Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Doc Ophthalmol ; 147(1): 29-43, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37106219

RESUMEN

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.


Asunto(s)
Electrorretinografía , Células Ganglionares de la Retina , Ratones , Animales , Células Ganglionares de la Retina/fisiología , Electrorretinografía/métodos , Ratones Endogámicos C57BL , Retina , Nervio Óptico , Modelos Animales de Enfermedad
2.
Gene Ther ; 29(3-4): 147-156, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34363035

RESUMEN

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.


Asunto(s)
Glaucoma , Enfermedades Neurodegenerativas , Animales , Axones , Modelos Animales de Enfermedad , Terapia Genética , Glaucoma/genética , Glaucoma/terapia , Presión Intraocular , Ratones , Células Ganglionares de la Retina/metabolismo
3.
Hum Mol Genet ; 25(21): 4787-4803, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28173139

RESUMEN

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.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X/genética , Proteína Nuclear Ligada al Cromosoma X/genética , Talasemia alfa/genética , Animales , Cromatina , Modelos Animales de Enfermedad , Interneuronas/metabolismo , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutación , Neuronas/metabolismo , Proteínas Nucleares/genética , Retina/metabolismo , Células Bipolares de la Retina/metabolismo , Proteína Nuclear Ligada al Cromosoma X/metabolismo , Talasemia alfa/metabolismo
4.
Dev Biol ; 411(1): 85-100, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26795056

RESUMEN

Neurogenesis is regulated by the dynamic and coordinated activity of several extracellular signalling pathways, but the basis for crosstalk between these pathways remains poorly understood. Here we investigated regulatory interactions between two pathways that are each required for neural progenitor cell maintenance in the postnatal retina; Hedgehog (Hh) and Notch signalling. Both pathways are activated in progenitor cells in the postnatal retina based on the co-expression of fluorescent pathway reporter transgenes at the single cell level. Disrupting Notch signalling, genetically or pharmacologically, induces a rapid downregulation of all three Gli proteins and inhibits Hh-induced proliferation. Ectopic Notch activation, while not sufficient to promote Hh signalling or proliferation, increases Gli2 protein. We show that Notch regulation of Gli2 in Müller glia renders these cells competent to proliferate in response to Hh. These data suggest that Notch signalling converges on Gli2 to prime postnatal retinal progenitor cells and Müller glia to proliferate in response to Hh.


Asunto(s)
Células Ependimogliales/citología , Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Células-Madre Neurales/citología , Neurogénesis/fisiología , Receptores Notch/metabolismo , Animales , Proliferación Celular/fisiología , Femenino , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales/metabolismo , Proteínas Nucleares/genética , ARN Mensajero/genética , Receptores Notch/genética , Retina/citología , Retina/embriología , Transducción de Señal , Factores de Transcripción/genética , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc
5.
Hum Mol Genet ; 22(5): 1005-16, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23201751

RESUMEN

Norrie disease (ND) is a congenital disorder characterized by retinal hypovascularization and cognitive delay. ND has been linked to mutations in 'Norrie Disease Protein' (Ndp), which encodes the secreted protein Norrin. Norrin functions as a secreted angiogenic factor, although its role in neural development has not been assessed. Here, we show that Ndp expression is initiated in retinal progenitors in response to Hedgehog (Hh) signaling, which induces Gli2 binding to the Ndp promoter. Using a combination of genetic epistasis and acute RNAi-knockdown approaches, we show that Ndp is required downstream of Hh activation to induce retinal progenitor proliferation in the retina. Strikingly, Ndp regulates the rate of cell-cycle re-entry and not cell-cycle kinetics, thereby uncoupling the self-renewal and cell-cycle progression functions of Hh. Taken together, we have uncovered a cell autonomous function for Ndp in retinal progenitor proliferation that is independent of its function in the retinal vasculature, which could explain the neural defects associated with ND.


Asunto(s)
Ceguera/congénito , Proteínas del Ojo/genética , Factores de Transcripción de Tipo Kruppel/genética , Proteínas del Tejido Nervioso/genética , Enfermedades del Sistema Nervioso/genética , Retina/metabolismo , Espasmos Infantiles/genética , Vía de Señalización Wnt , Animales , Ceguera/genética , Ceguera/metabolismo , Ciclo Celular/genética , Proliferación Celular , Epistasis Genética , Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Enfermedades Genéticas Ligadas al Cromosoma X , Proteínas Hedgehog/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Transgénicos , Mutación , Neovascularización Fisiológica , Proteínas del Tejido Nervioso/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Unión Proteica , Retina/crecimiento & desarrollo , Degeneración Retiniana , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/metabolismo , Espasmos Infantiles/metabolismo , Vía de Señalización Wnt/genética , Proteína Gli2 con Dedos de Zinc
6.
Transl Vis Sci Technol ; 12(11): 24, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37982768

RESUMEN

Purpose: This systematic review evaluates the safety and efficacy of ocular gene therapy using adeno-associated virus (AAV). Methods: MEDLINE, Embase, Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov were searched systematically for controlled or non-controlled interventional gene therapy studies using key words related to retinal diseases, gene therapy, and AAV vectors. The primary outcome measure was safety, based on ocular severe adverse events (SAEs). Secondary outcome measures evaluated efficacy of the therapy based on best corrected visual acuity (BCVA) and improvements in visual sensitivity and systemic involvement following ocular delivery. Pooling was done using a DerSimonian Laird random effects model. Risk of bias was assessed using the Cochrane Risk of Bias Tool, version 1. Results: Our search identified 3548 records. Of these, 80 publications met eligibility criteria, representing 28 registered clinical trials and 5 postmarket surveillance studies involving AAV gene therapy for Leber congenital amaurosis (LCA), choroideremia, Leber hereditary optic neuropathy (LHON), age-related macular degeneration (AMD), retinitis pigmentosa (RP), X-linked retinoschisis, and achromatopsia. Overall, AAV therapy vectors were associated with a cumulative incidence of at least one SAE of 8% (95% confidence intervals [CIs] of 5% to 12%). SAEs were often associated with the surgical procedure rather than the therapeutic vector itself. Poor or inconsistent reporting of adverse events (AEs) were a limitation for the meta-analysis. The proportion of patients with any improvement in BCVA and visual sensitivity was 41% (95% CIs of 31% to 51%) and 51% (95% CIs of 31% to 70%), respectively. Systemic immune involvement was associated with a cumulative incidence of 31% (95% CI = 21% to 42%). Conclusions: AAV gene therapy vectors appear to be safe but the surgical procedure required to deliver them is associated with some risk. The large variability in efficacy can be attributed to the small number of patients treated, the heterogeneity of the population and the variability in dosage, volume, and follow-up. Translational Relevance: This systematic review will help to inform and guide future clinical trials.


Asunto(s)
Degeneración Macular , Degeneración Retiniana , Retinitis Pigmentosa , Humanos , Degeneración Retiniana/terapia , Dependovirus/genética , Degeneración Macular/tratamiento farmacológico , Terapia Genética/efectos adversos
7.
Nat Methods ; 6(2): 127-30, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19122667

RESUMEN

We developed retrograde, transsynaptic pseudorabies viruses (PRVs) with genetically encoded activity sensors that optically report the activity of connected neurons among spatially intermingled neurons in the brain. Next we engineered PRVs to express two differentially colored fluorescent proteins in a time-shifted manner to define a time period early after infection to investigate neural activity. Finally we used multiple-colored PRVs to differentiate and dissect the complex architecture of brain regions.


Asunto(s)
Proteínas Fluorescentes Verdes/análisis , Herpesvirus Suido 1/metabolismo , Proteínas Luminiscentes/análisis , Transmisión Sináptica/fisiología , Vías Visuales/virología , Animales , Técnicas Biosensibles/métodos , Encéfalo/citología , Encéfalo/fisiología , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Herpesvirus Suido 1/genética , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Ratones , Neuronas/fisiología , Neuronas/virología , Factores de Tiempo , Vías Visuales/fisiología , Proteína Fluorescente Roja
8.
Nat Neurosci ; 11(6): 667-75, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18432197

RESUMEN

Genetically encoded optical neuromodulators create an opportunity for circuit-specific intervention in neurological diseases. One of the diseases most amenable to this approach is retinal degeneration, where the loss of photoreceptors leads to complete blindness. To restore photosensitivity, we genetically targeted a light-activated cation channel, channelrhodopsin-2, to second-order neurons, ON bipolar cells, of degenerated retinas in vivo in the Pde6b(rd1) (also known as rd1) mouse model. In the absence of 'classical' photoreceptors, we found that ON bipolar cells that were engineered to be photosensitive induced light-evoked spiking activity in ganglion cells. The rescue of light sensitivity was selective to the ON circuits that would naturally respond to increases in brightness. Despite degeneration of the outer retina, our intervention restored transient responses and center-surround organization of ganglion cells. The resulting signals were relayed to the visual cortex and were sufficient for the animals to successfully perform optomotor behavioral tasks.


Asunto(s)
Luz , Células Bipolares de la Retina/fisiología , Degeneración Retiniana , Rodopsina/fisiología , Visión Ocular/fisiología , Animales , Conducta Animal , Modelos Animales de Enfermedad , Electroporación/métodos , Potenciales Evocados Visuales/efectos de los fármacos , Potenciales Evocados Visuales/fisiología , Potenciales Evocados Visuales/efectos de la radiación , Antagonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/genética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/fisiología , Actividad Motora/efectos de la radiación , Técnicas de Placa-Clamp , Estimulación Luminosa/métodos , Piperazinas/farmacología , Quinoxalinas/farmacología , Células Bipolares de la Retina/efectos de la radiación , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Degeneración Retiniana/terapia , Células Ganglionares de la Retina/fisiología , Factores de Tiempo , Visión Ocular/efectos de la radiación , Vías Visuales/efectos de los fármacos , Vías Visuales/fisiología , Vías Visuales/efectos de la radiación
9.
Neuroscience ; 452: 169-180, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33197500

RESUMEN

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.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X , Proteína Nuclear Ligada al Cromosoma X , Talasemia alfa , Animales , Ratones , Ratones Endogámicos C57BL , Retina , Proteína Nuclear Ligada al Cromosoma X/genética
10.
Invest Ophthalmol Vis Sci ; 61(8): 49, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32735323

RESUMEN

Purpose: Leber hereditary optic neuropathy (LHON) is a genetic form of vision loss that occurs primarily owing to mutations in the nicotinamide adenine dinucleotide dehydrogenase (ND) subunits that make up complex I of the electron transport chain. LHON mutations result in the apoptotic death of retinal ganglion cells. We tested the hypothesis that gene therapy with the X-linked inhibitor of apoptosis (XIAP) would prevent retinal ganglion cell apoptosis and reduce disease progression in a vector-induced mouse model of LHON that carries the ND4 mutation. Methods: Adeno-associated virus (AAV) encoding full length hemagglutinin-tagged XIAP (AAV2.HA-XIAP) or green fluorescent protein (AAV2.GFP) was injected into the vitreous of DBA/1J mice. Two weeks later, the LHON phenotype was induced by AAV delivery of mutant ND4 (AAV2.mND4FLAG) to the vitreous. Retinal function was assessed by pattern electroretinography. Optic nerves were harvested at 4 months, and the effects of XIAP therapy on nerve fiber layer and optic nerve integrity were evaluated using immunohistochemistry, transmission electron microscopy and magnetic resonance imaging. Results: During LHON disease progression, retinal ganglion cell axons are lost. Apoptotic cell bodies are seen in the nuclei of astrocytes or oligodendrocytes in the optic nerve, and there is thinning of the optic nerve and the nerve fiber layer of the retina. At 4 months after disease onset, XIAP gene therapy protects the nerve fiber layer and optic nerve architecture by preserving axon health. XIAP also decreases nuclear fragmentation in resident astrocytes or oligodendrocytes and decreases glial cell infiltration. Conclusions: XIAP therapy improves optic nerve health and delays disease progression in LHON.


Asunto(s)
Terapia Genética/métodos , Atrofia Óptica Hereditaria de Leber , Nervio Óptico , Retina , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Animales , Apoptosis , Modelos Animales de Enfermedad , Electrorretinografía/métodos , Inmunohistoquímica , Imagen por Resonancia Magnética/métodos , Ratones , Atrofia Óptica Hereditaria de Leber/genética , Atrofia Óptica Hereditaria de Leber/metabolismo , Atrofia Óptica Hereditaria de Leber/terapia , Nervio Óptico/diagnóstico por imagen , Nervio Óptico/fisiopatología , Retina/diagnóstico por imagen , Retina/fisiopatología , Células Ganglionares de la Retina/metabolismo , Resultado del Tratamiento
11.
Invest Ophthalmol Vis Sci ; 44(7): 2841-50, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12824221

RESUMEN

PURPOSE: The gene elongation of very long chain fatty acids-4 (ELOVL4) has been shown to underlie phenotypically heterogeneous forms of autosomal dominant macular degeneration. In this study, the extent of evolutionary conservation and the existence and localization of retinal expression of this gene was investigated across a wide variety of species. METHODS: Southern blot analysis of genomic DNA and bioinformatic analysis using the human ELOVL4 cDNA and protein sequences, respectively, were performed to identify species in which ELOVL4 orthologues and/or homologues are present. Retinal RNA and protein extracts derived from different species were assessed by Northern hybridization and immunoblot techniques to assess evolutionary conservation of gene expression. Immunohistochemical analysis of tissue sections prepared from various mammalian retinas was performed to determine the distribution of ELOVL4 and homologous proteins within specific retinal cell layers. RESULTS: The existence of ELOVL4 sequence orthologues and homologues was confirmed by both Southern blot analysis and in silico searches of protein sequence databases. Phylogenetic analysis places ELOVL4 among a large family of known and putative fatty acid elongase proteins. Northern blot analysis revealed the presence of multiple transcripts corresponding to ELOVL4 homologues expressed in the retina of several different mammalian species. Conserved proteins were also detected among retinal extracts of different mammals and were found to localize predominantly to the photoreceptor cell layer within retinal tissue preparations. CONCLUSIONS: The ELOVL4 gene is highly conserved throughout evolution and is expressed in the photoreceptor cells of the retina in a variety of different species, which suggests that it plays a critical role in retinal cell biology.


Asunto(s)
Proteínas del Ojo/genética , Expresión Génica , Proteínas de la Membrana/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Secuencia de Aminoácidos , Animales , Northern Blotting , Southern Blotting , Gatos , Bovinos , Pollos , Secuencia Conservada , Electroforesis en Gel de Poliacrilamida , Evolución Molecular , Humanos , Macaca mulatta , Ratones , Datos de Secuencia Molecular , ARN/aislamiento & purificación , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
12.
EMBO Mol Med ; 6(9): 1175-90, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25092770

RESUMEN

In this report, we describe the development of a modified adeno-associated virus (AAV) capsid and promoter for transduction of retinal ON-bipolar cells. The bipolar cells, which are post-synaptic to the photoreceptors, are important retinal targets for both basic and preclinical research. In particular, a therapeutic strategy under investigation for advanced forms of blindness involves using optogenetic molecules to render ON-bipolar cells light-sensitive. Currently, delivery of adequate levels of gene expression is a limiting step for this approach. The synthetic AAV capsid and promoter described here achieves high level of optogenetic transgene expression in ON-bipolar cells. This evokes high-frequency (~100 Hz) spiking responses in ganglion cells of previously blind, rd1, mice. Our vector is a promising vehicle for further development toward potential clinical use.


Asunto(s)
Dependovirus/genética , Células Bipolares de la Retina/virología , Transducción Genética/métodos , Animales , Vectores Genéticos , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas
13.
Nat Commun ; 5: 4181, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24946904

RESUMEN

Chromatin compaction mediates progenitor to post-mitotic cell transitions and modulates gene expression programs, yet the mechanisms are poorly defined. Snf2h and Snf2l are ATP-dependent chromatin remodelling proteins that assemble, reposition and space nucleosomes, and are robustly expressed in the brain. Here we show that mice conditionally inactivated for Snf2h in neural progenitors have reduced levels of histone H1 and H2A variants that compromise chromatin fluidity and transcriptional programs within the developing cerebellum. Disorganized chromatin limits Purkinje and granule neuron progenitor expansion, resulting in abnormal post-natal foliation, while deregulated transcriptional programs contribute to altered neural maturation, motor dysfunction and death. However, mice survive to young adulthood, in part from Snf2l compensation that restores Engrailed-1 expression. Similarly, Purkinje-specific Snf2h ablation affects chromatin ultrastructure and dendritic arborization, but alters cognitive skills rather than motor control. Our studies reveal that Snf2h controls chromatin organization and histone H1 dynamics for the establishment of gene expression programs underlying cerebellar morphogenesis and neural maturation.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Cerebelo/embriología , Ensamble y Desensamble de Cromatina/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Histonas/metabolismo , Morfogénesis/fisiología , Células-Madre Neurales/fisiología , Análisis de Varianza , Animales , Western Blotting , Bromodesoxiuridina , Inmunoprecipitación de Cromatina , Femenino , Fluorescencia , Galactósidos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Indoles , Masculino , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Microscopía Electrónica de Transmisión , Morfogénesis/genética , Células-Madre Neurales/metabolismo , Células de Purkinje/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Prueba de Desempeño de Rotación con Aceleración Constante , Cloruro de Tolonio
14.
J Ocul Biol Dis Infor ; 4(3): 111-20, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23289056

RESUMEN

Retinal neurons are highly vulnerable to a diverse array of neurotoxic stimuli that leads to their degeneration, which is a major contributor to blindness. This review summarizes the role of epigenetic factors in mediating neuronal homeostasis and survival to protect against cell death and neurodegenerative conditions. Studies in human patients and mouse models implicate numerous chromatin modifications in neuroprotective processes including histone protein acetylation and methylation, DNA methylation, and ATP-dependent nucleosome remodeling. Recent research has begun to uncover specific epigenetic mechanisms invoked by neurotoxic stimuli. Continued investigation in this area will be the key to the generation of therapeutic strategies for the intervention of retinal neurodegenerative diseases.

16.
Genomics ; 83(4): 615-25, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15028284

RESUMEN

Elongation of very long chain fatty acids 4 (ELOVL4) is a novel member of the ELO family of genes that are involved in fatty acid metabolism. ELOVL4 encodes a putative transmembrane protein of 314 amino acids that carries a possible endoplasmic reticulum (ER) retention/retrieval signal (KXKXX) at the C-terminus. Two distinct mutations, a 5-bp deletion and a complex mutation from the same region in exon 6 of this gene, have been reported so far and are associated with autosomal dominant atrophic macular degeneration (adMD/STGD3). Both of these deletions could result in C-terminal truncation and loss of the ER retention signal in the mutant protein. We expressed the wild-type and mutant proteins in COS-7 and CHO cells to study the intracellular distribution of ELOVL4 and to identify possible implications of the above mutations in its localization. Immunofluorescence analysis of these proteins along with organelle marker antibodies revealed predominant ER localization for wild-type ELOVL4. Targeted deletion of the dilysine motif at the C-terminus of the protein resulted in the loss of ER localization. Immunoelectron microscopy and immunofluorescence analysis revealed a similar ER localization pattern for the protein in human photoreceptors. These data indicate that ELOVL4 is an ER-resident protein, which supports its suggested function in fatty acid elongation. We also demonstrate that the localization of both mutant proteins was dramatically changed from an ER to a Golgi distribution. Our observations suggest that the consequences of defective protein trafficking could underlie the molecular mechanism associated with degeneration of the macula in the patients with adMD/STGD3.


Asunto(s)
Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Degeneración Macular/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Western Blotting , Brefeldino A/farmacología , Células CHO , Células COS , Cricetinae , ADN/metabolismo , Electroforesis en Gel de Poliacrilamida , Retículo Endoplásmico/metabolismo , Colorantes Fluorescentes/farmacología , Eliminación de Gen , Genes Dominantes , Aparato de Golgi/metabolismo , Proteínas Fluorescentes Verdes , Humanos , Immunoblotting , Inmunohistoquímica , Proteínas Luminiscentes/metabolismo , Lisina/química , Degeneración Macular/patología , Microscopía Fluorescente , Microscopía Inmunoelectrónica , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Nocodazol/farmacología , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Retina/metabolismo , Homología de Secuencia de Aminoácido , Fracciones Subcelulares , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA