Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Recept Signal Transduct Res ; 31(6): 402-7, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22060185

RESUMEN

In this study, we investigated the neuroprotective effects of paclitaxel in transient cerebral ischemia and possible regulatory mechanism of these neuroprotection. Our data showed that paclitaxel can down-regulate the increased MLK3, JNK3, c-Jun, Bcl-2, and caspase-3 phosphorylation induced by ischemia injury. Cresyl violet staining and immunohistochemistry results demonstrated that paclitaxel had neuroprotective effect against ischemia/reperfusion-induced neuronal cell death. These results indicated that paclitaxel has neuroprotection in ischemic injury through JNK3 signaling pathway and provided a novel possible drug in therapeutics of brain ischemia.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Isquemia Encefálica/enzimología , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Fármacos Neuroprotectores/farmacología , Paclitaxel/farmacología , Daño por Reperfusión/enzimología , Moduladores de Tubulina/farmacología , Animales , Caspasa 3/metabolismo , Muerte Celular , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Neuronas/efectos de los fármacos , Neuronas/enzimología , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
2.
J Recept Signal Transduct Res ; 29(1): 38-43, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19519168

RESUMEN

Here we examined the effects of ischemia preconditioning and ketamine, an NMDA receptor antagonist, on the activation and its nucleus translocation of ERK5 in hippocampal CA1 region. Our results showed ERK5 was not activated in rat hippocampus CA1 region. But in cytosol extracts preconditioned with 3 min of sublethal ischaemia, ERK5 activation was enhanced significantly, with two peaks occurring at 3 hr and 3 days, respectively. This activation returned to base level 3 days later. The results lead us to conclude that preconditioning increased the activations of ERK5 during reperfusion after lethal ischemia through NMDA receptor. Preconditioning increased the activation and nucleus translocation of ERK5 during reperfusion after lethal ischemia through the NMDA receptor. These findings might provide some clues to understanding the mechanism underlying ischemia tolerance and to finding clinical therapies for stroke using the endogenous neuroprotection.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/metabolismo , Isquemia/metabolismo , Precondicionamiento Isquémico , Ketamina/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Transducción de Señal/fisiología , Transporte Activo de Núcleo Celular/fisiología , Animales , Activación Enzimática , Hipocampo/citología , Hipocampo/metabolismo , Masculino , Proteína Quinasa 7 Activada por Mitógenos/genética , Ratas , Ratas Sprague-Dawley
3.
Neurochem Res ; 34(11): 2008-21, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19449206

RESUMEN

Previous studies have shown that KA receptor subunit GluR6 mediated c-Jun N-terminal protein kinase (JNK) signaling is involved in global ischemia injury. Our present study indicates that focal ischemic brain insult on rat middle cerebral artery occlusion (MACo) model enhances the assembly of the GluR6-PSD95-MLK3 module and facilitates the phosphorylation of JNK. Most importantly, a peptide containing the TAT protein transduction sequence, Tat-GluR6-9c, can perturb the assembly of the GluR6-PSD95-MLK3 signaling module and suppress the activation of MLK3, MKK7/4 and JNK. As result, the inhibition of JNK activation caused by Tat-GluR6-9c diminishes the phosphorylation of the transcription factor c-Jun, down-regulates FasL expression and attenuates bax translocation, release of cytochrome c and the activation of caspase-3. Furthermore, MCAo induced infract volume is reduced by intracerebroventricular injection of Tat-Glur6-9c. Oxygen-glucose-deprivation (OGD) cultured cortical neuronal cell also shows an improved cell viability by application of Tat-GluR6-9c. Taken together, our findings strongly suggest that GluR6-PSD95-MLK3 signaling module mediated activation of nuclear and non-nuclear pathways of JNK activation are involved in focal ischemia injury and OGD. Tat-GluR6-9c, the peptide we constructed, gives a new insight into the therapy for ischemic stroke.


Asunto(s)
Glucosa/deficiencia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ataque Isquémico Transitorio/prevención & control , Proteínas de la Membrana/metabolismo , Fármacos Neuroprotectores/farmacología , Oxígeno/farmacología , Receptores de Ácido Kaínico/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Animales , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Homólogo 4 de la Proteína Discs Large , Técnicas In Vitro , Ataque Isquémico Transitorio/metabolismo , Masculino , Mutación , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores de Ácido Kaínico/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Receptor de Ácido Kaínico GluK2
4.
Hippocampus ; 19(1): 79-89, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18680160

RESUMEN

Our previous study showed that kainate (KA) receptor subunit GluR6 played an important role in ischemia-induced MLK3 and JNK activation and neuronal degeneration through the GluR6-PSD95-MLK3 signaling module. However, whether the KA receptors subunit GluR6 is involved in the activation of p38 MAP kinase during the transient brain ischemia/reperfusion (I/R) in the rat hippocampal CA1 subfield is still unknown. In this present study, we first evaluated the time-course of phospho-p38 MAP kinase at various time-points after 15 min of ischemia and then observed the effects of antagonist of KA receptor subunit GluR6, GluR6 antisence oligodeoxynucleotides on the phosphorylation of p38 MAP kinase induced by I/R. Results showed that inhibiting KA receptor GluR6 or suppressing the expression of KA receptor GluR6 could down-regulate the elevation of phospho-p38 MAP kinase induced by I/R. These drugs also reduced the phosphorylation of MLK3, MKK3/MKK6, MKK4, and MAPKAPK2. Additionally, our results indicated administration of three drugs, including p38 MAP kinase inhibitor before brain ischemia significantly decreased the number of TUNEL-positive cells detected at 3 days of reperfusion and increased the number of the surviving CA1 pyramidal cells at 5 days of reperfusion after 15 min of ischemia. Taken together, we suggest that GluR6-contained KA receptors can mediate p38 MAP kinase activation through a kinase cascade, including MLK3, MKK3/MKK6, and MKK4 and then induce increased phosphorylation of MAPKAPK-2 during ischemia injury and ultimately result in neuronal cell death in the rat hippocampal CA1 region.


Asunto(s)
Hipocampo/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Receptores de Ácido Kaínico/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Infarto Encefálico/metabolismo , Infarto Encefálico/patología , Infarto Encefálico/fisiopatología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Ácido Glutámico/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Hipoxia-Isquemia Encefálica/patología , Hipoxia-Isquemia Encefálica/fisiopatología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , MAP Quinasa Quinasa 3/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Oligodesoxirribonucleótidos Antisentido/farmacología , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Transmisión Sináptica/fisiología , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno , Receptor de Ácido Kaínico GluK2
5.
Zhonghua Yi Xue Za Zhi ; 87(17): 1211-3, 2007 May 08.
Artículo en Chino | MEDLINE | ID: mdl-17686245

RESUMEN

OBJECTIVE: To investigate the protective effects of the induction of heme oxygenase-1 (HO-1) on ischemia/reperfusion lung injury. METHODS: Forty Sprague-Dawley rats were randomly divided into four equal groups: sham group, lung ischemia/reperfusion injury (I/R) group, undergoing ligaturing of the left lung hilum for 30 minutes followed by reperfusion for 120 minutes; hemin group, undergoing intraperitoneal injection of hemin, an inducer of HO-1, 48 hours before the ligation and reperfusion; zinc protoporphyrin (ZnPP) group, undergoing intravenous injection of ZnPP, an inhibitor of heme oxygenase, 15 min after the ischemia-reperfusion; and sham operation group, undergoing sham operation. Two hours after the I/R arterial blood samples were collected and then the left lungs of the rats were taken out. Plasma tumor necrosis factor-alpha (TNF-alpha) and lung superoxide dismutase (SOD) activity were examined. Lung wet-to-dry weight (W/D) ratio was measured. The ultrastructure of the pulmonary alveoli and its capillaries were studied by using transmissional electronmicroscopy. RESULTS: The lung W/D ratio of the hemin group was 5.92 +/- 0.66, significantly lower than that of the I/R group (7.55 +/- 0.66, P < 0.01), and that of the ZnPP group (7.34 +/- 0.39, P < 0.01). The SOD activity of the hemin group was 6.5 +/- 0.6 U/mg protein, significantly higher than those of the I/R group and ZnPP group (2.8 +/- 0.4 U/mg protein and 3.0 +/- 0.4 U/mg protein respectively, both P < 0.01). The plasma TNF-alpha was 180.36 +/- 12.46, significantly lower than those of the I/R and ZnPP groups (452.26 +/- 22.59 and 438.59 +/- 30.26 respectively, both P < 0.01). Transmissional electronmicroscopy showed that the microscopic structure of the sham group was normal and that the pathological changes of hemin group were milder then those of the T/R and ZnPP groups. CONCLUSION: The induction of heme oxygenase-1 can protect effectively the lesion of lung pathology in ischemia reperfusion in vivo.


Asunto(s)
Hemo-Oxigenasa 1/biosíntesis , Enfermedades Pulmonares/patología , Daño por Reperfusión/patología , Animales , Modelos Animales de Enfermedad , Inducción Enzimática , Hemo-Oxigenasa 1/antagonistas & inhibidores , Inyecciones Intravenosas , Pulmón/metabolismo , Pulmón/patología , Pulmón/ultraestructura , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/prevención & control , Microscopía Electrónica de Transmisión , Protoporfirinas/administración & dosificación , Protoporfirinas/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/sangre
6.
J Neurosci Res ; 85(13): 2960-70, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17639597

RESUMEN

We investigated the possible relationships between KA2 subunit and GluR6 subunit, as well as the role of KA2 subunit in neuronal death induced by cerebral ischemia/reperfusion. Our results indicated that intracerebroventricular infusion of KA2 antisense oligodeoxynucleotides (AS) not only knocked down the expressions of KA2 and GluR6, but also suppressed the assembly of the GluR6/KA2-PSD95-MLK3 signaling module, and inhibited JNK activation and phosphorylation of c-jun. In addition, infusion of KA2 AS increased neuronal survival in CA1 region after 5 days of reperfusion. More interestingly, we found that the combination of KA2 and GluR6 AS exerted more significant effects than when pretreated with KA2 AS or GluR6 AS alone. Our results suggest that the KA2 subunit is involved in delayed neuronal death induced by cerebral ischemia, at the same time, it is noteworthy that the functional cooperation between KA2 and GluR6 subunits plays a critical role in the ischemic brain injury by PSD95-MLK3-MKK4/7-JNK3 signal pathway.


Asunto(s)
Isquemia Encefálica/metabolismo , Receptores de Ácido Kaínico/metabolismo , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Muerte Celular/efectos de los fármacos , Homólogo 4 de la Proteína Discs Large , Hipocampo/efectos de los fármacos , Hipocampo/patología , Etiquetado Corte-Fin in Situ/métodos , Inyecciones Intraventriculares , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Oligodesoxirribonucleótidos Antisentido/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Ácido Kaínico/química , Reperfusión/métodos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Tiempo , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno , Receptor de Ácido Kaínico GluK2
7.
Cell Signal ; 19(9): 1844-56, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17555943

RESUMEN

Increasing evidence suggests that the Bcl-2 family proteins play pivotal roles in regulation of the mitochondria cell-death pathway on transient cerebral ischemia. Bad, a BH3-only proapoptotic Bcl-2 family protein, has been shown to be phosphorylated extensively on serine by kinds of kinases. However, the exact mechanisms of the upstream kinases in regulation of Bad signaling pathway remain unknown. Here, we reported that Bad could be phosphorylated not only by Akt1 but also by JNK1/2 after transient global ischemia in rat hippocampal CA1 region. Our data demonstrated that Akt1 mediated the phosphorylation of Bad at serine 136, which increased the interaction of serine 136-phosphorylated Bad with 14-3-3 proteins and prevented the dimerization of Bad with Bcl-Xl, inhibited the release of cytochrome c to the cytosol and the death effector caspase-3 activation, leading to the survival of neuron. In contrast, JNK1/2 induced the phosphorylation of Bad at a novel site of serine 128 after brain ischemia/reperfusion, which inhibited the interaction of PI3K/Akt-induced serine 136-phosphorylated Bad with 14-3-3 proteins, thereby promoted the apoptotic effect of Bad. In addition, activated Akt1 inhibited the activation of Bad(S128) through downregulating JNK1/2 activation, thus inhibiting JNK-mediated Bad apoptosis pathway. Furthermore, the fate of cell to survive or to die was determined by a balance between prosurvival and proapoptotic signals. Taken together, our studies reveal that Bad phosphorylation at two distinct sites induced by Akt1 and JNK1/2 have opposing effects on ischemic brain injury, and present the possibility of Bad as a potential therapeutic target for stroke treatment.


Asunto(s)
Isquemia Encefálica/enzimología , Isquemia Encefálica/patología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Letal Asociada a bcl/metabolismo , Proteínas 14-3-3/metabolismo , Animales , Antracenos/farmacología , Apoptosis/efectos de los fármacos , Isquemia Encefálica/metabolismo , Activación Enzimática/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Hipocampo/patología , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fármacos Neuroprotectores/farmacología , Compuestos Organometálicos/farmacología , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Unión Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteína bcl-X/metabolismo
8.
Life Sci ; 80(22): 2067-75, 2007 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-17459422

RESUMEN

Accumulating evidence suggests that c-Jun N-terminal kinase (JNK) signaling pathway plays a critical role in renal ischemia/reperfusion injury. However, the downstream mechanism that accounts for the proapoptotic actions of JNK during renal ischemia/reperfusion has not been elucidated. We report that SP600125, a potent, cell-permeable, selective, and reversible inhibitor of c-Jun N-terminal kinase (JNK), potently decreased renal epithelial tubular cell apoptosis induced by renal ischemia/reperfusion via suppression of the extrinsic pathway. This corresponds to the decrease in JNK phosphorylation at 20 min and c-Jun phosphorylation (Ser63/73) at 3 h after renal ischemia. Additionally, SP600125 attenuated the increased expression of FasL induced by ischemia/reperfusion at 3 h. The administration of SP600125 prior to ischemia was also protective. Thus, our findings imply that SP600125 can inhibit the activation of the JNK-c-Jun-FasL pathway and protect renal tubular epithelial cells against ischemia/reperfusion-induced apoptosis. Taken together, these results indicate that targeting the JNK pathway provides a promising therapeutic approach for renal ischemia/reperfusion injury.


Asunto(s)
Antracenos/farmacología , Apoptosis/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Enfermedades Renales/tratamiento farmacológico , Daño por Reperfusión/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Riñón/enzimología , Riñón/patología , Enfermedades Renales/enzimología , Enfermedades Renales/patología , Necrosis/metabolismo , Sustancias Protectoras/farmacología , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/enzimología , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
9.
Biochemistry ; 46(13): 4006-16, 2007 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-17348686

RESUMEN

Our previous study indicates that global ischemia facilitates the assembly of the GluR6.PSD-95.MLK3 signaling module, which in turn activated MLK3, leading to exacerbated ischemic neuron death. In addition, JIP1, functioning as a scaffold protein, could couple MLK3-MKK7-JNK to form a specific signaling module and facilitate the activation of the JNK signal pathway. However, the organization, regulation, and function between the two signaling modules and the effects they have on MLK3 activation remain incompletely understood. Here, we show that JIP1 maintains MLK3 in an inactive and monomeric state; once activated, MLK3 binds to PSD-95 and then dimerizes and autophosphorylates. In addition, a GluR6 C-terminus-containing peptide (Tat-GluR6-9c) and antisense oligonucleotides (AS-ODNs) against PSD-95 inhibit the integration of PSD-95 and MLK3 and the dimerization of MLK3, facilitate the interaction of JIP1 and MLK3, and, consequently, perform neuroprotection on neuron death. However, AS-ODNs against JIP1 play a negative role compared to that mentioned above. The findings show that the crosstalk occurs between PSD-95 and the JIP1-mediated signaling module, which may be involved in brain ischemic injury and contribute to the regulation of MLK3 activation. Thus, specific blockade of PSD-95-MLK3 coupling may reduce the extent of ischemia-reperfusion-induced neuronal cell death.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Isquemia Encefálica/fisiopatología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Quinasas Quinasa Quinasa PAM/fisiología , Proteínas de la Membrana/fisiología , Transducción de Señal , Animales , Dimerización , Homólogo 4 de la Proteína Discs Large , Productos del Gen tat/farmacología , Hipocampo/fisiopatología , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , MAP Quinasa Quinasa 7/fisiología , Masculino , Modelos Biológicos , Oligodesoxirribonucleótidos Antisentido/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Ácido Kaínico/fisiología , Daño por Reperfusión/fisiopatología , Transducción de Señal/efectos de los fármacos , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno , Receptor de Ácido Kaínico GluK2
10.
J Neurochem ; 98(1): 170-9, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16805806

RESUMEN

Mitogen-activated protein kinase kinase 4 (MKK4), as an upstream activator of c-Jun NH(2)-terminal kinase (JNK), plays a critical role in response to cellular stresses and pro-inflammatory cytokines. In this study, we investigated the subcellular localization and activation of MKK4 in response to global cerebral ischemia. Our results indicated that MKK4 had two activation peaks in both the cytosol and the nucleus, and translocated from the cytosol to the nucleus at 30 min and 6 h of reperfusion. We also detected the interaction of JNK-interacting protein 3 (JIP3) and MKK4, which reached a maximum at 6 h of reperfusion. To elucidate the mechanism of translocation and activation, we administered N-acetylcysteine, an antioxidant reagent, and a glutamate receptor 6 C-terminus-containing peptide (Tat-GluR6-9c) to rats. The data showed that N-acetylcysteine limited the translocation and activation at 30 min of reperfusion; however, the peptide perturbed the subcellular localization and activation at 6 h of reperfusion, and subsequently provided a protective role against delayed neuronal cell death. Taken together, these results demonstrate that the translocation and activation of MKK4 during early reperfusion are closely associated with reactive oxygen species, whereas, at late reperfusion, MKK4 activation may be involved in brain ischemic injury.


Asunto(s)
MAP Quinasa Quinasa 4/metabolismo , Neuronas/metabolismo , Transporte de Proteínas/fisiología , Daño por Reperfusión/patología , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Antioxidantes/farmacología , Western Blotting/métodos , Cistina/análogos & derivados , Cistina/farmacología , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inmunohistoquímica/métodos , Inmunoprecipitación/métodos , Masculino , Neuronas/efectos de los fármacos , Neuronas/patología , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Factores de Tiempo
11.
Brain Res ; 1092(1): 36-46, 2006 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-16674927

RESUMEN

Our previous studies and the others have strongly suggested that JNK signaling pathway plays a critical role in ischemic brain injury. Here, we reported that SP600125, a potent, cell-permeable, selective, and reversible inhibitor of c-Jun N-terminal kinase (JNK), potently decrease neuronal apoptosis induced by global ischemia/reperfusion in the vulnerable hippocampal CA1 subregion. As a result, SP600125 diminished the increased phosphorylation of c-Jun and the increased expression of FasL induced by ischemia/reperfusion in the vulnerable hippocampal CA1 subregion. At the same time, through inhibiting phosphorylation of Bcl-2 and the release of Bax from Bcl-2/Bax dimers, SP600125 attenuated Bax translocation to mitochondria and the release of cytochrome c induced by ischemia/reperfusion (I/R). Furthermore, the activation of caspase-3 induced by ischemia/reperfusion was also significantly suppressed by preinfusion of SP600125. Importantly, the same neuropotective effect was showed by administration of SP600125 both before and after ischemia. Thus, our findings imply that SP600125 can inhibit the activation of JNK signaling pathway and induce neuroprotection against ischemia/reperfusion in rat hippocampal CA1 region via suppressing the extrinsic and intrinsic pathways of apoptosis. Taken together, these results indicate that targeting the JNK pathway provides a promising therapeutic approach for ischemic brain injury.


Asunto(s)
Antracenos/farmacología , Apoptosis/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Infarto Cerebral/tratamiento farmacológico , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Degeneración Nerviosa/tratamiento farmacológico , Animales , Apoptosis/fisiología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Caspasa 3 , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Infarto Cerebral/metabolismo , Infarto Cerebral/fisiopatología , Citocromos c/metabolismo , Inhibidores Enzimáticos/farmacología , Proteína Ligando Fas , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Glicoproteínas de Membrana/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Degeneración Nerviosa/etiología , Degeneración Nerviosa/prevención & control , Fármacos Neuroprotectores/farmacología , Proteínas Proto-Oncogénicas c-jun/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Necrosis Tumoral/metabolismo , Proteína X Asociada a bcl-2/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
12.
J Biol Chem ; 281(25): 17432-17445, 2006 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-16624817

RESUMEN

Previous studies have suggested that glutamate receptor 6 (GluR6) subunit- and JNK-deficient mice can resist kainate-induced epileptic seizure and neuronal toxicity (Yang, D. D., Kuan, C.-Y., Whitmarsh, A. J., Rinocn, M., Zheng, T. S., Davis, R. J., Rakic, P., and Flavell, R. A. (1997) Nature 389, 865-870; Mulle, C., Seiler, A., Perez-Otano, I., Dickinson-Anson, H., Castillo, P. E., Bureau, I., Maron, C., Gage, F. H., Mann, J. R., Bettler, B., and Heinemmann, S. F. (1998) Nature 392, 601-605). In this study, we show that kainate can enhance the assembly of the GluR6-PSD95-MLK3 module and facilitate the phosphorylation of JNK in rat hippocampal CA1 and CA3/dentate gyrus (DG) subfields. More important, a peptide containing the Tat protein transduction sequence (Tat-GluR6-9c) perturbed the assembly of the GluR6-PSD95-MLK3 signaling module and suppressed the activation of MLK3, MKK7, and JNK. As a result, the inhibition of JNK activation by Tat-GluR6-9c diminished the phosphorylation of the transcription factor c-Jun and down-regulated Fas ligand expression in hippocampal CA1 and CA3/DG regions. The inhibition of JNK activation by Tat-Glur6-9c attenuated Bax translocation, the release of cytochrome c, and the activation of caspase-3 in CA1 and CA3/DG subfields. Furthermore, kainate-induced neuronal loss in hippocampal CA1 and CA3 subregions was prevented by intracerebroventricular injection of Tat-Glur6 - 9c. Taken together, our findings strongly suggest that the GluR6-PSD95-MLK3 signaling module mediates activation of the nuclear and non-nuclear pathways of JNK, which is involved in brain injury induced by kainate. Tat-GluR6-9c, the peptide we constructed, gives new insight into seizure therapy.


Asunto(s)
Núcleo Celular/metabolismo , Hipocampo/metabolismo , Ácido Kaínico/metabolismo , Neuronas/metabolismo , Receptores de Ácido Kaínico/metabolismo , Animales , Diseño de Fármacos , Proteína Ligando Fas , Productos del Gen tat/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Sprague-Dawley , Convulsiones/tratamiento farmacológico , Factores de Necrosis Tumoral/metabolismo , Receptor de Ácido Kaínico GluK2
13.
Brain ; 129(Pt 2): 465-79, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16330502

RESUMEN

It is well documented that N-methyl-D-aspartate and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptors play a pivotal role in ischaemic brain injury. Recent studies have shown that kainate (KA) receptors are involved in neuronal cell death induced by seizure, which is mediated by the GluR6*PSD-95*MLK3 signalling module and subsequent c-Jun N-terminal kinase (JNK) activation. Here we investigate whether GluR6 mediated JNK activation is correlated with ischaemic brain injury. Our results show that cerebral ischaemia followed by reperfusion can enhance the assembly of the GluR6*PSD-95*MLK3 signalling module and JNK activation. As a result, activated JNK can not only phosphorylate the transcription factor c-Jun and up-regulate Fas L expression but can also phosphorylate 14-3-3 and promote Bax translocation to mitochondria, increase the release of cytochrome c and increase caspase-3 activation. These results indicate that GluR6 mediated JNK activation induced by ischaemia/reperfusion ultimately results in neuronal cell death via nuclear and non-nuclear pathways. Furthermore, the peptides we constructed, Tat-GluR6-9c, show a protective role against neuronal death induced by cerebral ischaemia/reperfusion through inhibiting the GluR6 mediated signal pathway. In summary, our results indicate that the KA receptor subunit GluR6 mediated JNK activation is involved in ischaemic brain injury and provides a new approach for stroke therapy.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Hipocampo/metabolismo , Receptores de Ácido Kaínico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Transducción de Señal , Animales , Isquemia Encefálica/metabolismo , Células Cultivadas , Productos del Gen tat/genética , Productos del Gen tat/metabolismo , Inmunohistoquímica/métodos , Etiquetado Corte-Fin in Situ , Quinasas Quinasa Quinasa PAM , Masculino , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Neuronas/metabolismo , Técnicas de Placa-Clamp , Ingeniería de Proteínas , Ratas , Ratas Sprague-Dawley , Receptores de Ácido Kaínico/efectos de los fármacos , Receptores de Ácido Kaínico/genética , Receptores de N-Metil-D-Aspartato/genética , Daño por Reperfusión/metabolismo , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno , Receptor de Ácido Kaínico GluK2
14.
Neurosci Lett ; 393(2-3): 226-30, 2006 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-16243436

RESUMEN

Our previous studies and those of others have strongly suggested that c-Jun N-terminal kinase (JNK) signaling pathway plays a critical role in ischemic brain injury. But the downstream mechanism that accounts for the proapoptotic actions of JNK during cerebral ischemia/reperfusion still remains to be investigated in detail. DP5, one of the mammalian BH3-only proteins, was cloned as a neuronal apoptosis-inducing gene. In this study, we examined the changes of protein level of DP5 and its interaction with Bcl-2 family members in a rat model of global ischemia and reperfusion by immunoprecipitation and immunoblotting; furthermore, we investigated the effect of activated JNK on DP5-signaling pathway. We show here that DP5 was induced and interacted with Bcl-2 but not Bax in hippocampal CA1 6 h to 3 days after ischemia, while the interaction of Bcl-2 with Bax decreased. Systemic administration of SP600125, a small molecule JNK-specific inhibitor, diminished the induction of DP5 and its interaction with Bcl-2 after 2 days of ischemia. At the same time, SP600125 increased the interaction of Bax with Bcl-2 after 2 days of reperfusion. Thus, these results indicate that brain ischemia/reperfusion-induced activation of DP5 signaling pathway is mediated by JNK in postischemic rat hippocampal CA1.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Isquemia Encefálica/metabolismo , Regulación de la Expresión Génica/fisiología , Neuropéptidos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Reperfusión , Proteína X Asociada a bcl-2/metabolismo , Análisis de Varianza , Animales , Antracenos/farmacología , Western Blotting/métodos , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Inmunoprecipitación/métodos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
15.
J Neurosci Res ; 82(5): 642-9, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16267825

RESUMEN

To investigate whether the kainate (KA) receptors subunit GluR6 is involved in the neuronal cell death induced by cerebral ischemia followed by reperfusion, the antisense oligodeoxynucleotides (ODNs) of GluR6 were used to suppress the expression of GluR6 by intracerebroventricular infusion once per day for 3 days before ischemia. Transient brain ischemia was induced by four-vessel occlusion in Sprague-Dawley rats. The effects of GluR6 antisense ODNs on the phosphorylation of MLK3 and JNK and the interactions of MLK3 and PSD-95 with GluR6 were examined by immunoprecipitation and immunoblotting. Our results show that GluR6 antisense ODNs can knock down the expression of GluR6 and suppress the assembly of the GluR6.PSD-95.MLK3 signaling module and, therefore, inhibit JNK activation and phosphoralation of c-jun. On the other hand, the GluR6 antisense ODNs also show a protective role against neuronal cell death induced by cerebral ischemia/reperfusion. Administration of GluR6 antisense ODNs once per day for 3 days before cerebral ischemia significantly decreased neuronal degeneration. In conclusion, our results demonstrate that kainate receptor subunit GluR6 plays an important role in neuronal death induced by cerebral ischemia followed by reperfusion.


Asunto(s)
Isquemia Encefálica/metabolismo , Hipocampo/metabolismo , Degeneración Nerviosa/metabolismo , Oligodesoxirribonucleótidos Antisentido/farmacología , Receptores de Ácido Kaínico/metabolismo , Daño por Reperfusión/metabolismo , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Infarto Cerebral/tratamiento farmacológico , Infarto Cerebral/genética , Infarto Cerebral/metabolismo , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Inyecciones Intraventriculares , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Quinasas Quinasa Quinasa PAM , Masculino , Proteínas de la Membrana/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/genética , Fármacos Neuroprotectores/farmacología , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Ácido Kaínico/efectos de los fármacos , Receptores de Ácido Kaínico/genética , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/genética , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno , Receptor de Ácido Kaínico GluK2
16.
Brain Res ; 1061(1): 57-66, 2005 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-16256962

RESUMEN

Kainate receptor glutamate receptor 6 (GluR6) binds to the postsynaptic density protein 95 (PSD-95), which in turn anchors mixed lineage kinase 3 (MLK3) via SH3 domain in rat brain tissue. MLK3 subsequently activates c-Jun NH(2)-terminal kinase (JNK) via MAP kinase kinases (MKKs). We investigated the association of PSD-95 with GluR6 and MLK3, MLK3 autophosphorylation, the interaction of MLK3 with JNK3, and JNK3 phosphorylation following cerebral ischemia in rat hippocampus. Our results indicate that the GluR6.PSD-95.MLK3 complex peaked at 6 h of reperfusion. Furthermore, MLK3 autophosphorylation and the interaction of MLK3 with JNK3 occurred with the alteration of GluR6.PSD-95.MLK3 signaling module. To further prove whether JNK3 activation in ischemic hippocampus is mediated by GluR6.PSD-95.MLK3 signaling pathway, the AMPA/KA receptor antagonist 6,7-dinitroquinoxaline-2, (1H, 4H)-dione (DNQX), the GluR6 antagonist 6,7,8,9-Tetrahydro-5-nitro-1H-benz[g]indole-2,3-dione-3-oxime (NS102), the AMPA receptor antagonist 1-(4-aminophenyl)-4-methyl-7,8-methylenedioxy-5H-2,3-benzo diazepine (GYKI52466), and the NMDA receptor antagonist ketamine were given to the rats 20 min prior to ischemia. Our findings indicate that both DNQX and NS102 significantly attenuated the association of PSD-95 with GluR6 and MLK3, MLK3 autophosphorylation, interaction of MLK3 with JNK3, and JNK3 phosphorylation, while GYKI52466 and ketamine had no effect. Moreover, administration of NS102 before cerebral ischemia significantly increased the number of the surviving hippocampal CA1 pyramidal cells at 5 days of reperfusion. Consequently, GluR6, one subunit of kainate receptor, plays a critical role in inducing JNK3 activation after ischemic injury.


Asunto(s)
Isquemia Encefálica/enzimología , Hipocampo/enzimología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Receptores de Ácido Kaínico/metabolismo , Análisis de Varianza , Animales , Western Blotting/métodos , Homólogo 4 de la Proteína Discs Large , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Antagonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Inmunoprecipitación/métodos , Quinasas Quinasa Quinasa PAM , Masculino , Fosforilación , Ratas , Reperfusión/métodos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Tiempo , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno , Receptor de Ácido Kaínico GluK2
17.
Neurosci Lett ; 391(1-2): 38-42, 2005 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-16154687

RESUMEN

Mixed lineage kinase-3 (MLK3) is a recently described member of the MLK subfamily of Ser/Thr protein kinases that interacts with mitogen-activated protein kinase (MAPK) pathways. In this study, we investigated the translocation of MLK3 during transient cerebral ischemia in rat hippocampus. Transient brain ischemia was induced by the four-vessel occlusion in Sprague-Dawley rats. Our data show that MLK3 can translocate from cytosolic fraction to the membrane fraction during ischemia and the increased MLK3 in the membrane fraction bind to postsynaptic density protein 95 (PSD-95). The antioxidant N-acetylcysteine (NAC) could inhibit the translocation of MLK3 from cytosolic fraction to the membrane fraction and decrease the interactions of MLK3 and PSD-95 in the membrane fraction. Consequently, these results indicate that reactive oxygen species (ROS) was closely associated with MLK3 translocation induced by transient global ischemia in rat hippocampus.


Asunto(s)
Acetilcisteína/administración & dosificación , Isquemia Encefálica/metabolismo , Membrana Celular/metabolismo , Citosol/metabolismo , Hipocampo/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Animales , Membrana Celular/efectos de los fármacos , Citosol/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Hipocampo/efectos de los fármacos , Masculino , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
18.
Brain Res Mol Brain Res ; 140(1-2): 55-62, 2005 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-16126302

RESUMEN

Recent studies have shown that GluR6 is involved in the modulation of neuronal cell death. It has been shown that PKA can phosphorylate recombinant GluR6 homomeric receptors and that this phosphorylation of GluR6 was suggested to underlie an enhancement of whole-cell current responses. Here, we try to find out whether brain ischemia and reperfusion could induce any change in the serine phosphorylation of GluR6. Our results showed that the serine phosphorylation of GluR6 increased in hippocampus during brain ischemia and early reperfusion period. Then, we used several drugs to investigate the mechanism of modulating the serine phosphorylation of GluR6. KT5720, a specific cell-permeable inhibitor of protein kinase A (PKA), had no effect on the increase in serine phosphorylation of GluR6 induced by brain ischemia or reperfusion. On the other hand, KN-62, a selective inhibitor of rat brain Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), diminished the increase in serine phosphorylation of GluR6. Moreover, our results showed that either MK801 (a NMDA receptor antagonist) or Nifedipine (a L-type Ca2+ channel (L-VGCC) blocker) decreased the increase in serine phosphorylation. In conclusion, our results suggest that CaMKII, activated through NMDA receptors and L-VGCCs, mediated the serine phosphorylation of GluR6 during brain ischemia and early reperfusion period.


Asunto(s)
Isquemia Encefálica/fisiopatología , Canales de Calcio Tipo L/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Hipocampo/fisiología , Fosfoserina/metabolismo , Receptores de Ácido Kaínico/metabolismo , Receptores de N-Metil-D-Aspartato/fisiología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Análisis de Varianza , Animales , Canales de Calcio Tipo L/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Inhibidores Enzimáticos/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/fisiopatología , Masculino , Nifedipino/farmacología , Fosforilación , Ratas , Ratas Sprague-Dawley , Reperfusión , Receptor de Ácido Kaínico GluK2
19.
Brain Res ; 1052(1): 1-9, 2005 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-16018989

RESUMEN

Current studies demonstrated that cell survival is determined by a balance among signaling cascades, including those that recruit the Akt and JNK pathways. In our present work, the relationship between Akt1 and JNK1/2 was evaluated after cerebral ischemia-reperfusion in the hippocampus in a four-vessel occlusion model of Sprague-Dawley rats. This paper was based on our present and previous studies. Firstly, Akt1 had one active peak during reperfusion following 15 min ischemia. Secondly, two peaks of JNK1/2 activation occurred during reperfusion, respectively. Thirdly, the phosphorylation of JNK substrates c-Jun and Bcl-2, and the activation of a key protease of caspase-3 were detected. They only had one active peak, respectively, during reperfusion. To clarify the mechanism of Akt1 activation and further define whether JNK1/2 activation could be regulated by Akt1 through PI3K pathway, LY294002 and insulin were, respectively, administrated to the rats prior to ischemia. Our research indicated that LY294002, a PI3K inhibitor, significantly suppressed Akt1 activation. Furthermore, LY294002 significantly strengthened both peaks of JNK1/2 activation, c-Jun activation, Bcl-2 phosphorylation, and the activation of caspase-3 during reperfusion. In contrast, insulin, a PI3K agonist, not only obviously activated Akt1 during early and later reperfusion, but also inhibited phosphorylation of JNK1/2, c-Jun, and Bcl-2 and attenuated the activation of caspase-3. In addition, pretreatment of insulin significantly increased the number of the surviving CA1 pyramidal cells at 5 days of reperfusion. Consequently, our results indicated that the cross-talk between Akt1 and JNK1/2 could be mediated by insulin receptor through PI3K in rat hippocampus during reperfusion. This signaling pathway might play a neuroprotective role against ischemic insults via inhibition of the JNK pathway, involving the death effector of caspase-3.


Asunto(s)
Isquemia Encefálica/prevención & control , Hipocampo/efectos de los fármacos , Insulina/uso terapéutico , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Análisis de Varianza , Animales , Western Blotting/métodos , Cromonas/farmacología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Masculino , Morfolinas/farmacología , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Sprague-Dawley , Reperfusión/métodos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
20.
Neurosci Lett ; 385(3): 230-3, 2005 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-15970382

RESUMEN

Our previous investigation has shown that postsynaptic density protein 95 (PSD-95) is critical for the Src family kinases-mediated tyrosine phosphorylation of N-methyl-d-aspartate receptor subunit 2A (NR2A) in the postischemic hippocampus. To clarify the roles of PSD-95 in the ischemic brain damage, histological method was performed to examine the effects of PSD-95 antisense oligonucleotides (AS) on the postischemic delayed cell death in rat hippocampus. Transient (15 min) brain ischemia was induced by the four-vessel occlusion method in Sprague-Dawley rats. Five days of reperfusion following brain ischemia (I/R5d) led to hippocampal CA1 pyramidal cell death upward of 90%. Intracerebroventricular infusion of AS (every 24 h for 3 days before ischemia) not only decreased the PSD-95 expression but also increased the number of surviving pyramidal neurons, while missense oligonucleotides (MS) had no effects. To further investigate the mechanisms underlying the neuroprotection of PSD-95 deficiency, the interaction of proline-rich tyrosine kinase 2 (Pyk2) with NR2A as well as autophosphorylation (Tyr402) of Pyk2 were detected. Immunoprecipitation and immunoblot analysis showed that preischemic treatment with AS, but not MS or vehicle, attenuated the I/R6h-induced increases in Pyk2-NR2A association and Pyk2 autophosphorylation. The protein levels of NR2A and Pyk2 had no differences under the above conditions. Our data suggest that the recruitments of ion channels and signaling molecules may be involved in the PSD-95 neurotoxicity in the postischemic hippocampus.


Asunto(s)
Isquemia Encefálica/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Oligonucleótidos Antisentido/administración & dosificación , Células Piramidales/efectos de los fármacos , Animales , Isquemia Encefálica/patología , Muerte Celular/efectos de los fármacos , Homólogo 4 de la Proteína Discs Large , Quinasa 2 de Adhesión Focal , Immunoblotting , Inmunoprecipitación , Inyecciones Intraventriculares , Péptidos y Proteínas de Señalización Intracelular , Masculino , Proteínas de la Membrana , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas Tirosina Quinasas/efectos de los fármacos , Células Piramidales/patología , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Daño por Reperfusión/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA