Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Graefes Arch Clin Exp Ophthalmol ; 255(8): 1581-1592, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28523456

RESUMEN

PURPOSE: Ischemia-associated retinal degeneration is one of the leading causes of vision loss, and to date, there are no effective treatment options. We hypothesized that delayed injection of bone-marrow stem cells (BMSCs) 24 h after the onset of ischemia could effectively rescue ischemic retina from its consequences, including apoptosis, inflammation, and increased vascular permeability, thereby preventing retinal cell loss. METHODS: Retinal ischemia was induced in adult Wistar rats by increasing intraocular pressure (IOP) to 130-135 mmHg for 55 min. BMSCs harvested from rat femur were injected into the vitreous 24 h post-ischemia. Functional recovery was assessed 7 days later using electroretinography (ERG) measurements of the a-wave, b-wave, P2, scotopic threshold response (STR), and oscillatory potentials (OP). The retinal injury and anti-ischemic effects of BMSCs were quantitated by measuring apoptosis, autophagy, inflammatory markers, and retinal-blood barrier permeability. The distribution and fate of BMSC were qualitatively examined using real-time fundus imaging, and retinal flat mounts. RESULTS: Intravitreal delivery of BMSCs significantly improved recovery of the ERG a- and b-waves, OP, negative STR, and P2, and attenuated apoptosis as evidenced by decreased TUNEL and caspase-3 protein levels. BMSCs significantly increased autophagy, decreased inflammatory mediators (TNF-α, IL-1ß, IL-6), and diminished retinal vascular permeability. BMSCs persisted in the vitreous and were also found within ischemic retina. CONCLUSIONS: Taken together, our results indicate that intravitreal injection of BMSCs rescued the retina from ischemic damage in a rat model. The mechanisms include suppression of apoptosis, attenuation of inflammation and vascular permeability, and preservation of autophagy.


Asunto(s)
Células de la Médula Ósea/citología , Isquemia/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Degeneración Retiniana/terapia , Vasos Retinianos/patología , Animales , Apoptosis , Barrera Hematorretinal , Permeabilidad Capilar , Modelos Animales de Enfermedad , Electrorretinografía , Etiquetado Corte-Fin in Situ , Inyecciones Intravítreas , Isquemia/diagnóstico , Isquemia/metabolismo , Masculino , Ratas , Ratas Wistar , Degeneración Retiniana/diagnóstico , Degeneración Retiniana/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/fisiopatología
2.
Exp Eye Res ; 93(4): 340-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21094639

RESUMEN

We previously described the phenomenon of retinal ischemic pre-conditioning (IPC) and we have shown the role of various signaling proteins in the protective pathways, including the mitogen-activated protein kinase p38. In this study we examined the role in IPC of mitogen-activated protein kinase phosphatase-1 (MKP-1), which inactivates p38. Ischemia was produced by elevation of intraocular pressure above systolic arterial blood pressure in adult Wistar rats. Preconditioning was produced by transient retinal ischemia for 5 min, 24 h prior to ischemia. Small interfering RNA (siRNA) to MKP-1 or a control non-silencing siRNA, was injected into the vitreous 6 h prior to IPC. Recovery was assessed by electroretinography (ERG) and histology. The a-and b-waves, and oscillatory potentials (OPs), measured before and 1 week after ischemia, were then normalized relative to pre-ischemic baseline, and corrected for diurnal variation in the normal non-ischemic eye. The P2, or post-photoreceptor component of the ERG (which reflects function of the rod bipolar cells in the inner retina), was derived using the Hood-Birch model. MKP-1 was localized in specific retinal cells using immunohistochemistry; levels of mitogen-activated protein kinases were measured using Western blotting. Injection of siRNA to MKP-1 significantly attenuated the protective effect of IPC as reflected by decreased recovery of the electroretinogram a and b-waves and the P2 after ischemia. The injection of siRNA to MKP-1 reduced the number of cells in the retinal ganglion cell and outer nuclear layers after IPC and ischemia. Blockade of MKP-1 by siRNA also increased the activation of p38 at 24 h following IPC. MKP-1 siRNA did not alter the levels of phosphorylated jun N-terminal kinase (JNK) or extracellular signal-regulated kinase (ERK) after IPC. The results suggest the involvement of dual-specificity phosphatase MKP-1 in IPC and that MKP-1 is involved in IPC by regulating levels of activated MAPK p38.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/fisiología , Precondicionamiento Isquémico , Daño por Reperfusión/prevención & control , Enfermedades de la Retina/prevención & control , Vasos Retinianos/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Western Blotting , Técnicas de Cultivo de Célula , Electrorretinografía , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , MAP Quinasa Quinasa 4/metabolismo , Fosforilación , Interferencia de ARN , Ratas , Ratas Wistar , Daño por Reperfusión/enzimología , Daño por Reperfusión/fisiopatología , Retina/enzimología , Retina/fisiopatología , Células Bipolares de la Retina/fisiología , Enfermedades de la Retina/enzimología , Enfermedades de la Retina/fisiopatología , Células Ganglionares de la Retina/metabolismo
3.
Exp Eye Res ; 92(6): 521-7, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21501608

RESUMEN

In previous studies, it was shown that post-conditioning, a transient period of brief ischemia following prolonged severe ischemia in the retina, could provide significant improvement in post-ischemic recovery, attenuation of cell loss, and decreased apoptosis. These studies showed that post-conditioning effectively prevented damage after retinal ischemia when it was instituted early (within 1 h) in the post-ischemic period. While post-ischemic conditioning holds high promise of clinical translation, patients often present late after the onset of retinal ischemia and therefore immediate application of this anti-ischemic maneuver is generally not feasible. In this study, we examined the hypothesis that application of a post-conditioning stimulus at 24 h or greater following the end of prolonged ischemia would decrease the extent of ischemic injury. Ischemia was induced in rat retina in vivo. Recovery after ischemia followed by 5 min of post-conditioning brief ischemia 24 or 48 h after prolonged ischemia was assessed functionally (electroretinography) and histologically at 7 days after ischemia and post-conditioning or sham post-conditioning. We found that the brief ischemic stimulus applied 24, but not 48 h after prolonged ischemia significantly improved functional recovery and decreased histological damage induced by prolonged ischemia. We conclude that within a defined time window, delayed post-ischemic conditioning ameliorated post-ischemic injury in rats. Compared to earlier studies, the present work demonstrates for the first time the novel ability of a significantly delayed ischemic stimulus to provide robust neuroprotection in the retina following ischemia.


Asunto(s)
Poscondicionamiento Isquémico , Daño por Reperfusión/prevención & control , Enfermedades de la Retina/prevención & control , Vasos Retinianos/fisiología , Animales , Apoptosis/fisiología , Presión Sanguínea/fisiología , Recuento de Células , Citoprotección , Electrorretinografía , Presión Intraocular/fisiología , Ratas , Ratas Wistar , Recuperación de la Función , Daño por Reperfusión/fisiopatología , Enfermedades de la Retina/fisiopatología , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/fisiología , Tonometría Ocular
4.
Exp Eye Res ; 91(6): 844-52, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20599964

RESUMEN

Ischemic pre-conditioning (IPC) provides neuroprotection in the rat retina from the damaging effects of severe ischemia. Recently, neuroprotection by retinal ischemic post-conditioning (Post-C), i.e., transient ischemia after more lengthy, damaging ischemia, was described, but its mechanisms are not yet known. One possible explanation of the effectiveness of Post-C is that it augments intrinsic neuroprotective mechanisms initiated during ischemia. Increasing duration of the damaging ischemic insult may therefore impact the effectiveness of Post-C. IPC, in contrast, sets in motion a series of neuroprotective events prior to the onset of ischemia. Thus, IPC and Post-C may operate by differing mechanisms. Accordingly, we examined the effect of retinal ischemic duration on post-ischemic outcome in vivo in rats after adding Post-C, and the impact of combining pre- and post-conditioning. Recovery after ischemia performed 24 h after IPC, or after Post-C performed 5 min after ischemia ended, was assessed functionally (electroretinography) and histologically at 7 days after ischemia. Durations of ischemia of 45 and 55 min were studied. Since recovery with IPC or Post-C alone, with 55 min of ischemia, did not achieve the same degree of effect (i.e., not complete recovery) exhibited in our previous studies of IPC using a different ischemia model, we also combined IPC and Post-C to test the hypothesis of the possible additive effects of the IPC and Post-C. We found that the recovery after Post-C was enhanced to a greater degree when ischemia was of longer duration. Post-C led to greater post-ischemic recovery compared to IPC. Both IPC and Post-C also attenuated structural damage to the retina. Contrary to our hypothesis, IPC and Post-C did not combine to enhance recovery after ischemia. In earlier studies, IPC attenuated post-ischemic apoptosis. To begin to examine the mechanism of Post-C, we studied its impact on apoptosis following ischemia. We examined apoptosis by determining the percentage of TUNEL-positive cells at 24 h after ischemia. Post-C attenuated apoptosis, but when combined with IPC, TUNEL was similar in the combined group to that of ischemia alone. We also examined the role of the recruitment of an inflammatory response in ischemia and Post-C. We found that inflammatory markers increased by ischemia were not altered by Post-C. We conclude that Post-C effectiveness depends upon the duration of ischemia; Post-C is not additive with IPC, and Post-C functions, in part, by preventing apoptotic damage to the inner retina. Post-C has considerable promise for clinical translation to eye diseases that cause blindness by ischemia.


Asunto(s)
Poscondicionamiento Isquémico , Daño por Reperfusión/prevención & control , Enfermedades de la Retina/prevención & control , Vasos Retinianos/fisiopatología , Animales , Apoptosis/fisiología , Electrorretinografía , Técnica del Anticuerpo Fluorescente Indirecta , Etiquetado Corte-Fin in Situ , Mediadores de Inflamación/metabolismo , Presión Intraocular/fisiología , Ratas , Ratas Wistar , Daño por Reperfusión/metabolismo , Daño por Reperfusión/fisiopatología , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/fisiopatología , Factores de Tiempo , Isquemia Tibia
5.
Exp Eye Res ; 89(5): 782-90, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19631642

RESUMEN

In previous studies, inhibition of mitogen-activated protein kinase (MAP) p38 significantly improved recovery and attenuated apoptosis after retinal ischemia in rats. Yet, ischemic preconditioning (IPC) attenuated the ischemia-induced increase in p38 expression. We hypothesized that p38 was required for induction of ischemic tolerance by IPC. We examined the mechanisms of involvement of p38 in IPC neuroprotection. IPC or ischemia was induced in rat retina in vivo. Recovery after ischemia performed 24h after IPC was assessed functionally (electroretinography) and histologically at 7d after ischemia in the presence or absence of inhibition of p38. We examined the role of p38alpha in the mimicking of IPC produced by opening mitochondrial KATP channels using diazoxide, or stimulation of p38 activation by anisomycin. The importance of adenosine receptors in p38 activation after IPC was assessed using specific blockers of adenosine A1 and A2a receptors. Interfering RNA (siRNA) or SB203580 was used to block p38alpha. Phosphorylated p38 levels were measured. Phosphorylated p38 protein increased with IPC. Interfering RNA (siRNA) to p38alpha prior to IPC, or inhibiting p38 activation with SB203580, with ischemia following 24h later, significantly attenuated the neuroprotective effect of IPC. Anisomycin administered to increase p38 mimicked IPC, an effect blocked by SB203580. IPC-mimicking with diazoxide, an opener of mitochondrial KATP channels, was diminished with p38alpha siRNA. Adenosine receptor blockade did not decrease the elevated levels of phosphorylated p38 after IPC. Specific inhibition of p38alpha suggests that this MAPK is involved in the protective effects of IPC, and that p38 is downstream of mitochondrial KATP channels, but not adenosine receptors, in this neuroprotection.


Asunto(s)
Isquemia/terapia , Precondicionamiento Isquémico , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Daño por Reperfusión/prevención & control , Neuronas Retinianas/enzimología , Vasos Retinianos/fisiopatología , Antagonistas del Receptor de Adenosina A1 , Antagonistas del Receptor de Adenosina A2 , Animales , Anisomicina/farmacología , Diazóxido/farmacología , Modelos Animales de Enfermedad , Electrorretinografía , Activadores de Enzimas/farmacología , Imidazoles/farmacología , Presión Intraocular , Isquemia/complicaciones , Isquemia/enzimología , Isquemia/patología , Isquemia/fisiopatología , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/genética , Fosforilación , Canales de Potasio/agonistas , Canales de Potasio/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A2A/metabolismo , Daño por Reperfusión/enzimología , Daño por Reperfusión/etiología , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Neuronas Retinianas/patología , Transducción de Señal , Factores de Tiempo
6.
Exp Eye Res ; 88(3): 512-21, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19084003

RESUMEN

Potent endogenous protection from ischemia can be induced in the retina by ischemic preconditioning (IPC). Protein kinase B/Akt is a cellular survival factor. We hypothesized that Akt was integral to IPC based upon differential effects of Akt subtypes. Rats were subjected to retinal ischemia after IPC or IPC-mimicking by the opening of mitochondrial KATP (mKATP) channels. The effects of blocking Akt using wortmannin, API-2, or small interfering RNA (siRNA) were examined. Electroretinography assessed functional recovery after ischemia, and TUNEL examined retinal ganglion cell apoptosis. We studied the relationship between Akt activation and known initiators of IPC, including adenosine receptor stimulation and the opening of mKATP channels. The PI-3 kinase inhibitor wortmannin 1 or 4 mg/kg (i.p.), the specific Akt inhibitor API-2, 5-500 microM in the vitreous, or intravitreal siRNA directed against Akt2 or -3, but not Akt1, significantly attenuated the neuroprotective effect of IPC. Interfering RNA against any of the three Akt subtypes significantly but time-dependently attenuated mKATP channel opening to mimic IPC. Adenosine A1 receptor blockade (DPCPX), A2a blockade (CSC), or the mKATP channel blocker 5-hydroxydecanoic acid significantly attenuated Akt activation after IPC. Interfering RNA directed against Akt subtypes prevented the ameliorative effect of IPC on post-ischemic apoptosis. All three Akt subtypes are involved in functional retinal neuroprotection by IPC or IPC-mimicking. Akt is downstream of adenosine A1 and A2a receptors and mKATP channel opening. The results indicate the presence in the retina of robust and redundant endogenous neuroprotection based upon subtypes of Akt.


Asunto(s)
Isquemia/enzimología , Precondicionamiento Isquémico/métodos , Proteínas Proto-Oncogénicas c-akt/fisiología , Vasos Retinianos/enzimología , Androstadienos/farmacología , Animales , Apoptosis , Western Blotting/métodos , Clorpropamida/análogos & derivados , Clorpropamida/farmacología , Diazóxido/farmacología , Relación Dosis-Respuesta a Droga , Electrorretinografía/efectos de los fármacos , Electrorretinografía/métodos , Isquemia/prevención & control , Isoenzimas/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Vasos Retinianos/patología , Wortmanina
7.
Anesthesiology ; 110(4): 774-80, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19322943

RESUMEN

BACKGROUND: The purpose of this study was to examine the role of erythropoietin in retinal ischemic preconditioning (IPC). METHODS: Rats were subjected to retinal ischemia after IPC. Electroretinography assessed functional recovery after ischemia; retinal sections were examined to determine loss of retinal ganglion cells, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling was used to assess apoptosis. Levels of downstream mediators were measured in retinal homogenates by Western blotting. To assess the involvement of erythropoietin in IPC, Western blotting was used to measure levels of erythropoietin and its receptor (EPO-R) in retinal homogenates after IPC. To examine erythropoietin's role in IPC, the impact of blocking erythropoietin via intravitreal injection of soluble EPO-R (sEPO-R) before IPC was studied. RESULTS: Erythropoietin levels did not change after IPC, but EPO-R increased. Intravitreal injection of sEPO-R significantly attenuated both the functional and histologic neuroprotection produced by IPC in comparison to control injection of denatured sEPO-R. Apoptotic damage after ischemia was enhanced in the sEPO-R-treated retinas as indicated by fluorescent terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling. Phosphorylated extracellular-signal-regulated kinase and heat shock protein 27, but not protein kinase B, upregulated in denatured sEPO-R-treated retinae, were attenuated in eyes injected with sEPO-R. CONCLUSIONS: These results indicate that EPO-R upregulation is a critical component of the functional, histologic, and antiapoptotic protective effect of IPC on ischemia in the retina and that several downstream effectors may be involved in the neuroprotective actions of erythropoietin.


Asunto(s)
Eritropoyetina/análisis , Precondicionamiento Isquémico , Receptores de Eritropoyetina/análisis , Daño por Reperfusión/prevención & control , Retina/efectos de los fármacos , Vasos Retinianos , Animales , Apoptosis , Western Blotting , ADN Nucleotidilexotransferasa , Electrorretinografía , Eritropoyetina/farmacología , Eritropoyetina/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Proteínas de Choque Térmico HSP27 , Etiquetado Corte-Fin in Situ , Neuronas/citología , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/enzimología , Retina/citología , Solubilidad
8.
Exp Eye Res ; 87(4): 300-11, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18722601

RESUMEN

The purpose of our study was to determine the specific subtypes of protein kinase C involved in the neuroprotection afforded by retinal ischemic preconditioning (IPC), their relationship to the opening of mitochondrial KATP (mKATP) channels, and their role in apoptosis after preconditioning and ischemia. Rats were subjected to retinal ischemia after IPC, or retinas were rendered ischemic after pharmacological opening of mKATP channels. Using immunohistochemistry and image analysis, we determined cellular localization of PKC subtypes. We blocked PKC-delta and -epsilon to study the effect on protection with IPC or with IPC-mimicking by the opening of mKATP channels. PKC subtypes were inhibited pharmacologically or with interfering RNA. Electroretinography assessed functional recovery after ischemia. IPC was effectively mimicked by injection of diazoxide to open the mKATP channel. IPC and/or its mimicking were attenuated by the PKC-delta inhibitor rottlerin and by interfering RNA targeting PKC-delta or -epsilon. Using TUNEL staining and Western blotting for caspase-3 and fodrin breakdown we assessed apoptosis. The injection of interfering RNA to PKC-delta and -epsilon before preconditioning significantly enhanced TUNEL staining as well as the cleavage of caspase-3 and fodrin after ischemia. In summary, our experiments have shown that both PKC-delta and -epsilon subtypes are involved in the cellular signaling that results in neuroprotection from IPC and that both are downstream of the opening of mKATP channels.


Asunto(s)
Precondicionamiento Isquémico , Proteína Quinasa C/fisiología , Daño por Reperfusión/prevención & control , Enfermedades de la Retina/prevención & control , Vasos Retinianos/enzimología , Acetofenonas/farmacología , Animales , Apoptosis , Benzopiranos/farmacología , Western Blotting , Células Cultivadas , Diazóxido/farmacología , Electrorretinografía , Inhibidores Enzimáticos/farmacología , Etiquetado Corte-Fin in Situ , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Isoenzimas/fisiología , Canales de Potasio/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , ARN Interferente Pequeño/farmacocinética , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/enzimología , Enfermedades de la Retina/enzimología , Enfermedades de la Retina/patología , Vasos Retinianos/patología , Vasodilatadores/farmacología
9.
Invest Ophthalmol Vis Sci ; 47(5): 2114-24, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16639023

RESUMEN

PURPOSE: To examine the mechanisms of ischemic preconditioning (IPC) related to the opening of mitochondrial KATP (mKATP) channels in the retina. METHODS: Rats were subjected to retinal ischemia after IPC, or retinas were rendered ischemic after pharmacological opening of mKATP channels. The effects of blocking mKATP channel opening, nitric oxide synthase (NOS) subtypes, or protein kinase C (PKC) on the protective effect of IPC or on the opening of mKATP channels were studied. Electroretinography assessed functional recovery after ischemia. Immunohistochemistry and image analysis were used to measure changes in levels of reactive oxygen species (ROS) and NOS subtypes and to determine their cellular localization. RESULTS: IPC was effectively mimicked by injection of the mKATP channel opener diazoxide. Both IPC and its mimicking by diazoxide were completely attenuated by the mKATP channel blocker 5-hydroxydecanoic acid (5-HD). Nonspecific blockade of NOS by N(omega)-nitro-L-arginine (L-NNA), but not by specific inducible (i)NOS or neuronal (n)NOS inhibitors, blunted IPC and IPC-mimicking, as did blockade of PKC. IPC and diazoxide IPC-mimicking significantly enhanced mitochondrial ROS production in the inner retina, an effect blocked by 5-HD. Mitochondrial ROS colocalized with e- and nNOS in retinal cells after stimulation with diazoxide. CONCLUSIONS: The results showed that IPC in the retina requires opening of the mKATP channel, and that IPC could be effectively mimicked using the mKATP channel opener diazoxide. eNOS-generated nitric oxide, PKC, and ROS are activated by opening of the mKATP channel.


Asunto(s)
Precondicionamiento Isquémico , Canales de Potasio/metabolismo , Daño por Reperfusión/prevención & control , Retina/metabolismo , Vasos Retinianos/fisiología , Animales , Ácidos Decanoicos/farmacología , Diazóxido/farmacología , Electrorretinografía , Técnica del Anticuerpo Fluorescente Indirecta , Hidroxiácidos/farmacología , Mitocondrias/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Nitroarginina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
10.
Invest Ophthalmol Vis Sci ; 57(7): 3522-32, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27367588

RESUMEN

PURPOSE: We have previously demonstrated the protective effect of bone marrow stem cell (BMSC)-conditioned medium in retinal ischemic injury. We hypothesized here that hypoxic preconditioning of stem cells significantly enhances the neuroprotective effect of the conditioned medium and thereby augments the protective effect in ischemic retina. METHODS: Rats were subjected to retinal ischemia by increasing intraocular pressure to 130 to 135 mm Hg for 55 minutes. Hypoxic-preconditioned, hypoxic unconditioned, or normoxic medium was injected into the vitreous 24 hours after ischemia ended. Recovery was assessed 7 days after injections by comparing electroretinography measurements, histologic examination, and apoptosis (TUNEL, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay). To compare proteins secreted into the medium in the groups and the effect of hypoxic exposure, we used rat cytokine arrays. RESULTS: Eyes injected with hypoxic BMSC-conditioned medium 24 hours after ischemia demonstrated significantly enhanced return of retinal function, decreased retinal ganglion cell layer loss, and attenuated apoptosis compared to those administered normoxic or hypoxic unconditioned medium. Hypoxic-preconditioned medium had 21 significantly increased protein levels compared to normoxic medium. CONCLUSIONS: The medium from hypoxic-preconditioned BMSCs robustly restored retinal function and prevented cell loss after ischemia when injected 24 hours after ischemia. The protective effect was even more pronounced than in our previous studies of normoxic conditioned medium. Prosurvival signals triggered by the secretome may play a role in this neuroprotective effect.


Asunto(s)
Células de la Médula Ósea/citología , Medios de Cultivo Condicionados/farmacología , Hipoxia/fisiopatología , Isquemia/terapia , Precondicionamiento Isquémico , Trasplante de Células Madre Mesenquimatosas/métodos , Retina/patología , Enfermedades de la Retina/terapia , Animales , Apoptosis , Células Cultivadas , Modelos Animales de Enfermedad , Electrorretinografía , Etiquetado Corte-Fin in Situ , Inyecciones Intraoculares , Isquemia/patología , Isquemia/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley , Retina/fisiopatología , Enfermedades de la Retina/patología , Enfermedades de la Retina/fisiopatología , Células Ganglionares de la Retina/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA