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.
Doc Ophthalmol ; 147(1): 29-43, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37106219

RESUMO

PURPOSE: The uniform field electroretinogram (UF-ERG) has been suggested as an alternative to the pattern electroretinogram (PERG) for non-invasive assessment of retinal ganglion cell (RGC) function in primates. We evaluated the validity of the UF-ERG to assess mouse RGC activity in vivo. METHODS: Unilateral optic nerve crush (ONC) was performed on adult C57BL/6J mice. Contralateral eyes with uncrushed optic nerves and eyes from surgically naive mice served as experimental controls. Electrophysiological visual assessment was performed at 12 weeks post-ONC. Flash-mediated visual-evoked cortical potentials (VEPs) were measured to confirm the robustness of the ONC procedure. Full-field flash ERGs were used to interrogate photoreceptor and retinal bipolar cell function. RGC function was assessed with pattern ERGs. Summed onset and offset UF-ERG responses to alternating dark and light uniform field flash stimuli of different intensities and wavelengths were recorded from ONC and control eyes, and relative differences were compared to the PERG results. Following electrophysiological analysis, RGC loss was monitored by immunohistochemical staining of the RGC marker protein, RBPMS, in post-mortem retinal tissues. RESULTS: ONC dramatically impacts RGC integrity and optic nerve function, demonstrated by reduced RGC counts and near complete elimination of VEPs. ONC did not affect scotopic ERG a-wave and b-wave amplitudes, while PERG amplitudes of eyes subjected to ONC were reduced by approximately 50% compared to controls. Summation of ON and OFF UF-ERG responses did not reveal statistically significant differences between ONC and control eyes, regardless of visual stimulus. CONCLUSIONS: PERG responses are markedly impaired upon ONC, while UF-ERG responses are not significantly affected by surgical trauma to RGC axons in mice. The more closely related pattern and uniform field ERGs recorded in primates suggests species-specific differences in RGC features or subpopulations corresponding to PERG and UF-ERG response generators, limiting the utility of the UF-ERG for mouse RGC functional analysis.


Assuntos
Eletrorretinografia , Células Ganglionares da Retina , Camundongos , Animais , Células Ganglionares da Retina/fisiologia , Eletrorretinografia/métodos , Camundongos Endogâmicos C57BL , Retina , Nervo Óptico , Modelos Animais de Doenças
2.
J Clin Invest ; 132(22)2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36136598

RESUMO

Preterm birth is the leading cause of death in children under 5 years of age. Premature infants who receive life-saving oxygen therapy often develop bronchopulmonary dysplasia (BPD), a chronic lung disease. Infants with BPD are at a high risk of abnormal neurodevelopment, including motor and cognitive difficulties. While neural progenitor cells (NPCs) are crucial for proper brain development, it is unclear whether they play a role in BPD-associated neurodevelopmental deficits. Here, we show that hyperoxia-induced experimental BPD in newborn mice led to lifelong impairments in cerebrovascular structure and function as well as impairments in NPC self-renewal and neurogenesis. A neurosphere assay utilizing nonhuman primate preterm baboon NPCs confirmed impairment in NPC function. Moreover, gene expression profiling revealed that genes involved in cell proliferation, angiogenesis, vascular autoregulation, neuronal formation, and neurotransmission were dysregulated following neonatal hyperoxia. These impairments were associated with motor and cognitive decline in aging hyperoxia-exposed mice, reminiscent of deficits observed in patients with BPD. Together, our findings establish a relationship between BPD and abnormal neurodevelopmental outcomes and identify molecular and cellular players of neonatal brain injury that persist throughout adulthood that may be targeted for early intervention to aid this vulnerable patient population.


Assuntos
Displasia Broncopulmonar , Disfunção Cognitiva , Hiperóxia , Nascimento Prematuro , Recém-Nascido , Feminino , Camundongos , Humanos , Animais , Hiperóxia/complicações , Hiperóxia/metabolismo , Animais Recém-Nascidos , Displasia Broncopulmonar/genética , Neurogênese , Disfunção Cognitiva/etiologia , Cognição , Pulmão/metabolismo
3.
Gene Ther ; 29(3-4): 147-156, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34363035

RESUMO

Glaucoma is a prevalent neurodegenerative disease that is characterized by progressive visual field loss. It is the leading cause of irreversible blindness in the world. The main risk factor for glaucoma is elevated intraocular pressure that results in the damage and death of retinal ganglion cells (RGCs) and their axons. The death of RGCs has been shown to be apoptotic. We tested the hypothesis that blocking the activation of apoptosis may be an effective strategy to prevent RGC death and preserve functional vision in glaucoma. In the magnetic microbead mouse model of induced ocular hypertension, inhibition of RGC apoptosis was targeted through viral-mediated ocular delivery of the X-linked inhibitor of apoptosis (XIAP) gene, a potent caspase inhibitor. Pattern electroretinograms revealed that XIAP therapy resulted in significant protection of both somal and axonal RGC function in glaucomatous eyes. Histology confirmed that the treated optic nerves showed preservation of axon counts and reduced glial cell infiltration. These results show that XIAP is able to provide both functional and structural protection of RGCs in the microbead model of glaucoma and provide important proof-of-principle for XIAP's efficacy as a neuroprotective treatment for glaucoma.


Assuntos
Glaucoma , Doenças Neurodegenerativas , Animais , Axônios , Modelos Animais de Doenças , Terapia Genética , Glaucoma/genética , Glaucoma/terapia , Pressão Intraocular , Camundongos , Células Ganglionares da Retina/metabolismo
4.
Neuroscience ; 452: 169-180, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33197500

RESUMO

Mutation of the α-thalassemia/mental retardation syndrome X-linked protein, ATRX, causes intellectual disability and is associated with pleiotropic defects including ophthalmological abnormalities. We have previously demonstrated that Atrx deficiency in the mouse retina leads to the selective loss of inhibitory interneurons and inner retinal dysfunction. Onset of the amacrine cell neurodegenerative phenotype in Atrx-deficient retinas occurs postnatally after neuronal specification, and coincides with eye opening. Given this timing, we sought to interrogate the influence of light-dependent visual signaling on Atrx-mediated neuronal survival and function in the mouse retina. Retina-specific Atrx conditional knockout (cKO) mice were subjected to light deprivation using two different paradigms: (1) a dark-rearing regime, and (2) genetic deficiency of metabotropic glutamate receptor 6 (mGluR6) to block the ON retinal signaling pathway. Scotopic electroretinography was performed for adult dark-reared Atrx cKO mice and controls to measure retinal neuron function in vivo. Retinal immunohistochemistry and enumeration of amacrine cells were performed for both light deprivation paradigms. We observed milder normalized a-wave, b-wave and oscillatory potential (OP) deficits in electroretinograms of dark-reared Atrx cKO mice compared to light-exposed counterparts. In addition, amacrine cell loss was partially limited by genetic restriction of retinal signaling through the ON pathway. Our results suggest that the temporal features of the Atrx cKO phenotype are likely due to a combined effect of light exposure upon eye opening and coincident developmental processes impacting the retinal circuitry. In addition, this study reveals a novel activity-dependent role for Atrx in mediating post-replicative neuronal integrity in the CNS.


Assuntos
Deficiência Intelectual Ligada ao Cromossomo X , Proteína Nuclear Ligada ao X , Talassemia alfa , Animais , Camundongos , Camundongos Endogâmicos C57BL , Retina , Proteína Nuclear Ligada ao X/genética
5.
Hum Gene Ther ; 28(6): 482-492, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28335619

RESUMO

Retinal detachment is an acute disorder in humans that is caused by trauma or disease, and it can often lead to permanent visual deficits that result from the death of photoreceptors in the retina. The final pathway for photoreceptor cell death is apoptosis and necroptosis. The X-linked inhibitor of apoptosis (XIAP) has been shown to block both of these cell death pathways. This study tested the effects of XIAP on photoreceptor survival in a feline model of retinal detachment. The study was performed in 12 cats, divided into two experimental groups. Six animals received a subretinal injection of adeno-associated virus (AAV) carrying XIAP, and six animals received AAV carrying green fluorescent protein (GFP) as a control. Three weeks after viral delivery, retinas were detached by injecting C3F8 gas into the subretinal space. Optical coherence tomography revealed that the retinal detachments resolved within 3-6 weeks as the gas was slowly resorbed. Analysis of histological sections through the plane of the detachment showed significant preservation of the photoreceptor layer in AAV-XIAP-treated animals compared to AAV-GFP-treated animals at 9 weeks after the detachment. XIAP-treated detached retinas were similar to intact controls. These studies support the potential for XIAP therapy in the treatment of human retinal detachment.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Descolamento Retiniano/terapia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Animais , Apoptose/genética , Gatos , Linhagem Celular , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Fluorocarbonos/administração & dosagem , Expressão Gênica , Genes Reporter , Vetores Genéticos/administração & dosagem , Vetores Genéticos/química , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Injeções Intraoculares , Células Fotorreceptoras Retinianas Cones/patologia , Descolamento Retiniano/genética , Descolamento Retiniano/metabolismo , Descolamento Retiniano/patologia , Transdução de Sinais , Tomografia de Coerência Óptica , Transgenes , Resultado do Tratamento , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
6.
Hum Mol Genet ; 25(21): 4787-4803, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28173139

RESUMO

ATRX is a chromatin remodeling protein that is mutated in several intellectual disability disorders including alpha-thalassemia/mental retardation, X-linked (ATR-X) syndrome. We previously reported the prevalence of ophthalmological defects in ATR-X syndrome patients, and accordingly we find morphological and functional visual abnormalities in a mouse model harboring a mutation occurring in ATR-X patients. The visual system abnormalities observed in these mice parallels the Atrx-null retinal phenotype characterized by interneuron defects and selective loss of amacrine and horizontal cells. The mechanisms that underlie selective neuronal vulnerability and neurodegeneration in the central nervous system upon Atrx mutation or deletion are unknown. To interrogate the cellular specificity of Atrx for its retinal neuroprotective functions, we employed a combination of temporal and lineage-restricted conditional ablation strategies to generate five different conditional knockout mouse models, and subsequently identified a non-cell-autonomous requirement for Atrx in bipolar cells for inhibitory interneuron survival in the retina. Atrx-deficient retinal bipolar cells exhibit functional, structural and molecular alterations consistent with impairments in neuronal activity and connectivity. Gene expression changes in the Atrx-null retina indicate defective synaptic structure and neuronal circuitry, suggest excitotoxic mechanisms of neurodegeneration, and demonstrate that common targets of ATRX in the forebrain and retina may contribute to similar neuropathological processes underlying cognitive impairment and visual dysfunction in ATR-X syndrome.


Assuntos
Deficiência Intelectual Ligada ao Cromossomo X/genética , Proteína Nuclear Ligada ao X/genética , Talassemia alfa/genética , Animais , Cromatina , Modelos Animais de Doenças , Interneurônios/metabolismo , Masculino , Deficiência Intelectual Ligada ao Cromossomo X/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação , Neurônios/metabolismo , Proteínas Nucleares/genética , Retina/metabolismo , Células Bipolares da Retina/metabolismo , Proteína Nuclear Ligada ao X/metabolismo , Talassemia alfa/metabolismo
7.
Acta Biomater ; 9(8): 7855-64, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23623991

RESUMO

Chitosan microparticles (CMPs) have previously been developed for topical applications to the eye, but their safety and efficacy in delivering proteins to the retina have not been adequately evaluated. This study examines the release kinetics of CMPs in vitro, and assesses their biocompatibility and cytotoxicity on retinal cells in vitro and in vivo. Two proteins were used in the encapsulation and release studies: BSA (bovine serum albumin) and tat-EGFP (enhanced green fluorescent protein fused to the transactivator of transcription peptide). Not surprisingly, the in vitro release kinetics were dependent on the protein encapsulated, with BSA showing higher release than tat-EGFP. CMPs containing encapsulated tat-EGFP were tested for cellular toxicity in photoreceptor-derived 661W cells. They showed no signs of in vitro cell toxicity at a low concentration (up to 1mgml(-1)), but at a higher concentration of 10mgml(-1) they were associated with cytotoxic effects. In vivo, CMPs injected into the subretinal space were found beneath the photoreceptor layer of the retina, and persisted for at least 8weeks. Similar to the in vitro studies, the lower concentration of CMPs was generally well tolerated, but the higher concentration resulted in cytotoxic effects and in reduced retinal function, as assessed by electroretinogram amplitudes. Overall, this study suggests that CMPs are effective long-term delivery agents to the retina, but the concentration of chitosan may affect cytotoxicity.


Assuntos
Cápsulas/síntese química , Quitosana/química , Proteínas/administração & dosagem , Proteínas/farmacocinética , Retina/metabolismo , Animais , Cápsulas/administração & dosagem , Células Cultivadas , Difusão , Injeções Intraoculares , Teste de Materiais , Taxa de Depuração Metabólica , Camundongos , Ratos Long-Evans , Retina/efeitos dos fármacos
8.
Dev Neurobiol ; 71(2): 153-69, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20862739

RESUMO

The eye is an excellent model for the study of neuronal development and pathogenesis of central nervous system disorders because of its relative ease of accessibility and the well-characterized cellular makeup. We have used this model to study spinal muscular atrophy (SMA), an autosomal recessive neuromuscular disease caused by deletions or mutations in the survival of motor neuron 1 gene (SMN1). We have investigated the expression pattern of mouse Smn mRNA and protein in the neural retina and the optic nerve of wild type mice. Smn protein is present in retinal ganglion cells and amacrine cells within the neural retina as well as in glial cells in the optic nerve. Histopathological analysis in phenotype stage SMA mice revealed that Smn deficiency is associated with a reduction in ganglion cell axon and glial cell number in the optic nerve, as well as compromised cellular processes and altered organization of neurofilaments in the neural retina. Whole mount preparation and retinal neuron primary culture provided further evidence of abnormal synaptogenesis and neurofilament accumulation in the neurites of Smn-deficient retinal neurons. A subset of amacrine cells is absent, in a cell-autonomous fashion, in the retina of SMA mice. Finally, the retinas of SMA mice have altered electroretinograms. Altogether, our study has demonstrated defects in axodendritic outgrowth and cellular composition in Smn-depleted retinal neurons, indicating a role for Smn in neuritogenesis and neurogenesis, and providing us with an insight into pathogenesis of SMA.


Assuntos
Atrofia Muscular Espinal/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , Retina/metabolismo , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Animais , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Eletrorretinografia , Expressão Gênica , Perfilação da Expressão Gênica , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patologia , Neurônios/citologia , Nervo Óptico/citologia , Nervo Óptico/metabolismo , Retina/citologia , Retina/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína 1 de Sobrevivência do Neurônio Motor/genética
9.
Biomaterials ; 31(12): 3414-21, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20149443

RESUMO

The efficient and controlled delivery of genes and proteins to retinal cells remains a challenge. In this study, we evaluated polyethylene glycol-polylactic acid (PEG-PLA) microparticles for encapsulation and delivery of a Transactivator of transcription-enhanced green fluorescent protein fusion (Tat-EGFP) to retinal cells. Our main objective was to develop a microparticle system that delivers Tat-EGFP with an initial rapid release (within 24 h) followed by a sustained release. We prepared four different formulations of Tat-EGFP encapsulated PEG-PLA particles to investigate the effects of protein and polymer concentrations on particle morphology and protein release, using scanning electron microscopy (SEM) and fluorometry techniques. The optimum formulation was selected based on higher protein release, and smaller particle size. The optimum formulation was then tested in vitro for cell biocompatibility and protein internalization, and in vivo for cellular toxicity following sub-retinal injections into rat eyes. The results suggest that PEG-PLA microparticles can deliver proteins in cell culture allowing protein internalization in as little as 1 h. In vivo, protein was shown to localize within the photoreceptor layer of the retina, and persist for at least 9 weeks with no observed toxicity.


Assuntos
Portadores de Fármacos , Produtos do Gene tat/administração & dosagem , Proteínas de Fluorescência Verde/administração & dosagem , Ácido Láctico/química , Polietilenoglicóis/química , Polímeros/química , Retina/metabolismo , Animais , Células Cultivadas , Fluorometria , Microscopia Eletrônica de Varredura , Microesferas , Poliésteres , Ratos
10.
Invest Ophthalmol Vis Sci ; 50(3): 1448-53, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19060276

RESUMO

PURPOSE: To evaluate the ability of X-linked inhibitor of apoptosis (XIAP) gene therapy to provide neuroprotection to cells of the outer nuclear layer (ONL) of the retina after retinal detachment. METHODS: Subretinal injections of a recombinant adenoassociated virus (rAAV) encoding either XIAP or green fluorescent protein (GFP; injection control) were performed in the left eye of Brown Norway rats. Two weeks later, retinal detachments were created at the site of viral injection by delivering sodium hyaluronate into the subretinal space. Retinal tissue was harvested at 24 hours after retinal detachment and was analyzed for caspase 3 and 9 activity. Histologic analysis was conducted on samples taken at 3 days and 2 months after detachment to confirm the presence of XIAP or GFP expression and to assess levels of apoptosis and changes in retinal thickness. RESULTS: Caspase assays performed 24 hours after detachment confirmed an expected increase in caspase 3 and 9 activity in the detached regions of GFP-treated retinas, whereas XIAP-treated detached retinas behaved comparably to attached controls. TUNEL analysis of 3-day tissue samples showed fewer apoptotic cells in XIAP-treated detachments than in GFP-treated detachments. At 2 months after the detachment, histology and immunohistochemistry confirmed the preservation of the ONL at sites of XIAP overexpression, whereas the GFP-treated detached retinas had significantly deteriorated. CONCLUSIONS: The results suggest that XIAP confers structural neuroprotection of photoreceptors for at least 2 months after retinal detachment.


Assuntos
Apoptose/efeitos dos fármacos , Dependovirus/genética , Terapia Genética , Células Fotorreceptoras de Vertebrados/patologia , Descolamento Retiniano/terapia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Animais , Caspase 3/metabolismo , Caspase 9/metabolismo , Modelos Animais de Doenças , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Marcação In Situ das Extremidades Cortadas , Masculino , Fármacos Neuroprotetores , Células Fotorreceptoras de Vertebrados/enzimologia , Ratos , Ratos Endogâmicos BN , Descolamento Retiniano/enzimologia , Transfecção
11.
PLoS One ; 2(3): e314, 2007 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-17375200

RESUMO

BACKGROUND: Retinitis pigmentosa (RP) is a blinding genetic disorder that is caused by the death of photoreceptors in the outer nuclear layer of the retina. To date, 39 different genetic loci have been associated with the disease, and 28 mutated genes have been identified. Despite the complexity of the underlying genetic basis for RP, the final common pathway is photoreceptor cell death via apoptosis. METHODOLOGY/PRINCIPAL FINDINGS: In this study, P23H and S334ter rhodopsin transgenic rat models of RP were used to test the neuroprotective effects of anti-apoptotic gene therapy. Adeno-associated viruses (AAV) carrying the X-linked inhibitor of apoptosis (XIAP) or green fluorescent protein (GFP) were delivered subretinally into the eye of transgenic rat pups. Histological and functional measures were used to assess neuroprotection. XIAP is known to block apoptosis by inhibiting the action of caspases-3, -7 and -9. The results show that XIAP gene therapy provides long-term neuroprotection of photoreceptors at both structural and functional levels. CONCLUSIONS/SIGNIFICANCE: Our gene therapy strategy targets the apoptotic cascade, which is the final common pathway in all forms of retinitis pigmentosa. This strategy holds great promise for the treatment of RP, as it allows for the broad protection of photoreceptors, regardless of the initial disease causing mutation.


Assuntos
Células Fotorreceptoras de Vertebrados/fisiologia , Retinose Pigmentar/prevenção & controle , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/fisiologia , Animais , Animais Geneticamente Modificados , Apoptose , Inibidores de Caspase , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/genética , Ratos , Ratos Long-Evans , Degeneração Retiniana/prevenção & controle , Cromossomo X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...