identify possible processes and pathways associated with ageRD. To identify additional candidates, database analyses (Positional Medline or PosMed) were used. Based on differential expression, PosMed, and the presence of reported polymorphisms, five genes per QTL were selected for further study by sequencing analysis and qRT-PCR. Tumor necrosis factor, alpha- induced protein 3 (Tnfaip3; on a C57BL/6J (B6) background) was phenotypically tested. Single nucleotide polymorphisms (SNPs) flanking this gene were correlated with outer nuclear layer thickness (ONL), and eight-month-old Tnfaip3(+/-) mice were tested for ageRD. RESULTS: Polymorphisms were found in the coding regions of eight genes. Changes in gene expression were identified by qRT-PCR for Hexokinase 2 (Hk2) and Docking protein 1 (Dok1) at four months and for Dok1 and Tnfaip3 at eight months. Tnfaip3 was selected for phenotypic testing due to differential expression and the presence of two nonsynonymous mutations. However, when ONL thickness was compared in eight-month-old congenic Tnfaip3(+/-) and Tnfaip3(+/+) mice, no differences were found, suggesting that Tnfaip3 is not the quantitative trait gene (QTG) for the Chr 10 QTL. The GO analysis revealed that GO terms associated with stress and cell remodeling are overrepresented in the ageRD-sensitive C strain compared with the B6a strain with age (eight months). In the ageRD-resistant B6a strain, compared with the C strain, GO terms associated with antioxidant response and the regulation of blood vessel size are overrepresented with age. CONCLUSIONS: The analyses of differentially expressed genes and the PosMed database yielded candidate genes for the Chr 6 and Chr 10 QTL. HtrA serine peptidase 2 (Htra2), Dok1, and Tnfaip3 were deemed most promising because of their known roles in apoptosis and our finding of nonsynonymous substitutions between B6a and C strains. While Tnfaip3 was excluded as the QTG for the Chr 10 QTL, Dok1 and Htra2 remain good candidates for the Chr 6 QTL. Finally, the GO term analysis further supports the general hypothesis that oxidative stress is involved in ageRD.


Assuntos
Mapeamento Cromossômico , Estudos de Associação Genética , Locos de Características Quantitativas , Degeneração Retiniana/genética , Animais , Cisteína Endopeptidases/genética , Proteínas de Ligação a DNA/genética , Feminino , Serina Peptidase 2 de Requerimento de Alta Temperatura A , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Análise em Microsséries , Proteínas Mitocondriais/genética , Estresse Oxidativo , Fenótipo , Fosfoproteínas/genética , Polimorfismo de Nucleotídeo Único , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina Endopeptidases/genética , Proteína 3 Induzida por Fator de Necrose Tumoral alfa
14.
Proc Natl Acad Sci U S A ; 107(13): 5961-6, 2010 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-20231467

RESUMO

The P23H mutation within the rhodopsin gene (RHO) causes rhodopsin misfolding, endoplasmic reticulum (ER) stress, and activates the unfolded protein response (UPR), leading to rod photoreceptor degeneration and autosomal dominant retinitis pigmentosa (ADRP). Grp78/BiP is an ER-localized chaperone that is induced by UPR signaling in response to ER stress. We have previously demonstrated that BiP mRNA levels are selectively reduced in animal models of ADRP arising from P23H rhodopsin expression at ages that precede photoreceptor degeneration. We have now overexpressed BiP to test the hypothesis that this chaperone promotes the trafficking of P23H rhodopsin to the cell membrane, reprograms the UPR favoring the survival of photoreceptors, blocks apoptosis, and, ultimately, preserves vision in ADRP rats. In cell culture, increasing levels of BiP had no impact on the localization of P23H rhodopsin. However, BiP overexpression alleviated ER stress by reducing levels of cleaved pATF6 protein, phosphorylated eIF2alpha and the proapoptotic protein CHOP. In P23H rats, photoreceptor levels of cleaved ATF6, pEIF2alpha, CHOP, and caspase-7 were much higher than those of wild-type rats. Subretinal delivery of AAV5 expressing BiP to transgenic rats led to reduction in CHOP and photoreceptor apoptosis and to a sustained increase in electroretinogram amplitudes. We detected complexes between BiP, caspase-12, and the BH3-only protein BiK that may contribute to the antiapoptotic activity of BiP. Thus, the preservation of photoreceptor function resulting from elevated levels of BiP is due to suppression of apoptosis rather than to a promotion of rhodopsin folding.


Assuntos
Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Rodopsina/genética , Rodopsina/metabolismo , Visão Ocular/genética , Visão Ocular/fisiologia , Substituição de Aminoácidos , Animais , Apoptose , Sequência de Bases , Dependovirus/genética , Modelos Animais de Doenças , Eletrorretinografia , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Células HeLa , Humanos , Camundongos , Modelos Biológicos , Complexos Multiproteicos , Mutação de Sentido Incorreto , Células Fotorreceptoras de Vertebrados/patologia , Células Fotorreceptoras de Vertebrados/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Retina/patologia , Retina/fisiopatologia , Retinose Pigmentar/genética , Retinose Pigmentar/patologia , Retinose Pigmentar/fisiopatologia , Retinose Pigmentar/terapia , Rodopsina/química , Estresse Fisiológico , Fator de Transcrição CHOP/metabolismo , Transfecção , Resposta a Proteínas não Dobradas/genética , Resposta a Proteínas não Dobradas/fisiologia
15.
J Comp Neurol ; 511(6): 724-35, 2008 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-18925574

RESUMO

Several neurotrophic factors (NTFs) are effective in protecting retinal photoreceptor cells from the damaging effects of constant light and slowing the rate of inherited photoreceptor degenerations. It is currently unclear whether, if continuously available, all NTFs can be protective for many or most retinal degenerations (RDs). We used transgenic mice that continuously overexpress the neurotrophin NT-3 from lens fibers under the control of the alphaA-crystallin promoter to test for neuroprotection in light-damage experiments and in four naturally occurring or transgenically induced RDs in mice. Lens-specific expression of NT-3 mRNA was demonstrated both by in situ hybridization in embryos and by reverse-transcriptase polymerase chain reaction (RT-PCR) in adult mice. Furthermore, NT-3 protein was found in abundance in the lens, ocular fluids, and retina by enzyme-linked immunosorbent assay (ELISA) and immunocytochemistry. Overexpression of NT-3 had no adverse effects on the structure or function of the retina for up to at least 14 months of age. Mice expressing the NT-3 transgene were protected from the damaging effects of constant light to a much greater degree than those receiving bolus injections of NT-3. When the NT-3 transgene was transferred into rd/rd, Rds/+, Q344ter mutant rhodopsin or Mertk knockout mice, overexpression of NT-3 had no protective effect on the RDs in these mice. Thus, specificity of the neuroprotective effect of NT-3 is clearly demonstrated, and different molecular mechanisms are inferred to mediate the protective effect in light-induced and inherited RDs.


Assuntos
Citoproteção/genética , Células Epiteliais/metabolismo , Cristalino/metabolismo , Neurotrofina 3/genética , Neurotrofina 3/metabolismo , Degeneração Retiniana/terapia , Animais , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Cristalino/fisiopatologia , Cristalino/efeitos da radiação , Luz/efeitos adversos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Células Fotorreceptoras de Vertebrados/efeitos da radiação , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/genética , Degeneração Retiniana/etiologia , Degeneração Retiniana/fisiopatologia , Fatores de Tempo , Transgenes/genética , c-Mer Tirosina Quinase
16.
Invest Ophthalmol Vis Sci ; 48(12): 5756-66, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18055829

RESUMO

PURPOSE: CNTF is a neuroprotective agent for retinal degenerations that can cause reduced electroretinogram (ERG) amplitudes. The goal of the present study was to determine the effects of intraocular delivery of CNTF on normal rat visual function. METHODS: Full-field scotopic and photopic ERG amplitudes and spatial frequency thresholds of the optokinetic response (OKR) of adult Long-Evans rats were measured before and after intravitreous injection of CNTF or subretinal delivery of adenoassociated virus-vectored CNTF (AAV-CNTF) into one eye. Visual acuity was also measured by using the Visual Water Task in AAV-CNTF-injected animals. Multiunit luminance thresholds were recorded in the superior colliculus after CNTF injection, and the eyes were examined histologically. RESULTS: In eyes injected with a high dose of CNTF, ERG amplitudes and OKR thresholds measured through CNTF-injected eyes were decreased by 45% to 70% within 6 days after injection. ERG amplitudes had begun to recover by 21 days, whereas OKR thresholds only began to recover after 56 days. Neither OKR thresholds nor ERG amplitudes fully recovered until 90 to 100 days. When measured in the superior colliculus at 2 weeks after CNTF injection, luminance thresholds were elevated by 0.35 log units. In AAV-CNTF-injected eyes, OKR thresholds, and visual acuity were reduced by approximately 50% for at least 6 months, and scotopic and photopic ERG b-waves were reduced by 30% to 50%. Photoreceptor loss occurred in the injected regions in some of the eyes. By contrast, comparison of dose-response analysis with a dose-response study of light damage strongly suggests that therapeutic doses of CNTF exist that do not suppress ERG responses. CONCLUSIONS: Intraocular delivery of CNTF, which preserves photoreceptors in animal models of retinal degeneration, impairs visual function in normal rats at very high doses, but not at lower doses that still provide protection from constant light damage.


Assuntos
Fator Neurotrófico Ciliar/toxicidade , Eletrorretinografia/efeitos dos fármacos , Nistagmo Optocinético/efeitos dos fármacos , Transtornos da Visão/induzido quimicamente , Acuidade Visual/efeitos dos fármacos , Animais , Fator Neurotrófico Ciliar/genética , Dependovirus/genética , Relação Dose-Resposta a Droga , Vetores Genéticos , Injeções , Luz/efeitos adversos , Células Fotorreceptoras de Vertebrados/efeitos da radiação , Lesões Experimentais por Radiação/etiologia , Lesões Experimentais por Radiação/prevenção & controle , Ratos , Ratos Long-Evans , Limiar Sensorial/efeitos dos fármacos , Colículos Superiores/efeitos dos fármacos , Corpo Vítreo
17.
Hum Gene Ther ; 18(10): 871-80, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17892416

RESUMO

The selective silencing of target genes in specific cell types by RNA interference (RNAi) represents a powerful approach both to gene therapy of dominantly active mutant alleles, and to the investigation of normal gene function in animal models in vivo. We established a simple and versatile in vitro method for screening the efficacy of DNA-based short hairpin RNAs (shRNAs), and identified a highly effective shRNA targeting basic fibroblast growth factor (bFGF), a gene thought to play important roles in endogenous neuroprotective responses in the rat retina. We used two viral vectors, based on lentivirus and adeno-associated virus (AAV), to deliver shRNAs and silence bFGF in retinal pigment epithelial cells in vivo. The AAV experiments made use of a "stabilized double-stranded" version of these vectors with rapid onset of gene expression. In the rat retinal pigment epithelium, shRNAs delivered by either vector reduced bFGF immunoreactivity to undetectable levels in transduced cells, whereas a nonfunctional control construct incorporating a two-base pair mutation had no measurable effect on bFGF expression. Silencing commenced within a few days after injection of virus and remained stable throughout the period of observation, as long as 60 days. Viral delivery of RNAi constructs offers a powerful and versatile approach for both gene therapy and the analysis of fundamental questions in retinal biology.


Assuntos
Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Terapia Genética , Vetores Genéticos , Epitélio Pigmentado Ocular/metabolismo , RNA Interferente Pequeno/metabolismo , Animais , Linhagem Celular , Dependovirus/genética , Marcação de Genes , Lentivirus/genética , Epitélio Pigmentado Ocular/citologia , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos
18.
Invest Ophthalmol Vis Sci ; 48(3): 1389-400, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17325188

RESUMO

PURPOSE: To characterize molecular and cellular changes induced by sustained expression of ciliary neurotrophic factor (CNTF) in the rds mutant mouse retina. METHODS: Recombinant adeno-associated virus (rAAV) expressing CNTF was injected subretinally, for transduction of peripherin/rds(+/)(-) transgenic mice that carry the P216L mutation found in human retinitis pigmentosa. Characterization of retinal neurons and glia was performed by immunocytochemistry with cell-type-specific markers. Activation of signaling molecules was examined by Western blot and immunostaining. Alterations of gene transcription profiles were studied by microarray analyses. RESULTS: CNTF viral transduction maintained rhodopsin expression in surviving rod photoreceptors, but greatly reduced both S- and M-opsin normally expressed in cones. In addition, CNTF treatment resulted in increased numbers and dispersion of Müller glia and Chx10-positive bipolar cells within the inner nuclear layer. Persistent CNTF signaling also caused enhanced phosphorylation of STAT1, STAT3, and p42/44 ERK, as well as their levels of expression. Moreover, altered transcription profiles were detected for a large number of genes. Among these, Crx and Nrl involved in photoreceptor differentiation and several genes involved in phototransduction were suppressed. CONCLUSIONS: Despite the rescue from cell death, continuous exposure to CNTF changed photoreceptor cell profiles, especially resulting in the loss of cone immunoreactivity. In addition, the Müller glia and bipolar cells became disorganized, and the number of cells expressing Müller and bipolar cell markers increased. Constitutive CNTF production resulted in sustained activation of cytokine signal transduction and altered the expression of a large number of genes. Therefore, stringent regulation of CNTF may be necessary for its therapeutic application in preventing retinal degeneration.


Assuntos
Fator Neurotrófico Ciliar/genética , Regulação da Expressão Gênica/fisiologia , Neuroglia/patologia , Células Fotorreceptoras de Vertebrados/metabolismo , Retinose Pigmentar/metabolismo , Animais , Western Blotting , Dependovirus/genética , Modelos Animais de Doenças , Eletrorretinografia , Vetores Genéticos , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neuroglia/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Células Fotorreceptoras de Vertebrados/patologia , Células Bipolares da Retina/metabolismo , Células Bipolares da Retina/patologia , Retinose Pigmentar/patologia , Rodopsina/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Transfecção
19.
Novartis Found Symp ; 255: 179-88; discussion 188-94, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14750604

RESUMO

Viable strategies for retinal gene therapy must be designed to cope with the genetic nature of the disease and/or the primary pathologic process responsible for retinal malfunction. For dominant gene defects the aim must be to destroy the presumably toxic gene product, for recessive gene defects the direct approach aims to provide a wild-type copy of the gene to the affected retinal cell type, and for diseases of either complex or unknown genetic origin, more general cell survival strategies that deal with preserving affected retinal cells are often the best and only option. Hence examples of each type of therapy will be briefly discussed in several animal models, including ribozyme therapy for autosomal dominant retinitis pigmentosa in the transgenic P23H opsin rat, beta-PDE gene augmentation therapy for autosomal recessive retinitis pigmentosa in the rd mouse, glial cell-derived neurotrophic factor (GDNF) gene therapy for autosomal dominant RP in the transgenic S334ter opsin rat and pigment epithelial cell-derived neurotrophic factor (PEDF) gene therapy for neovascular retinal disease in rodents. Each employs a recombinant AAV vectored passenger gene controlled by one of several promoters supporting either photoreceptor-specific expression or more general retinal cell expression depending on the therapeutic requirements.


Assuntos
Dependovirus , Proteínas do Olho , Terapia Genética , Vetores Genéticos , Doenças Retinianas/terapia , Animais , Genes Dominantes , Genes Recessivos , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Camundongos , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA Catalítico , Doenças Retinianas/genética , Neovascularização Retiniana/terapia , Serpinas/genética , Serpinas/metabolismo
20.
J Neurosci ; 23(14): 6050-7, 2003 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-12853423

RESUMO

Phenyl-N-tert-butylnitrone (PBN) protects rat retinas against light damage. Because the degenerative process involved in light damage and inherited retinal degeneration both lead to a common final cell death, apoptosis, we used transgenic rats with a P23H or S334ter rhodopsin mutation to test the effects of PBN on retinal degeneration and light damage and the susceptibility of the transgenic rats to light damage. In the first study, 3-week-old mutant and wild-type rats were given no drug, 0.25% PBN in drinking water, or 0.25% PBN in drinking water plus three daily intraperitoneal injections of PBN (100 mg/kg, i.p., every 8 hr). Electroretinograms were recorded at postnatal day 49, after which the rats were killed for morphometric analysis. There was no photoreceptor rescue by PBN in P23H or S334ter rats, as evidenced by equivalent loss of function and photoreceptor cells in the three treatment groups. In the second study, P23H, S334ter, and wild-type rats were exposed for 24 hr to 2700 lux light. The rats were untreated or treated with PBN (50 mg/kg per injection, every 6 hr, starting before exposure). ERGs were recorded before and 1 d after exposure. Animals were killed 6 d later for morphometric analysis. PBN protected wild-type and P23H but not S334ter retinas from light damage. S334ter retinas were relatively less susceptible to light damage than P23H and wild-type rats. The results suggest that the initiating event(s) that causes photoreceptor cell death in the mutated rats is different from that which occurs in light damage, although both ultimately undergo an apoptotic cell death.


Assuntos
Sequestradores de Radicais Livres/farmacologia , Luz/efeitos adversos , Óxidos de Nitrogênio/uso terapêutico , Degeneração Retiniana/tratamento farmacológico , Rodopsina/genética , Substituição de Aminoácidos , Animais , Animais Geneticamente Modificados , Óxidos N-Cíclicos , Modelos Animais de Doenças , Eletrorretinografia , Predisposição Genética para Doença , Mutação , Estimulação Luminosa , Fotoperíodo , Ratos , Ratos Sprague-Dawley , Retina/efeitos dos fármacos , Retina/patologia , Retina/efeitos da radiação , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Rodopsina/metabolismo , Limiar Sensorial/efeitos da radiação , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA