Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
1.
Mediators Inflamm ; 2017: 2810295, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28626342

RESUMEN

BACKGROUND: Protein tyrosine phosphatases (PTPs) in conjunction with protein tyrosine kinases (PTKs) regulate cellular processes by posttranslational modifications of signal transduction proteins. PTP nonreceptor type 1B (PTP-1B) is an enzyme of the PTP family. We have previously shown that hypoxia induces an increase in activation of a class of nonreceptor PTK, the Src kinases. In the present study, we investigated the changes that occur in the expression of PTP-1B in the cytosolic component of the brain of newborn piglets acutely after hypoxia as well as long term for up to 2 weeks. METHODS: Newborn piglets were divided into groups: normoxia, hypoxia, hypoxia followed by 1 day and 15 days in FiO2 0.21, and hypoxia pretreated with Src kinase inhibitor PP2, prior to hypoxia followed by 1 day and 15 days. Hypoxia was achieved by providing 7% FiO2 for 1 hour and PTP-1B expression was measured via immunoblotting. RESULTS: PTP-1B increased posthypoxia by about 30% and persisted for 2 weeks while Src kinase inhibition attenuated the expected PTP-1B-increased expression. CONCLUSIONS: Our study suggests that Src kinase mediates a hypoxia-induced increased PTP-1B expression.


Asunto(s)
Hipoxia Encefálica/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Familia-src Quinasas/metabolismo , Animales , Animales Recién Nacidos , Citosol/enzimología , Modelos Animales de Enfermedad , Porcinos
2.
Rev Neurosci ; 26(1): 105-17, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25720056

RESUMEN

Abstract Hypoxic or ischemic stress causes many serious brain injuries, including stroke and neonatal hypoxia ischemia encephalopathy. During brain hypoxia ischemia processes, nitric oxide (NO) may play either a neurotoxic or a neuroprotective role, depending upon factors such as the NO synthase (NOS) isoform, the cell type by which NO is produced, and the temporal stage after the onset of the hypoxic ischemic brain injury. Excessive NO production can be neurotoxic, leading to cascade reactions of excitotoxicity, inflammation, apoptosis, and deteriorating primary brain injury. In contrast, NO produced by endothelial NOS plays a neuroprotective role by maintaining cerebral blood flow and preventing neuronal injury, as well as inhibiting platelet and leukocyte adhesion. Sometimes, NO-derived inducible NOS and neuronal NOS in special areas may also play neuroprotective roles. Therefore, this review summarizes the different roles and the regulation of the three NOS isoforms in hypoxic or ischemic brain injury as revealed in research in recent years, focusing on the neurotoxic role of the three NOS isoforms involved in mechanisms of hypoxic or ischemic brain injury.


Asunto(s)
Isquemia Encefálica/enzimología , Hipoxia Encefálica/enzimología , Óxido Nítrico Sintasa/metabolismo , Animales , Isquemia Encefálica/patología , Humanos , Hipoxia Encefálica/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo
3.
Neurochem Res ; 40(11): 2270-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26342830

RESUMEN

The Src family kinases are a family of intracellular, non-receptor tyrosine kinases that are involved in a variety of cellular functions including the regulation of inflammation and apoptosis after brain hypoxia. Caspase-1 (C1) activates IL-1ß through the formation of complex structures, the inflammasomes, while caspase-8 (C8) is part of the extrinsic apoptotic pathway. C8 has been found to directly activate the production of IL-1ß. Previously, we observed that C1 and IL-1ß are increased in the acute phase after hypoxia in the brain of piglets, but they follow a different pattern long term, with C1 remaining activated throughout the period of observation, while IL-1ß returning to baseline at 15 days. Src kinase inhibition ameliorated the activation of C1 and IL-1ß early, but did not appear to have any effect long term. Prompted by these findings, we assessed the changes that occur over time (1 h and 15 days) in C1 and C8 activities after brain hypoxia as well as the effect of pretreatment with a Src kinase inhibitor, PP2 on these biochemical markers. Enzymatic activities were determined by spectrophotometry with measurements of C1 and C8 in each cytosolic brain sample (N = 4 in each group). We found that C1 and C8 activities increase in the acute phase following hypoxia in the brain of newborn piglets, with C8 relatively more than C1 (C8/C1 ratio increased from 2:1 as baseline to 3:1 in hypoxia). Fifteen days after hypoxia C8/C1 ratio decreased to about 1:1. In piglets that were pretreated with a Src kinase selective inhibitor (PP2) and then subjected to hypoxia, the C8/C1 ratio early increase was not observed. Immediately after hypoxia C8 and C1 follow a similar pattern of increase while long term this appears to dissociate. We propose that following this experimental methodology, the previously observed IL-1ß production after hypoxia might be associated with C8 rather than C1 and that Src kinase is involved in the above process.


Asunto(s)
Caspasa 8/metabolismo , Hipoxia Encefálica/enzimología , Familia-src Quinasas/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Caspasa 1/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Interleucina-1beta/biosíntesis , Fosfocreatina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Sus scrofa
4.
Biochem J ; 458(2): 203-11, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24328859

RESUMEN

Increased catalytic activity of CBS (cystathionine ß-synthase) was recently shown to mediate vasodilation of the cerebral microcirculation, which is initiated within minutes of the onset of acute hypoxia. To test whether chronic hypoxia was a stimulus for increased CBS expression, U87-MG human glioblastoma and PC12 rat phaeochromocytoma cells were exposed to 1% or 20% O2 for 24-72 h. CBS mRNA and protein expression were increased in hypoxic cells. Hypoxic induction of CBS expression was abrogated in cells transfected with vector encoding shRNA targeting HIF (hypoxia-inducible factor) 1α or 2α. Exposure of rats to hypobaric hypoxia (0.35 atm; 1 atm=101.325 kPa) for 3 days induced increased CBS mRNA, protein and catalytic activity in the cerebral cortex and cerebellum, which was blocked by administration of the HIF inhibitor digoxin. HIF-binding sites, located 0.8 and 1.2 kb 5' to the transcription start site of the human CBS and rat Cbs genes respectively, were identified by ChIP assays. A 49-bp human sequence, which encompassed an inverted repeat of the core HIF-binding site, functioned as a hypoxia-response element in luciferase reporter transcription assays. Thus HIFs mediate tissue-specific CBS expression, which may augment cerebral vasodilation as an adaptive response to chronic hypoxia.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Cistationina betasintasa/biosíntesis , Regulación Enzimológica de la Expresión Génica , Hipoxia Encefálica/enzimología , Animales , Encéfalo/irrigación sanguínea , Células Cultivadas , Cistationina betasintasa/genética , Células HEK293 , Humanos , Hipoxia Encefálica/genética , Hipoxia Encefálica/patología , Factor 1 Inducible por Hipoxia/fisiología , Masculino , Células PC12 , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Distribución Tisular/genética , Vasodilatación/genética
5.
Zh Evol Biokhim Fiziol ; 51(6): 427-30, 2015.
Artículo en Ruso | MEDLINE | ID: mdl-26983277

RESUMEN

Analysis of the effect of a caspase-3 inhibitor on the content of the amyloid-degrading neuropeptidase neprilysin (NEP) in the cortex of rats subjected to prenatal hypoxia (7% O2, 3 h) on the 14-th day of the embryonic development (E14) was performed. It was found that rats subjected to prenatal hypoxia on days 20-30 after birth have an increased content and activity of caspase-3 with reduced levels of NEP and of the C-terminal fragment of the amyloid precursor protein (AICD) regulating NEP expression. In hypoxic animals 3 days after a single injection of a caspase inhibitor (i. v., Ac-DEVD-CHO, P20) the content of AICD and NEP was found to be increased up to the levels observed in control rats. The data obtained suggest that the increase of caspase-3 enzyme activity could affect NEP expression via proteolytic degradation of its transcription factor AICD. These data for the first time demonstrate the role of caspases in AICD-dependent regulation of NEP production in the brain of mammals under hypoxic conditions.


Asunto(s)
Caspasa 3/biosíntesis , Corteza Cerebral/enzimología , Regulación Enzimológica de la Expresión Génica , Hipoxia Encefálica/enzimología , Neprilisina/biosíntesis , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Corteza Cerebral/patología , Femenino , Hipoxia Encefálica/patología , Ratas , Ratas Wistar
6.
Patol Fiziol Eksp Ter ; (3): 11-9, 2012.
Artículo en Ruso | MEDLINE | ID: mdl-23072108

RESUMEN

Group I of metabotropic glutamate receptors (ImGluRs) are a family of G-protein-coupled receptors which activate a multitude of signaling pathways important for modulating neuronal excitability and synaptic plasticity as well as anti- and prosurvival pathways initiated by hypoxia. However these functions are still not complete and sometimes controversial. The present work is a review of data concerning involvement of ImGluRs in mechanisms of cell response to hypoxia. We also present original data demonstrating their participation in forming pathogenic and adaptogenic intracellular events, appearing in rat neocortex during a day after severe or moderate hypobaric hypoxia, respectively. Ca2+ responses to ImGluRs stimulation in survival cortical slices and expression of ImGluRs, IP3Rs and PLCbeta1 in immunolabelled cortical preparations were estimated for these two different hypoxic models.


Asunto(s)
Encéfalo/metabolismo , Hipoxia Encefálica/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Transducción de Señal , Adaptación Fisiológica , Animales , Encéfalo/enzimología , Encéfalo/patología , Encéfalo/fisiopatología , Señalización del Calcio , Humanos , Hipoxia Encefálica/enzimología , Hipoxia Encefálica/patología , Hipoxia Encefálica/fisiopatología , Inmunohistoquímica , Fosfatos de Inositol/metabolismo , Neuronas/enzimología , Neuronas/metabolismo , Neuronas/patología , Fosfolipasas/metabolismo , Proteína Quinasa C/metabolismo
7.
Int J Neurosci ; 121(5): 279-88, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21348795

RESUMEN

High altitude (HA) generates a deleterious effect known as hypobaric hypoxia (HBH). This causes severe physiological and psychological changes such as acute mountain sickness (AMS) and cognitive functions in terms of learning and memory. The present study has evaluated the effect of cholinesterase inhibitors on memory consolidation following HBH. Adult male Sprague Dawley rats (80-90 days old) with an average body weight of 250 ± 25 g were used. Rats were assessed memory consolidation by using Morris water maze (MWM) for 8 days. After assessment of memory consolidation, rats were then exposed to HBH in stimulated chamber for 7 days at 6,100 m. After exposure to HBH, the memory consolidation of rats has been assessed in MWM. The results showed that there was memory consolidation impairment in HBH-exposed rats as compared to normoxic rats in terms of time spent in quaradents, rings, and counters. The rats which have been treated with physostigmine (PHY) and galantamine (GAL) showed better time spent in quaradents, rings, and counters as compared with hypoxic rats. In conclusion, the cholinesterase inhibitors could ameliorate the impairment of memory consolidation following HBH.


Asunto(s)
Mal de Altura/tratamiento farmacológico , Inhibidores de la Colinesterasa/farmacología , Hipoxia Encefálica/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Memoria/efectos de los fármacos , Acetilcolina/agonistas , Acetilcolina/fisiología , Presión del Aire , Mal de Altura/complicaciones , Mal de Altura/enzimología , Animales , Modelos Animales de Enfermedad , Galantamina/farmacología , Hipoxia Encefálica/complicaciones , Hipoxia Encefálica/enzimología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/etiología , Fisostigmina/farmacología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Resultado del Tratamiento
8.
Mil Med Res ; 8(1): 62, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34857032

RESUMEN

BACKGROUND: Disruption of the blood-brain barrier (BBB) after a stroke can lead to brain injury and neurological impairment. Previous work confirmed the involvement of the immunoproteasome subunit of low molecular mass peptide 2 (LMP2) in the pathophysiology of ischemia stroke. However, the relationship between the immunoproteasome LMP2 and the BBB remains unclear. METHODS: Adult male Sprague-Dawley rats were subjected to transient middle cerebral artery occlusion/reperfusion (MCAO/R). Three days before MCAO, the rats were treated with lentivirus-mediated LMP2 shRNA preparations by stereotactical injection into the ipsilateral hemispheric region. The rat brain microvascular endothelial cell (RBMVEC) line was exposed to oxygen-glucose deprivation/reperfusion (OGD/R) to mimic ischemic conditions in vitro. The RNA interference-mediated knockdown of LMP2 or ß-catenin was analysed in vivo and in vitro. Analysis of the quantity of extravasated Evans blue (EB) and cerebral fluorescent angiography were performed to evaluate the integrity of the BBB. Immunofluorescence and Western blotting were employed to detect the expression of target proteins. Cell migration was evaluated using a scratch migration assay. The results of immunofluorescence, Western blotting and cell migration were quantified using the software ImageJ (Version 1.53m). Parametric data from different groups were compared using one-way ANOVA followed by the least significant difference (LSD) test. RESULTS: Cerebral ischemia led to lower levels of structural components of the BBB such as tight junction proteins (occludin, claudin-1 and ZO-1) in the MCAO/R group compared with the sham group (P < 0.001). However, inhibition of the immunoproteasome LMP2 restored the expression of these proteins, resulting in higher levels of occludin, claudin-1 and ZO-1 in the LMP2-shRNA group compared with the control-shRNA group (P < 0.001). In addition, inhibition of the immunoproteasome LMP2 contributed to higher microvascular density and decreased BBB permeability [e.g., the quantity of extravasated EB: LMP2-shRNA group (58.54 ± 7.37) µg/g vs. control-shRNA group (103.74 ± 4.32) µg/g, P < 0.001], and promoted the upregulation of Wnt-3a and ß-catenin proteins in rats following MCAO/R. In vitro experiments, OGD/R induced marked upregulation of LMP2, proapoptotic protein Bax and cleaved caspase-3, and downregulation of occludin, claudin-1, ZO-1 and Bcl-2, as well as inhibition of the Wnt/ß-catenin pathway Wnt-3a and ß-catenin proteins in RBMVECs, compared with the control group under normal culture conditions (P < 0.001). However, silencing of LMP2 gene expression reversed these protein changes and promoted proliferation and migration of RBMVECs following OGD/R. Silencing of ß-catenin by transfection of RBMVECs with ß-catenin-siRNA aggravated the downregulation of tight junction proteins, and reduced the proliferation and migration of RBMVECs following OGD/R, compared with the control-siRNA group (P < 0.001). LMP2-siRNA and ß-catenin-siRNA co-transfection partly counteracted the beneficial effects of silencing LMP2-siRNA on the levels of tight junction proteins in RBMVECs exposed to OGD/R. CONCLUSION: This study suggests that inhibition of the immunoproteasome LMP2 ameliorates ischemia/hypoxia-induced BBB injury, and that the molecular mechanism involves the immunoproteasome-regulated activation of the Wnt/ß-catenin signalling pathway under ischemic conditions.


Asunto(s)
Barrera Hematoencefálica , Cisteína Endopeptidasas , Hipoxia Encefálica , Complejo de la Endopetidasa Proteasomal , Vía de Señalización Wnt , beta Catenina , Animales , Barrera Hematoencefálica/metabolismo , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Hipoxia Encefálica/enzimología , Hipoxia Encefálica/genética , Masculino , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Transfección , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
9.
J Neurosci ; 29(15): 4903-10, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19369559

RESUMEN

Respiratory motoneuron response to hypoxia is reflex in nature and carotid body sensory receptor constitutes the afferent limb of this reflex. Recent studies showed that repetitive exposures to hypoxia evokes long term facilitation of sensory nerve discharge (sLTF) of the carotid body in rodents exposed to chronic intermittent hypoxia (CIH). Although studies with anti-oxidants suggested the involvement of reactive oxygen species (ROS)-mediated signaling in eliciting sLTF, the source of and the mechanisms associated with ROS generation have not yet been investigated. We tested the hypothesis that ROS generated by NADPH oxidase (NOX) mediate CIH-evoked sLTF. Experiments were performed on ex vivo carotid bodies from rats and mice exposed either to 10 d of CIH or normoxia. Acute repetitive hypoxia evoked a approximately 12-fold increase in NOX activity in CIH but not in control carotid bodies, and this effect was associated with upregulation of NOX2 mRNA and protein, which was primarily localized to glomus cells of the carotid body. sLTF was prevented by NOX inhibitors and was absent in mice deficient in NOX2. NOX activation by CIH required 5-HT release and activation of 5-HT(2) receptors coupled to PKC signaling. Studies with ROS scavengers revealed that H(2)O(2) generated from O(2).(-) contributes to sLTF. Priming with H(2)O(2) elicited sLTF of carotid bodies from normoxic control rats and mice, similar to that seen in CIH-treated animals. These observations reveal a novel role for NOX-induced ROS signaling in mediating sensory plasticity of the carotid body.


Asunto(s)
Cuerpo Carotídeo/enzimología , Hipoxia Encefálica/enzimología , NADPH Oxidasas/fisiología , Plasticidad Neuronal/fisiología , Animales , Cuerpo Carotídeo/metabolismo , Enfermedad Crónica , Hipoxia Encefálica/metabolismo , Hipoxia Encefálica/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vías Nerviosas/enzimología , Vías Nerviosas/metabolismo , Proteína Quinasa C/fisiología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptores de Serotonina 5-HT2/fisiología , Serotonina/fisiología
10.
Med Hypotheses ; 134: 109419, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31622925

RESUMEN

To remedy carotid artery stenosis and prevent stroke surgical intervention is commonly used, and the gold standard being carotid endarterectomy (CEA). During CEA cerebrovascular hemoglobin oxygen saturation decreases and when this decrease reaches critical levels it leads to cerebral hypoxia that causes neuronal damage. One of the proposed mechanism that affects changes during CEA and contribute to acute brain ischemia (ABI) is oxidative stress. The increased production of reactive oxygen species and reactive nitrogen species during ABI may cause an unregulated inflammatory response and further lead to structural and functional injury of neurons. Antioxidant activity are involved in the protection against neuronal damage after cerebral ischemia. We hypothesized that neuronal injury and poor outcomes in patients undergoing CEA may be results of oxidative stress that disturbed function of antioxidant enzymes and contributed to the DNA damage in lymphocytes.


Asunto(s)
Isquemia Encefálica/enzimología , Catalasa/biosíntesis , Endarterectomía Carotidea/efectos adversos , Hipoxia Encefálica/enzimología , Complicaciones Intraoperatorias/enzimología , Linfocitos/enzimología , Superóxido Dismutasa-1/biosíntesis , Superóxido Dismutasa/biosíntesis , Isquemia Encefálica/etiología , Estenosis Carotídea/enzimología , Estenosis Carotídea/cirugía , Catalasa/sangre , Catalasa/genética , Daño del ADN , Radicales Libres , Regulación Enzimológica de la Expresión Génica , Humanos , Hipoxia Encefálica/etiología , Complicaciones Intraoperatorias/etiología , Mitocondrias/metabolismo , Modelos Biológicos , Estrés Oxidativo , Daño por Reperfusión/enzimología , Daño por Reperfusión/etiología , Superóxido Dismutasa/sangre , Superóxido Dismutasa/genética , Superóxido Dismutasa-1/sangre , Superóxido Dismutasa-1/genética
11.
Anesth Analg ; 108(2): 601-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19151295

RESUMEN

BACKGROUND: The neuroprotective efficacy of anesthetics observed in experimental models remains unproven in the clinical setting. The nonreceptor tyrosine kinase focal adhesion kinase (FAK) has been suggested to be involved in the neuroprotective effect of anesthetics observed experimentally. In the present work, we investigated whether FAK and the duration of ischemia play a role in the preconditioning effect of sevoflurane on brain tissue. METHODS: Rat acute hippocampal slices were subjected to oxygen and glucose deprivation (OGD) challenge during increasing periods of time (10, 20, 30, 45, 50, and 60 min) followed by 1 h reperfusion. A preconditioning sevoflurane concentration (10(-4) M, 1 h) was applied 3 h before initiation of OGD. Protein expression of FAK and cleaved caspase 3 (a marker of activation of the apoptotic cascade) was measured by immunoblotting. Cell death was assessed by propidium iodide (PI) fluorescence. RESULTS: Both PI fluorescence and expression of cleaved caspase 3 significantly increased with duration of ischemia until reaching a ceiling effect for durations of ischemia longer than 30 min. Sevoflurane (10(-4) M) increased FAK expression and markedly reduced the increase in PI fluorescence and cleaved caspase 3 expression for periods of ischemia of 10, 20, and 30 min. In contrast, the protective effect was no longer observed for periods of ischemia longer than 30 min. 4-amino-5-(4-chlorophenyl)-7-(t-butyl) pyrazolo[3,4-d] pyrimidine (PP2, 10(-5) M, an inhibitor of src tyrosine kinases) application 60 min before and throughout that of sevoflurane significantly reduced the neuroprotective effect of sevoflurane on both caspase 3 expression and PI fluorescence. CONCLUSION: In the OGD rat acute hippocampal slice, the preconditioning effect of a clinically relevant concentration of sevoflurane was very likely to involve FAK and was observed only for periods of ischemia

Asunto(s)
Anestésicos por Inhalación/farmacología , Isquemia Encefálica/fisiopatología , Proteína-Tirosina Quinasas de Adhesión Focal/fisiología , Glucosa/deficiencia , Hipocampo/fisiopatología , Hipoxia Encefálica/fisiopatología , Éteres Metílicos/farmacología , Actinas/metabolismo , Animales , Análisis de los Gases de la Sangre , Isquemia Encefálica/enzimología , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Hipocampo/irrigación sanguínea , Hipocampo/enzimología , Concentración de Iones de Hidrógeno , Hipoxia Encefálica/enzimología , Masculino , Fármacos Neuroprotectores , Fosforilación , Ratas , Ratas Sprague-Dawley , Sevoflurano , Transducción de Señal/fisiología
12.
Neurosci Lett ; 438(1): 38-41, 2008 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-18468794

RESUMEN

Previous studies have shown that cerebral hypoxia results in increased activity of caspase-9, a key initiator of programmed cell death, in the cytosolic fractions of the cerebral cortex of newborn piglets. The present study tests the hypothesis that hypoxia results in increased expression of procaspase-9 and procaspase-3 in neuronal nuclear, mitochondrial and cytosolic fractions of the cerebral cortex of newborn piglets. To test this hypothesis, expression of procaspase-9 and procaspase-3 was determined in 10 newborn piglets divided into two groups: normoxic (Nx, n=5) and hypoxic (Hx, n=5). The hypoxic piglets were exposed to an FiO(2) of 0.06 for 1h. Tissue hypoxia was documented by ATP and phosphocreatinine (PCr) levels. Neuronal nuclear, mitochondrial and cytosolic fractions were isolated and the expression of procaspase-9 and procaspase-3 was determined by immunoblotting using specific anti-procaspase-9 and anti-procaspase-3 antibodies. ATP levels (micromol/g brain) were 4.34+/-0.36 in the Nx and 1.43+/-0.28 in the Hx (p<0.001 vs. Nx) groups. PCr levels (micromol/g brain) were 3.75+/-0.27 in the Nx and 0.69+/-0.26 in the Hx (p<0.001 vs. Nx) group. Cytosolic procaspase-9 density (ODxmm(2)) was 88.82+/-17.55 in the Nx and 215.54+/-22.77 in the Hx (p<0.001 vs. Nx). Mitochondrial procaspase-9 density (ODxmm(2)) was 104.67+/-12.75 in the Nx and 183.44+/-16.69 in the Hx (p<0.001 vs. Nx). Nuclear procaspase-9 density (ODxmm(2)) was 135.56+/-15.36 in the Nx and 190.66+/-29.35 in the Hx (p<0.001 vs. Nx). Cytosolic procaspase-3 density (ODxmm(2)) was 23.72+/-3.71 in the Nx and 92.44+/-8.46 in the Hx (p<0.001 vs. Nx). Mitochondrial procaspase-3 density (ODxmm(2)) was 22.12+/-2.97 in the Nx and 51.22+/-10.67 in the Hx (p<0.001 vs. Nx). Nuclear procaspase-3 density (ODxmm(2)) was 53.80+/-7.18 in the Nx and 84.67+/-5.63 in the Hx (p<0.001 vs. Nx). We conclude that procaspase-9 and procaspase-3 proteins increased in all cell compartments including cytosolic, mitochondrial and nuclear during hypoxia, indicating increased expression of procaspase-9 during hypoxia. We propose that following increased expression of procaspase-9 and procaspase-3, these molecules traffic among the various cell compartments and become available for their activation resulting in increased caspase-9 and caspase-3 activity.


Asunto(s)
Caspasa 3/metabolismo , Caspasa 9/metabolismo , Corteza Cerebral/enzimología , Hipoxia Encefálica/enzimología , Neuronas/enzimología , Adenosina Trifosfato/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Compartimento Celular/fisiología , Núcleo Celular/enzimología , Corteza Cerebral/fisiopatología , Citosol/enzimología , Modelos Animales de Enfermedad , Metabolismo Energético/fisiología , Activación Enzimática/fisiología , Hipoxia Encefálica/fisiopatología , Mitocondrias/enzimología , Fosfocreatina/metabolismo , Fosforilación , Fracciones Subcelulares/metabolismo , Sus scrofa , Regulación hacia Arriba/fisiología
13.
J Cereb Blood Flow Metab ; 27(5): 998-1009, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17033691

RESUMEN

Rho-kinase is a serine threonine kinase that increases vasomotor tone via its effects on both endothelium and smooth muscle. Rho-kinase inhibition reduces cerebral infarct size in wild type, but not endothelial nitric oxide synthase deficient (eNOS-/-) mice. The mechanism may be related to Rho-kinase activation under hypoxic/ischemic conditions and impaired vasodilation because of downregulation of eNOS activity. To further implicate Rho-kinase in impaired vascular relaxation during hypoxia/ischemia, we exposed isolated vessels from rat and mouse to 60 mins of hypoxia, and showed that hypoxia reversibly abolished acetylcholine-induced eNOS-dependent relaxation, and that Rho-kinase inhibitor hydroxyfasudil partially preserved this relaxation during hypoxia. We, therefore, hypothesized that if hypoxia-induced Rho-kinase activation acutely impairs vasodilation in ischemic cortex, in vivo, then Rho-kinase inhibitors would acutely augment cerebral blood flow (CBF) as a mechanism by which they reduce infarct size. To test this, we studied the acute cerebral hemodynamic effects of Rho-kinase inhibitors in ischemic core and penumbra during distal middle cerebral artery occlusion (dMCAO) in wild-type and eNOS-/- mice using laser speckle flowmetry. When administered 60 mins before or immediately after dMCAO, Rho-kinase inhibitors hydroxyfasudil and Y-27632 reduced the area of severely ischemic cortex. However, hydroxyfasudil did not reduce the area of CBF deficit in eNOS-/- mice, suggesting that its effect on CBF within the ischemic cortex is primarily endothelium-dependent, and not mediated by its direct vasodilator effect on vascular smooth muscle. Our results suggest that Rho-kinase negatively regulates eNOS activity in acutely ischemic brain, thereby worsening the CBF deficit. Therefore, rapid nontranscriptional upregulation of eNOS activity by small molecule inhibitors of Rho-kinase may be a viable therapeutic approach in acute stroke.


Asunto(s)
Isquemia Encefálica/fisiopatología , Circulación Cerebrovascular/efectos de los fármacos , Endotelio Vascular/fisiología , Inhibidores Enzimáticos/farmacología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Acetilcolina/farmacología , Animales , Isquemia Encefálica/tratamiento farmacológico , Regulación hacia Abajo/efectos de los fármacos , Hipoxia Encefálica/enzimología , Hipoxia Encefálica/metabolismo , Infarto de la Arteria Cerebral Media/patología , Flujometría por Láser-Doppler , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas , Estimulación Química , Vasodilatadores/farmacología , Quinasas Asociadas a rho
14.
Neurosci Lett ; 423(3): 219-24, 2007 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-17709198

RESUMEN

Accumulated studies have suggested that mitogen-activated protein kinase (MAPK) play a pivotal role in the development of cerebral hypoxic preconditioning (HPC). By using our "auto-hypoxia"-induced HPC mouse model, we have reported increased phosphorylation level of p38 MAPK, and decreased phosphorylation and protein expression levels of extracellular signal regulated kinases 1/2 (ERK1/2) in the brain of HPC mice. In the current study, we investigated the involvement of c-Jun N-terminal kinase (JNK) in the brain of HPC mice. By using Western blot analysis, we found that the phosphorylation levels of JNK at Thr183 and Tyr185 sites (phospho-Thr183/Tyr185 JNK), but not its protein expression, increased significantly (p<0.05, n=6 for each group) both in the hippocampus and frontal cortex of early (H1-H4) and delayed (H5 and H6) HPC mice than that of the normoxic group (H0, n=6). Similarly, enhanced phospho-Thr183/Tyr185 JNK was also observed by immunostaining in the hippocampus and frontal cortex of mice following series of hypoxic exposures (H3 and H6). In addition, we found that phospho-Thr183/Tyr185 JNK predominantly co-localized with a neuron-specific protein, neurogranin, in both the hippocampus and frontal cortex of HPC mice (H3) by using double-labeled immunofluorescence. These results suggest that the increased neuron-specific phosphorylation of JNK at Thr183/Tyr185, not protein expression, might be involved in the development of cerebral HPC of mice.


Asunto(s)
Encéfalo/enzimología , Hipoxia Encefálica/enzimología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas/enzimología , Animales , Encéfalo/metabolismo , Lóbulo Frontal/enzimología , Hipocampo/enzimología , Hipoxia Encefálica/metabolismo , Técnicas Inmunológicas , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Neurogranina/metabolismo , Fosforilación , Coloración y Etiquetado , Treonina , Distribución Tisular , Tirosina
15.
Anesth Analg ; 105(3): 739-43, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17717233

RESUMEN

BACKGROUND: Cognitive changes associated with moderate hypoxia in rodents may result from the diminished functioning of central cholinergic neurotransmission. We designed this study to examine whether treatment with physostigmine (PHY), an acetylcholinesterase inhibitor, could improve the impairment of working memory after hypoxic hypoxia. METHODS: We randomized 90 Swiss Webster, 30-35 g mice (6-8 wks) to three hypoxia groups at fraction of inspired oxygen, FiO2 = 0.10 (1. no treatment; 2. PHY 0.1 mg/kg intraperitoneally administered immediately before; or 3. after hypoxia), or to two room air groups (given either no treatment or PHY after an insult). An object recognition test was used to assess short-term memory function. The object recognition test exploits the tendency of mice to prefer exploring novel objects in an environment when a familiar object is also present. During the 15 min training trial, two identical objects were placed in two defined sites of the box. During the test trial performed 1 h later, one of the objects was replaced by a new object with a different shape. The time spent exploring the two objects was automatically recorded by a video camera and associated software. The performance was analyzed with ANOVA, followed by post hoc comparisons using the Newman-Keuls test when appropriate. P values <0.05 were considered significant. RESULTS: Untreated mice subjected to hypoxia at Fio2 = 0.1 spent significantly less time exploring a novel object on testing day 1 than did untreated mice breathing room air. Performance of the mice subjected to hypoxia, who received physostigmine after, but not before, the insult did not differ from the control group. CONCLUSION: Moderate hypoxia impairs rodents' performance in a working memory task. It appears that changes are transient, because the cognitive functioning of the mice returned to the baseline level 7 days after treatment. Postinsult administration of PHY prevented deterioration of cognitive function. An increased level of acetylcholine in the central nervous system may be responsible for the improved performance of the hypoxia-treated mice.


Asunto(s)
Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Inhibidores de la Colinesterasa/farmacología , Trastornos del Conocimiento/prevención & control , Cognición/efectos de los fármacos , Hipoxia Encefálica/tratamiento farmacológico , Nootrópicos/farmacología , Fisostigmina/farmacología , Acetilcolina/metabolismo , Acetilcolinesterasa/metabolismo , Animales , Encéfalo/enzimología , Inhibidores de la Colinesterasa/uso terapéutico , Trastornos del Conocimiento/enzimología , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Hipoxia Encefálica/enzimología , Hipoxia Encefálica/psicología , Masculino , Memoria/efectos de los fármacos , Ratones , Nootrópicos/uso terapéutico , Fisostigmina/uso terapéutico , Índice de Severidad de la Enfermedad , Factores de Tiempo
16.
Mol Med Rep ; 15(5): 2727-2731, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28447745

RESUMEN

The present study aimed to investigate the association between myocardial cell apoptosis and calpain-1/caspase-3 expression in a rat model of hypoxic-ischemic brain damage (HIBD). A total of 64 newborn rats were divided into control (n=8; sacrificed on day 7) and HIBD groups (n=56). HIBD group rats were sacrificed 2, 12 or 24 h, or 2, 3, 5 or 7 days following HIBD (n=8/group). A terminal deoxynucleotidyl transferase dUTP nick-end labeling assay was performed to detect myocardial apoptotic cells and calculate the apoptosis index (AI), reverse transcription-polymerase chain reaction was performed to detect myocardial calpain-1/caspase-3 mRNA expression levels and a western blot analysis was conducted to detect calpain­1 protein expression levels. The correlations between calpain­1 and caspase­3 expression levels and AI were analyzed. The results demonstrated that apoptotic myocardial cells in the HIBD groups were markedly increased compared with the control group, with AI peaking in the day 3 group. Caspase­3 and calpain­1 mRNA expression levels were increased from 2 and 12 h following HIBD, respectively, with the most elevated levels in the day 2 group. Compared with the control group, calpain­1 protein expression levels were increased from 2 h, with the greatest expression levels in the day 3 group (P<0.05). Calpain­1 mRNA and protein (76/80 kDa) expression levels demonstrated positive linear correlations with AI (r=0.786, P=0.001; and r=0.853, P=0.001, respectively) Caspase-3 mRNA expression levels were positively correlated with AI (r=0.894; P=0.001). In conclusion, the present study demonstrated that in rats with HIBD, there is a positive correlation between increased apoptosis of myocardial cells and expression levels of calpain-1 and caspase-3.


Asunto(s)
Apoptosis , Lesiones Encefálicas/enzimología , Calpaína/biosíntesis , Caspasa 3/biosíntesis , Regulación Enzimológica de la Expresión Génica , Hipoxia Encefálica/enzimología , Miocardio/enzimología , Animales , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Femenino , Hipoxia Encefálica/patología , Masculino , Miocardio/patología , Ratas , Ratas Wistar
17.
Neuroreport ; 28(12): 770-773, 2017 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-28658050

RESUMEN

Caspase-2 has features of both initiator and effector caspases. Previously, we have shown that brain hypoxia-induced production of caspases 1, 3, 8, and 9 is Src kinase mediated, a nonreceptor intracellular family of kinases. The present study tests the hypothesis that hypoxia results in increased expression of caspase-2 and this effect is mediated by Src kinase. Two to three days old newborn piglets were subjected to normoxia, hypoxia (Hx, FiO2 7%), and Src kinase inhibition (using PP2, 1 mg/kg, intravenous), followed by 30 min of acute hypoxia (Hx+PP2). ATP and phosphocreatine were determined biochemically to verify energy molecule depletion in the hypoxic groups. The cytosolic brain function was isolated and a western blot analysis was carried out using an antibody specific for the caspase-2. The immune-complex band density was expressed as OD/mm. Caspase-2 expression was increased two-fold in the Hx group. After Src kinase inhibition followed by hypoxia, caspase-2 expression was similar to normoxia levels. We conclude that hypoxia results in increased expression of caspase-2 protein in the cytosolic fraction of the cerebral cortex of the newborn piglets. This increase is mediated by Src kinase.


Asunto(s)
Caspasa 2/metabolismo , Hipoxia Encefálica/tratamiento farmacológico , Hipoxia Encefálica/enzimología , Proteínas Quinasas/farmacología , Familia-src Quinasas/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/enzimología , Citosol/enzimología , Modelos Animales de Enfermedad , Fosfocreatina/metabolismo , Pirimidinas/farmacología , Espectrofotometría , Porcinos , Familia-src Quinasas/metabolismo
18.
Neuroscience ; 140(3): 857-63, 2006 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-16581191

RESUMEN

Previously, we have shown that hypoxia results in increased generation of nitric oxide free radicals in the cerebral cortex of newborn piglets that may be due to up-regulation of nitric oxide synthases, neuronal nitric oxide synthase and inducible nitric oxide synthase. The present study tests the hypothesis that hypoxia results in increased expression of neuronal nitric oxide synthase and inducible nitric oxide synthase in the cerebral cortex of newborn piglets and that the increased expression is nitric oxide-mediated. Newborn piglets, 2-4 days old, were divided to normoxic (n=4), hypoxic (n=4) and hypoxic-treated with 7-nitro-indazole-sodium salt, a selective neuronal nitric oxide synthase inhibitor (hypoxic-7-nitro-indazole-sodium salt, n=6, 1 mg/kg, 60 min prior to hypoxia). Piglets were anesthetized, ventilated and exposed to an FiO2 of 0.21 or 0.07 for 60 min. Cerebral tissue hypoxia was documented biochemically by determining ATP and phosphocreatine. The expression of neuronal nitric oxide synthase and inducible nitric oxide synthase was determined by Western blot using specific antibodies for neuronal nitric oxide synthase and inducible nitric oxide synthase. Protein bands were detected by enhanced chemiluminescence, analyzed by imaging densitometry and the protein band density expressed as absorbance (OD x mm(2)). The density of neuronal nitric oxide synthase in the normoxic, hypoxic and hypoxic-7-nitro-indazole-sodium salt groups was: 41.56+/-4.27 in normoxic, 61.82+/-3.57 in hypoxic (P<0.05) and 47.80+/-1.56 in hypoxic-7-nitro-indazole-sodium salt groups (P=NS vs normoxic), respectively. Similarly, the density of inducible nitric oxide synthase in the normoxic, hypoxic and hypoxic-7-nitro-indazole-sodium salt groups was: 105.21+/-9.09, 157.71+/-13.33 (P<0.05 vx normoxic), 117.84+/-10.32 (p=NS vx normoxic), respectively. The data show that hypoxia results in increased expression of neuronal nitric oxide synthase and inducible nitric oxide synthase proteins in the cerebral cortex of newborn piglets and that the hypoxia-induced increased expression is prevented by the administration of 7-nitro-indazole-sodium salt. Furthermore, the neuronal nitric oxide synthase inhibition prevented the inducible nitric oxide synthase expression for a period of 7 days after hypoxia. Since administration of 7-nitro-indazole-sodium salt prevents nitric oxide generation by inhibiting neuronal nitric oxide synthase, we conclude that the hypoxia-induced increased expression of neuronal nitric oxide synthase and inducible nitric oxide synthase is mediated by neuronal nitric oxide synthase derived nitric oxide. We speculate that during hypoxia nitric oxide-mediated up-regulation of nitric oxide synthases will continue the perpetual cycle of nitric oxide generation-->NOS up-regulation-->nitric oxide generation resulting in hypoxic neuronal death.


Asunto(s)
Corteza Cerebral/enzimología , Hipoxia Encefálica/enzimología , Neuronas/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Animales Recién Nacidos , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiopatología , Infarto Cerebral/enzimología , Infarto Cerebral/fisiopatología , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Hipoxia Encefálica/fisiopatología , Indazoles/farmacología , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/fisiopatología , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Fosfocreatina/metabolismo , Sus scrofa , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
19.
Exp Gerontol ; 41(1): 25-31, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16260109

RESUMEN

The effect of aging on basal and hypoxia/reoxygenation levels of both oxidative stress (protein carbonyl and TBARS) and antioxidative-enzyme activity (Cu/Zn-SOD; Mn-SOD; Catalase, CAT; Se-independent and Se-dependent glutathione peroxidase, GPX; glutathione transferase, GST and glutathione reductase, GR) has been studied in the cerebral cortex of adult and old rats. Oxidative stress markers increased with aging and show an age-dependent post-hypoxic response. Moreover, aging caused either no change (GST, GR and CAT) or an increase (Se-GPX, Cu/Zn-SOD, Mn-SOD) in the basal activity of the enzymes analysed. Only Se-independent GPX activity decreases. However, we detected an age-dependent response of SODs to the hypoxic injury. The early and sustained Cu/Zn-SOD activity rise in adult animals became late and weak in aged animals. Meanwhile, aging slowed the Mn-SOD post-hypoxic response although this activity was consistently higher in aged rats. Aging eliminated the post-hypoxic CAT response, but, perhaps offset by increased GPX activity, did not affect the GST response and slightly reduced post-hypoxic GR activity. In conclusion, aging rise basal ROS production, does not diminish or even increase the antioxidative-enzyme activity, and may slow but does not usually eliminate the enzymatic antioxidant response to the increased post-hypoxic ROS generation.


Asunto(s)
Envejecimiento/fisiología , Corteza Cerebral/enzimología , Hipoxia Encefálica/fisiopatología , Animales , Antioxidantes/metabolismo , Hipoxia Encefálica/enzimología , Peroxidación de Lípido , Masculino , Oxidación-Reducción , Estrés Oxidativo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
20.
Neurosci Lett ; 397(3): 307-12, 2006 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-16406314

RESUMEN

Accumulated reports have suggested that activation of protein kinase C (PKC) isoforms may involve the activation of extracellular signal-regulated kinases 1/2 (ERK1/2) in the neuronal response to hypoxic stimuli. We have previously demonstrated that the membrane translocation or activation of conventional PKC (cPKC) betaII, gamma and novel PKC (nPKC) epsilon are increased in the early phase of cerebral hypoxic preconditioning in mice. However, the role of ERK1/2 in the development of cerebral hypoxic preconditioning is unclear. In the current study, we used Western blot analysis to investigate the effects of repetitive hypoxic exposure (H0-H6, n=6 for each group) on the levels of phosphorylation and protein expression of ERK1/2 in the frontal cortex and the whole hippocampus of mice. We found that the levels of phosphorylated ERK1/2, not protein expression of ERK1/2, decreased significantly in both cortex and hippocampus of the early hypoxic preconditioned mice (H1-H4), when compared to that of the normoxic group (p<0.05). In addition, a significant decrease (p<0.05) in the ERK1/2 protein expression, not the phosphorylated form of ERK1/2, was found both in the frontal cortex and hippocampus of mice followed hypoxia with previous hypoxia (H5 and H6). These results suggest that the decreased phosphorylation and downregulation of protein expression of ERK1/2 might be involved in the development of hypoxic preconditioning.


Asunto(s)
Encéfalo/enzimología , Hipoxia Encefálica/enzimología , Precondicionamiento Isquémico , Proteína Quinasa 1 Activada por Mitógenos/biosíntesis , Proteína Quinasa 3 Activada por Mitógenos/biosíntesis , Animales , Encéfalo/irrigación sanguínea , Lóbulo Frontal/irrigación sanguínea , Lóbulo Frontal/enzimología , Hipocampo/irrigación sanguínea , Hipocampo/enzimología , Masculino , Ratones , Ratones Endogámicos BALB C , Fosforilación
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda