Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 7(5): e37197, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615940

RESUMEN

PURPOSE: AAV-mediated gene therapy in the rd10 mouse, with retinal degeneration caused by mutation in the rod cyclic guanosine monophosphate phosphodiesterase ß-subunit (PDEß) gene, produces significant, but transient, rescue of photoreceptor structure and function. This study evaluates the ability of AAV-mediated delivery of X-linked inhibitor of apoptosis (XIAP) to enhance and prolong the efficacy of PDEß gene-replacement therapy. METHODS: Rd10 mice were bred and housed in darkness. Two groups of animals were generated: Group 1 received sub-retinal AAV5-XIAP or AAV5-GFP at postnatal age (P) 4 or 21 days; Group 2 received sub-retinal AAV5-XIAP plus AAV5- PDEß, AAV5-GFP plus AAV5- PDEß, or AAV- PDEß alone at age P4 or P21. Animals were maintained for an additional 4 weeks in darkness before being moved to a cyclic-light environment. A subset of animals from Group 1 received a second sub-retinal injection of AAV8-733-PDEß two weeks after being moved to the light. Histology, immunohistochemistry, Western blots, and electroretinograms were performed at different times after moving to the light. RESULTS: Injection of AAV5-XIAP alone at P4 and 21 resulted in significant slowing of light-induced retinal degeneration, as measured by outer nuclear thickness and cell counts, but did not result in improved outer segment structure and rhodopsin localization. In contrast, co-injection of AAV5-XIAP and AAV5-PDEß resulted in increased levels of rescue and decreased rates of retinal degeneration compared to treatment with AAV5-PDEß alone. Mice treated with AAV5-XIAP at P4, but not P21, remained responsive to subsequent rescue by AAV8-733-PDEß when injected two weeks after moving to a light-cycling environment. CONCLUSIONS: Adjunctive treatment with the anti-apoptotic gene XIAP confers additive protective effect to gene-replacement therapy with AAV5-PDEß in the rd10 mouse. In addition, AAV5-XIAP, when given early, can increase the age at which gene-replacement therapy remains effective, thus effectively prolonging the window of opportunity for therapeutic intervention.


Asunto(s)
Inhibidores de Caspasas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Terapia Genética/métodos , Degeneración Retiniana/terapia , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/biosíntesis , Dependovirus/genética , Modelos Animales de Enfermedad , Electrorretinografía , Vectores Genéticos , Luz/efectos adversos , Ratones , Ratones Endogámicos C57BL , Células Fotorreceptoras de Vertebrados/metabolismo , Degeneración Retiniana/genética , Proteína Inhibidora de la Apoptosis Ligada a X/uso terapéutico
2.
Invest Ophthalmol Vis Sci ; 52(7): 4193-9, 2011 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-21421874

RESUMEN

PURPOSE: To examine the activation of autophagy and its relationship to Fas-mediated photoreceptor apoptosis during experimental retinal detachment. METHODS: Retina-retinal pigment epithelium (RPE) separation was created in Brown-Norway rats by subretinal injection of 1% hyaluronic acid and the intraretinal levels of the autophagy proteins LC3 and Atg5, the time course of LC3-I to LC3-II conversion, and the activation of cathepsins B and D were assayed with Western blot analysis and immunohistochemistry. We measured the ability of a Fas-activating antibody to induce LC3-I to LC3-II conversion in 661W cells, and the in vivo effect of Met12, a small molecule inhibitor of the Fas receptor, on LC3-I to LC3-II conversion and Atg5 expression. Autophagy activation was inhibited using 3-methyladenine (3-MA) or siRNA knockdown of Atg5 and the effect on apoptosis was measured using a caspase 8 activity assay, caspase 8 immunoblots, and photoreceptor TUNEL staining. RESULTS: Retina-RPE separation resulted in a Fas-dependent activation of autophagy, with increased Atg5 levels and intraphotoreceptor conversion of LC3-I to LC3-II. Detached retinas had increased levels of autophagosome-associated lysosomal proteases, cathepsins B and D. Inhibition of autophagy by 3-MA or siAtg5 accelerated the time course of caspase 8 activation and photoreceptor TUNEL staining. CONCLUSIONS: Autophagy activation occurs in the photoreceptors after retina-RPE separation. This appears to be, at least in part, dependent on Fas receptor activation, and plays a role in regulating the level of photoreceptor apoptosis.


Asunto(s)
Apoptosis , Autofagia , Células Fotorreceptoras de Vertebrados , Desprendimiento de Retina/fisiopatología , Receptor fas/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia , Western Blotting , Caspasa 8/metabolismo , Catepsina B/metabolismo , Catepsina D/metabolismo , Línea Celular , Activación Enzimática/efectos de los fármacos , Inmunohistoquímica , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas/genética , ARN Interferente Pequeño/farmacología , Ratas , Ratas Endogámicas BN , Factores de Tiempo
3.
Invest Ophthalmol Vis Sci ; 51(4): 2177-84, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19850829

RESUMEN

Purpose. To test the effect of a small peptide inhibitor (Met12) of the Fas receptor on the activation of extrinsic and intrinsic apoptosis pathways after retinal detachment. Methods. Retinal-RPE separation was created in Brown Norway rats by subretinal injection of 1% hyaluronic acid. Met12, derived from the Fas-binding extracellular domain of the oncoprotein Met, was injected into the subretinal space at the time of separation. A mutant peptide and vehicle administered in a similar fashion acted as inactive controls. The extrinsic apoptotic pathway was induced in 661W cells using a Fas-activating antibody in the presence or absence of Met12. Caspase 3, caspase 8, and caspase 9 activities were measured with calorimetric and luminescent assays in retinal extracts and cell lysates. Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) was performed in retinal sections 3 days after separation. Histology was performed in retinal sections 2 months after retinal detachment. Results. Met12 inhibited Fas-induced caspase 8 activation in 661W cells. Similarly, administration of Met12 into the subretinal space inhibited the activation of caspase 3, caspase 8, and caspase 9 after retinal detachment. This corresponded to a decreased level of TUNEL-positive staining of photoreceptors after retinal-RPE separation in animals that received Met12, but not inactive mutant, peptide treatment. After 2 months, the outer nuclear layer was significantly thicker, and the photoreceptor count was higher in animals treated with subretinal Met12. Conclusions. The small peptide Met12 may serve as a photoreceptor-protective agent in the setting of retinal-RPE separation.


Asunto(s)
Apoptosis/efectos de los fármacos , Oligopéptidos/farmacología , Células Fotorreceptoras de Vertebrados/patología , Desprendimiento de Retina/prevención & control , Receptor fas/antagonistas & inhibidores , Animales , Western Blotting , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Caspasa 9/metabolismo , Recuento de Células , Técnicas de Cultivo de Célula , Supervivencia Celular , Etiquetado Corte-Fin in Situ , Masculino , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Ratas , Ratas Endogámicas BN , Receptor fas/metabolismo
4.
Invest Ophthalmol Vis Sci ; 50(3): 1448-53, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19060276

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Dependovirus/genética , Terapia Genética , Células Fotorreceptoras de Vertebrados/patología , Desprendimiento de Retina/terapia , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Animales , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Modelos Animales de Enfermedad , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Etiquetado Corte-Fin in Situ , Masculino , Fármacos Neuroprotectores , Células Fotorreceptoras de Vertebrados/enzimología , Ratas , Ratas Endogámicas BN , Desprendimiento de Retina/enzimología , Transfección
5.
Invest Ophthalmol Vis Sci ; 49(7): 3193-200, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18378582

RESUMEN

PURPOSE: To test the hypothesis that interleukin (IL)-6 prevents photoreceptor cell death during periods of retinal separation from the retinal pigment epithelium (RPE). METHODS: Retinal-RPE separation was created in wild-type C57BL mice, IL-6(-/-) mice, and Brown Norway rats by subretinal injection of 1% hyaluronic acid. In some animals, anti-IL-6 neutralizing antibody (NAB) or exogenous IL-6 was administered into the subretinal space at the time of separation or at specified times afterward. Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) was performed 3 days after separation to detect apoptotic photoreceptors. Photoreceptor cell counts were performed after 1 and 2 months. RESULTS: Loss of IL-6 function through genetic ablation (IL-6(-/-) mice) or injection of anti-IL-6 NAB resulted in significantly increased levels of TUNEL-positive staining 3 days after retinal-RPE separation. One month after separation, outer nuclear layer (ONL) cell counts were significantly lower in IL-6(-/-) mice or in animals injected with anti-IL-6 NAB than in controls. Gain of IL-6 function through the addition of exogenous IL-6 resulted in significantly increased ONL counts at 1 month but not at 2 months. Reinjection of IL-6 at 1 month led to continued preservation of ONL counts compared with controls. A window of opportunity for treatment was detected because delaying injection of exogenous IL-6 to 2 weeks after retinal-RPE separation still resulted in significantly greater ONL cell counts compared with controls. CONCLUSIONS: IL-6 may serve as a photoreceptor neuroprotectant in the setting of retinal-RPE separation.


Asunto(s)
Interleucina-6/farmacología , Fármacos Neuroprotectores/farmacología , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Desprendimiento de Retina/fisiopatología , Animales , Anticuerpos/farmacología , Apoptosis/efectos de los fármacos , Recuento de Células , Esquema de Medicación , Humanos , Interleucina-6/administración & dosificación , Interleucina-6/deficiencia , Interleucina-6/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/antagonistas & inhibidores , Fármacos Neuroprotectores/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Ratas , Ratas Endogámicas BN , Retina/efectos de los fármacos , Retina/patología , Desprendimiento de Retina/patología , Factores de Tiempo
6.
Arch Ophthalmol ; 125(10): 1389-95, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17923548

RESUMEN

OBJECTIVE: To determine whether inhibiting the Fas proapoptosis pathway will result in increased photoreceptor survival after separation of the retina from the retinal pigment epithelium (RPE). METHODS: Retina/RPE separation was induced in rat and mouse eyes by the subretinal injection of hyaluronic acid, 1%. Fas-pathway signaling was inhibited by the concomitant injection of a Fas receptor-neutralizing antibody, small inhibitory RNA against the Fas-receptor transcript (siFAS), or the use of the Fas-receptor defective mouse strain LPR. Indices of photoreceptor death included terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) staining, cell counts, and retinal thickness measurements. Retinas were immunostained with antibodies against rhodopsin and cone opsin to evaluate rod and cone photopigment production, respectively. RESULTS: Inhibition of Fas signaling using Fas receptor-neutralizing antibody, siFas, or LPR mice resulted in a significant reduction in the number of TUNEL-positive photoreceptor cells as well as in a significant preservation of outer nuclear layer cell counts and thickness as compared with retina/RPE separation in eyes with intact Fas signaling. Fas-pathway inhibition resulted in preservation of both rhodopsin- and cone opsin-positive cells. CONCLUSIONS: Inhibition of the Fas proapoptosis pathway results in significant photoreceptor preservation after retinal separation from the RPE. CLINICAL RELEVANCE: Fas-pathway inhibition might serve as a novel mechanism for preserving photoreceptor cells during retinal disease.


Asunto(s)
Células Fotorreceptoras de Vertebrados/fisiología , Transducción de Señal/fisiología , Receptor fas/fisiología , Animales , Anticuerpos Bloqueadores/farmacología , Apoptosis/efectos de los fármacos , Recuento de Células , Supervivencia Celular/fisiología , Técnica del Anticuerpo Fluorescente Indirecta , Immunoblotting , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , ARN/aislamiento & purificación , ARN Interferente Pequeño/farmacología , Ratas , Ratas Endogámicas BN , Desprendimiento de Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rodopsina/metabolismo , Opsinas de Bastones/metabolismo , Receptor fas/antagonistas & inhibidores
7.
Invest Ophthalmol Vis Sci ; 45(12): 4563-9, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15557468

RESUMEN

PURPOSE: To determine whether the FAS-mediated apoptosis pathway becomes activated in the retina after retinal detachment and to investigate the temporal relationship between the activation of the FAS-pathway and the intrinsic apoptosis pathway involving caspase-9 and cytochrome c. METHODS: Experimental retinal detachments were created in Brown-Norway rats by injecting 10% hyaluronic acid into the subretinal space. Retinal tissue was harvested at 2, 4, 8, 24, 72, and 168 hours after creation of the detachment. Immunoprecipitation was performed to assess for FAS-receptor/FAS-ligand complex formation, and activation of caspase-8 and BID (a member of the Bcl-2 family of proteins) was assessed by Western blot analysis. A caspase-9 activity assay and immunoprecipitation of the caspase-9/cytochrome c complex were performed at these same time points. Specific pathway inhibition was performed with the caspase-9 inhibitor zLEHD.fmk or neutralizing antibodies against either the FAS-receptor or FAS-ligand. Transcription levels of FAS and intrinsic pathway intermediates were assessed as a function of time after retinal detachment by using quantitative real-time polymerase chain reaction. RESULTS: Retinal detachment resulted in the time-dependent formation of the FAS-receptor/FAS-ligand complex that preceded the peak of caspase-9 activity and caspase-9/cytochrome c complex formation. Cleavage of caspase-8 and truncation of BID were also observed. Injection of zLEHD.fmk into the subretinal space of a detached retina resulted in decreased caspase-9 activity, as did injection of anti-FAS-receptor antibody into either the subretinal space or the vitreous. Retinal detachment resulted in the transcriptional upregulation of the FAS-receptor, FAS-ligand, caspase-8 and BID, but not caspase-9 and cytochrome c. CONCLUSIONS: The FAS-mediated apoptosis pathway becomes activated and transcriptionally upregulated after retinal detachment. The peak of FAS activation precedes that of the intrinsic pathway, and inhibition of FAS activation can decrease caspase-9 activity.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Citocromos c/metabolismo , Glicoproteínas de Membrana/metabolismo , Desprendimiento de Retina/fisiopatología , Receptor fas/metabolismo , Animales , Anticuerpos/farmacología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3 , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Caspasa 3 , Caspasa 8 , Caspasa 9 , Inhibidores de Caspasas , Caspasas/química , Caspasas/genética , Citocromos c/genética , Inhibidores Enzimáticos/farmacología , Proteína Ligando Fas , Masculino , Glicoproteínas de Membrana/genética , Oligopéptidos/farmacología , Fragmentos de Péptidos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas BN , Desprendimiento de Retina/enzimología , Factores de Tiempo , Receptor fas/genética , Receptor fas/inmunología
8.
Front Biosci ; 9: 448-56, 2004 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-14766381

RESUMEN

The role of Fas ligand (FasL) in tumor immune privilege is controversial. In this study, 22 human tumor cell lines reported to be FasL+ were reevaluated by Western blot analysis, ELISA, and a functional assay. None of the cells lines expressed FasL. To assess whether human tumors express FasL in vivo, susceptibility to FasL-mediated killing was evaluated. About 75% of the 22 tumors tested were sensitive to FasL-mediated apoptosis, suggesting, therefore, that only about 25% could possibly express FasL. To investigate whether "FasL+" human tumor cells could suppress the proinflammatory effects of FasL in vivo, FasL transfectants were generated from two prototype "FasL+" tumor cell lines. The transfectants expressing FasL were rejected by SCID mice. In contrast, all the mice inoculated with parental tumor cells developed large tumors. These results suggested that human tumor cells that express FasL and resist both FasL-mediated apoptosis and inflammation are rare or nonexistant. We concluded that FasL expression is not a main mechanism that tumors use to counteract antitumor immunity.


Asunto(s)
Glicoproteínas de Membrana/inmunología , Neoplasias/inmunología , Receptor fas/inmunología , Animales , Especificidad de Anticuerpos , Muerte Celular , Relación Dosis-Respuesta Inmunológica , Proteína Ligando Fas , Femenino , Humanos , Inmunidad , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Desnudos , Ratones SCID , Células Tumorales Cultivadas
9.
J Immunol ; 170(7): 3806-11, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12646647

RESUMEN

Dendritic cell (DC)-based antitumor vaccine is a novel cancer immunotherapy that is promising for reducing cancer-related mortality. However, results from early clinical trials were suboptimal. A possible explanation is that many tumors secrete immunosuppressive factors such as TGF-beta, which may hamper host immune response to DC vaccine. In this study, we demonstrated that TGF-beta produced by tumors significantly reduced the potency of DC/tumor fusion vaccines. TGF-beta-secreting (CT26-TGF-beta) stable mouse colon cancer cell lines were generated using a retroviral vector expressing TGF-beta. A non-TGF-beta-secreting (CT26-neo) cell line was generated using an empty retroviral vector. The efficacies of DC/tumor fusion vaccines were assessed in vitro and in vivo. DC/CT26-TGF-beta fusion cells failed to induce a strong T cell proliferative response in vitro, mainly due to the effect of TGF-beta on T cell responsiveness rather than DC stimulatory capability. Animals vaccinated with DC/CT26-TGF-beta fusion vaccine had lower tumor-specific CTL activity and had significantly lower survival after tumor challenge as compared with animals immunized with DC/CT26-neo hybrids (45 vs 77%, p < 0.05). Ex vivo exposure of DCs to TGF-beta did not appear to lessen the efficacy of DC vaccine. These data suggest that tumor-derived TGF-beta reduces the efficacy of DC/tumor fusion vaccine via an in vivo mechanism. Neutralization of TGF-beta produced by the fusion cells may enhance the effectiveness of DC-based immunotherapy.


Asunto(s)
Vacunas contra el Cáncer/efectos adversos , Células Dendríticas/trasplante , Proteínas de Neoplasias/efectos adversos , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Factores Supresores Inmunológicos/efectos adversos , Factor de Crecimiento Transformador beta/efectos adversos , Células Tumorales Cultivadas/trasplante , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Fraccionamiento Celular , Sistema Libre de Células/inmunología , Sistema Libre de Células/metabolismo , Técnicas de Cocultivo , Citotoxicidad Inmunológica/genética , Células Dendríticas/inmunología , Femenino , Inyecciones Intraperitoneales , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos BALB C , Proteínas de Neoplasias/sangre , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/mortalidad , Factores Supresores Inmunológicos/sangre , Factores Supresores Inmunológicos/genética , Factores Supresores Inmunológicos/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Factor de Crecimiento Transformador beta/administración & dosificación , Factor de Crecimiento Transformador beta/sangre , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1 , Células Tumorales Cultivadas/inmunología , Células Tumorales Cultivadas/metabolismo , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...