Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
1.
Mil Med Res ; 8(1): 62, 2021 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-34857032

RESUMO

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.


Assuntos
Barreira Hematoencefálica , Cisteína Endopeptidases , Hipóxia Encefálica , Complexo de Endopeptidases do Proteassoma , Via de Sinalização Wnt , beta Catenina , Animais , Barreira Hematoencefálica/metabolismo , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Hipóxia Encefálica/enzimologia , Hipóxia Encefálica/genética , Masculino , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Transfecção , Via de Sinalização Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
2.
Med Hypotheses ; 134: 109419, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31622925

RESUMO

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.


Assuntos
Isquemia Encefálica/enzimologia , Catalase/biossíntese , Endarterectomia das Carótidas/efeitos adversos , Hipóxia Encefálica/enzimologia , Complicações Intraoperatórias/enzimologia , Linfócitos/enzimologia , Superóxido Dismutase-1/biossíntese , Superóxido Dismutase/biossíntese , Isquemia Encefálica/etiologia , Estenose das Carótidas/enzimologia , Estenose das Carótidas/cirurgia , Catalase/sangue , Catalase/genética , Dano ao DNA , Radicais Livres , Regulação Enzimológica da Expressão Gênica , Humanos , Hipóxia Encefálica/etiologia , Complicações Intraoperatórias/etiologia , Mitocôndrias/metabolismo , Modelos Biológicos , Estresse Oxidativo , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/etiologia , Superóxido Dismutase/sangue , Superóxido Dismutase/genética , Superóxido Dismutase-1/sangue , Superóxido Dismutase-1/genética
3.
Mediators Inflamm ; 2017: 2810295, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28626342

RESUMO

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.


Assuntos
Hipóxia Encefálica/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Quinases da Família src/metabolismo , Animais , Animais Recém-Nascidos , Citosol/enzimologia , Modelos Animais de Doenças , Suínos
4.
Neuroreport ; 28(12): 770-773, 2017 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-28658050

RESUMO

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.


Assuntos
Caspase 2/metabolismo , Hipóxia Encefálica/tratamento farmacológico , Hipóxia Encefálica/enzimologia , Proteínas Quinases/farmacologia , Quinases da Família src/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/enzimologia , Citosol/enzimologia , Modelos Animais de Doenças , Fosfocreatina/metabolismo , Pirimidinas/farmacologia , Espectrofotometria , Suínos , Quinases da Família src/metabolismo
5.
Mol Med Rep ; 15(5): 2727-2731, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28447745

RESUMO

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.


Assuntos
Apoptose , Lesões Encefálicas/enzimologia , Calpaína/biossíntese , Caspase 3/biossíntese , Regulação Enzimológica da Expressão Gênica , Hipóxia Encefálica/enzimologia , Miocárdio/enzimologia , Animais , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Feminino , Hipóxia Encefálica/patologia , Masculino , Miocárdio/patologia , Ratos , Ratos Wistar
6.
Ross Fiziol Zh Im I M Sechenova ; 102(12): 1444-52, 2016 Dec.
Artigo em Russo | MEDLINE | ID: mdl-30198248

RESUMO

The effect of melatonin on the correlation between the intensity of the accumulation of the oxidative-modified protein content, activity of the antioxidant enzymes and the state of proteoly-sis in the basal nuclei (the nucleus caudatus, globus pallidus, nucleus accumbens, amigdaloid complex) of the brain under the conditions of acute hypoxia has been studied. Under the conditions of acute hypoxia in the basal nuclei an intensification of the protein peroxidation processes is observed, the activity of the antioxidant enzymes decreases, the intensity of the proteolysis increases. The injection of melatonin in a dose of 1 mg per kg of the body mass before the modeling of acute hypoxia results in the decreasing of protein peroxidation, increasing of the antioxidant enzyme activity and normalization of the parameters of proteolysis.


Assuntos
Antioxidantes/metabolismo , Gânglios da Base/enzimologia , Hipóxia Encefálica/enzimologia , Melatonina/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Doença Aguda , Animais , Gânglios da Base/patologia , Feminino , Hipóxia Encefálica/patologia , Masculino , Oxirredução/efeitos dos fármacos , Ratos
7.
Neurochem Res ; 40(11): 2270-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26342830

RESUMO

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.


Assuntos
Caspase 8/metabolismo , Hipóxia Encefálica/enzimologia , Quinases da Família src/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Caspase 1/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Interleucina-1beta/biossíntese , Fosfocreatina/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Sus scrofa
8.
J Cereb Blood Flow Metab ; 35(12): 2043-51, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26174325

RESUMO

Oxidative stress is a major brain injury mechanism after ischemic stroke. 12/15-lipoxygenase (12/15-LOX) is a key mediator of oxidative stress, contributing to neuronal cell death and vascular leakage. Nonetheless, the mechanism leading to its upregulation is currently unknown. We show here that Signal Transducers and Activators of Transcription (STATs), specifically STAT6 and possibly STAT1, increase transcription of 12/15-LOX in neuronal cells. Both p-STAT6 and -1 bound to specific STAT binding sites in the mouse 12/15-LOX promoter. Small interfering RNA (siRNA) knockdown showed STAT6 to be the dominant regulator, reducing 12/15-LOX promoter activation and cell death in oxidatively stressed HT22 cells. STAT6 siRNA efficiently prevented the increase of 12/15-LOX in murine primary neurons, both after induction of oxidative stress and after oxygen-glucose deprivation. Early activation of STAT6 and STAT1 in mice was consistent with a role in regulating 12/15-LOX in focal ischemia. Brains of human stroke patients showed increased p-STAT6 and p-STAT1 in the peri-infarct region, along with 12/15-LOX and markers of apoptosis. These results link STAT6 and STAT1 to the 12/15-LOX damage pathway and suggest disregulation of STAT-dependent transcription as injury mechanism in stroke. Selectively targeting STATs may thus be a novel therapeutic approach to reducing brain injury after a stroke.


Assuntos
Araquidonato 12-Lipoxigenase/metabolismo , Araquidonato 15-Lipoxigenase/biossíntese , Neurônios/patologia , Fatores de Transcrição STAT/metabolismo , Acidente Vascular Cerebral/enzimologia , Idoso , Animais , Apoptose , Feminino , Técnicas de Silenciamento de Genes , Glucose/deficiência , Humanos , Hipóxia Encefálica/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Estresse Oxidativo , RNA Interferente Pequeno , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Acidente Vascular Cerebral/patologia , Regulação para Cima
9.
J Cereb Blood Flow Metab ; 35(11): 1888-900, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26198177

RESUMO

During focal cerebral ischemia, the degradation of microvessel basal lamina matrix occurs acutely and is associated with edema formation and microhemorrhage. These events have been attributed to matrix metalloproteinases (MMPs). However, both known protease generation and ligand specificities suggest other participants. Using cerebral tissues from a non-human primate focal ischemia model and primary murine brain endothelial cells, astrocytes, and microglia in culture, the effects of active cathepsin L have been defined. Within 2 hours of ischemia onset cathepsin L, but not cathepsin B, activity appears in the ischemic core, around microvessels, within regions of neuron injury and cathepsin L expression. In in vitro studies, cathepsin L activity is generated during experimental ischemia in microglia, but not astrocytes or endothelial cells. In the acidic ischemic core, cathepsin L release is significantly increased with time. A novel ex vivo assay showed that cathepsin L released from microglia during ischemia degrades microvessel matrix, and interacts with MMP activity. Hence, the loss of microvessel matrix during ischemia is explained by microglial cathepsin L release in the acidic core during injury evolution. The roles of cathepsin L and its interactions with specific MMP activities during ischemia are relevant to strategies to reduce microvessel injury and hemorrhage.


Assuntos
Isquemia Encefálica/enzimologia , Isquemia Encefálica/patologia , Catepsina L/metabolismo , Microvasos/enzimologia , Microvasos/patologia , Animais , Astrócitos/enzimologia , Catepsina B/genética , Catepsina B/metabolismo , Catepsina L/genética , Células Cultivadas , Hemorragia Cerebral/enzimologia , Células Endoteliais/enzimologia , Hipóxia Encefálica/enzimologia , Masculino , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/enzimologia , Acoplamento Neurovascular , Papio
10.
Rev Neurosci ; 26(1): 105-17, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25720056

RESUMO

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.


Assuntos
Isquemia Encefálica/enzimologia , Hipóxia Encefálica/enzimologia , Óxido Nítrico Sintase/metabolismo , Animais , Isquemia Encefálica/patologia , Humanos , Hipóxia Encefálica/patologia , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo
11.
Zh Evol Biokhim Fiziol ; 51(6): 427-30, 2015.
Artigo em Russo | MEDLINE | ID: mdl-26983277

RESUMO

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.


Assuntos
Caspase 3/biossíntese , Córtex Cerebral/enzimologia , Regulação Enzimológica da Expressão Gênica , Hipóxia Encefálica/enzimologia , Neprilisina/biossíntese , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Córtex Cerebral/patologia , Feminino , Hipóxia Encefálica/patologia , Ratos , Ratos Wistar
12.
Brain Res ; 1569: 41-7, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24796880

RESUMO

Dynamic microvascular remodeling maintains an optimal continuous supply of oxygen and nutrients to the brain to account for prolonged environmental variations. The objective of this study was to determine the relative time course of capillary regression during re-oxygenation after exposure to prolonged moderate hypoxia and expression of the primary signaling factors involved in the process. Four-month old male C57BL/6 mice were housed and maintained in a hypobaric chamber at 290 Torr (0.4 atm) for 21 days and allowed to recover at normoxia (room air) for up to 21 days. The mice were either decapitated or perfused in-situ and brain samples collected were either homogenized for Western blot analysis or fixed and embedded in paraffin for immunohistochemistry. Hypoxia inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF) and erythropoietin (EPO) expression were increased during hypoxic exposure and diminished during subsequent re-oxygenation. However, cyclooxygenase-2 (COX-2) and angiopoietin-2 (Ang-2) were both elevated during hypoxia as well as subsequent re-oxygenation. Significantly increased capillary density at the end of the 3rd week of hypoxia regressed back toward normoxic baseline as the duration of re-oxygenation continued. In conclusion, elevated COX-2 and Ang-2 expression during hypoxia where angiogenesis occurs and re-oxygenation, when micro-vessels regress, identifies these proteins as vascular remodeling molecules crucial for angioplasticity.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Ciclo-Oxigenase 2/metabolismo , Hipóxia Encefálica/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Remodelação Vascular , Angiopoietina-2/metabolismo , Animais , Encéfalo/enzimologia , Eritropoetina/metabolismo , Hipóxia Encefálica/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Biochem J ; 458(2): 203-11, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24328859

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Cistationina beta-Sintase/biossíntese , Regulação Enzimológica da Expressão Gênica , Hipóxia Encefálica/enzimologia , Animais , Encéfalo/irrigação sanguínea , Células Cultivadas , Cistationina beta-Sintase/genética , Células HEK293 , Humanos , Hipóxia Encefálica/genética , Hipóxia Encefálica/patologia , Fator 1 Induzível por Hipóxia/fisiologia , Masculino , Células PC12 , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Distribuição Tecidual/genética , Vasodilatação/genética
14.
Brain Res ; 1515: 98-107, 2013 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-23562458

RESUMO

Immediately following traumatic brain injury (TBI) and TBI with hypoxia, there is a rapid and pathophysiological increase in extracellular glutamate, subsequent neuronal damage and ultimately diminished motor and cognitive function. N-acetyl-aspartyl glutamate (NAAG), a prevalent neuropeptide in the CNS, is co-released with glutamate, binds to the presynaptic group II metabotropic glutamate receptor subtype 3 (mGluR3) and suppresses glutamate release. However, the catalytic enzyme glutamate carboxypeptidase II (GCP II) rapidly hydrolyzes NAAG into NAA and glutamate. Inhibition of the GCP II enzyme with NAAG peptidase inhibitors reduces the concentration of glutamate both by increasing the duration of NAAG activity on mGluR3 and by reducing degradation into NAA and glutamate resulting in reduced cell death in models of TBI and TBI with hypoxia. In the following study, rats were administered the NAAG peptidase inhibitor PGI-02776 (10mg/kg) 30 min following TBI combined with a hypoxic second insult. Over the two weeks following injury, PGI-02776-treated rats had significantly improved motor function as measured by increased duration on the rota-rod and a trend toward improved performance on the beam walk. Furthermore, two weeks post-injury, PGI-02776-treated animals had a significant decrease in latency to find the target platform in the Morris water maze as compared to vehicle-treated animals. These findings demonstrate that the application of NAAG peptidase inhibitors can reduce the deleterious motor and cognitive effects of TBI combined with a second hypoxic insult in the weeks following injury.


Assuntos
Lesões Encefálicas/enzimologia , Transtornos Cognitivos/enzimologia , Glutamato Carboxipeptidase II/antagonistas & inibidores , Hipóxia Encefálica/enzimologia , Destreza Motora/efeitos dos fármacos , Destreza Motora/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Animais , Lesões Encefálicas/tratamento farmacológico , Transtornos Cognitivos/tratamento farmacológico , Modelos Animais de Doenças , Glutamato Carboxipeptidase II/fisiologia , Hipóxia Encefálica/tratamento farmacológico , Masculino , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley , Ureia/análogos & derivados , Ureia/farmacologia , Ureia/uso terapêutico
15.
Patol Fiziol Eksp Ter ; (3): 11-9, 2012.
Artigo em Russo | MEDLINE | ID: mdl-23072108

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Hipóxia Encefálica/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Transdução de Sinais , Adaptação Fisiológica , Animais , Encéfalo/enzimologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Sinalização do Cálcio , Humanos , Hipóxia Encefálica/enzimologia , Hipóxia Encefálica/patologia , Hipóxia Encefálica/fisiopatologia , Imuno-Histoquímica , Fosfatos de Inositol/metabolismo , Neurônios/enzimologia , Neurônios/metabolismo , Neurônios/patologia , Fosfolipases/metabolismo , Proteína Quinase C/metabolismo
16.
Naunyn Schmiedebergs Arch Pharmacol ; 384(2): 177-83, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21713382

RESUMO

R(-) enantiomers of the 2-arylpropionic acid derivatives ibuprofen and flurbiprofen weakly inhibit cyclooxygenase (COX) activity. However, a possible cytoprotective effect has been proposed. The aim of the study is to investigate the possible mechanism of this effect. An in vitro hypoxia-reoxygenation model in rat brain slices was used (n=6 rats per group). After reoxygenation, we measured LDH efflux (neuronal death), brain prostaglandin E(2) (PGE(2)) concentration, interleukins (IL)-1ß and 10, oxidative and nitrosative stress (lipid peroxides, glutathione, 3-nitrotyrosine, and nitrites/nitrates). Anti-COX activity was measured in human whole blood. Racemic, R(-), and S(+) enantiomers of ibuprofen and flurbiprofen were tested. All compounds had a cytoprotective effect with IC(50) values in the range of 10(-5) M. R(-) enantiomers did not significantly inhibit brain PGE(2). The concentration of IL-1ß was reduced by 53.1% by the racemic form, 30.6% by the S(+) and 43.2% by the R(-) enantiomer of ibuprofen. The IL-10 concentration increased significantly only with S(+)-flurbiprofen (33.1%) and R(-)-flurbiprofen (26.1%). Lipid peroxidation was significantly reduced by all three forms of flurbiprofen. Nitrite + nitrate concentrations were reduced by racemic, S(+), and R(-)-flurbiprofen. Peroxynitrite formation (3-nitrotyrosine) was significantly reduced by racemic and S(+)-ibuprofen. COX inhibition is not the main mechanism of cytoprotection for these compounds. Their influence on inflammatory mediators and oxidative and nitrosative stress could account for the potential cytoprotective effect of R(-) enantiomers.


Assuntos
Encéfalo/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Citoproteção/efeitos dos fármacos , Flurbiprofeno/farmacologia , Ibuprofeno/farmacologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Animais , Encéfalo/enzimologia , Encéfalo/imunologia , Encéfalo/patologia , Inibidores de Ciclo-Oxigenase/química , Citocinas/metabolismo , Dinoprostona/metabolismo , Flurbiprofeno/química , Hipóxia Encefálica/enzimologia , Hipóxia Encefálica/imunologia , Hipóxia Encefálica/patologia , Hipóxia Encefálica/prevenção & controle , Ibuprofeno/química , Técnicas In Vitro , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar , Estereoisomerismo , Tirosina/análogos & derivados , Tirosina/análise
17.
Brain Res ; 1395: 74-85, 2011 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-21555119

RESUMO

The enhanced carotid body (CB) chemosensory response to hypoxia induced by chronic intermittent hypoxia (CIH) has been attributed to oxidative stress, which is expected to increase the expression of chemosensory modulators including chemoexcitatory pro-inflammatory cytokines in the CB. Accordingly, we studied the time-course of the changes in the immunohistological expression of TNF-α, IL-1ß, IL-6, ET-1, iNOS, eNOS and 3-nitrotyrosine in the CB, along with the progression of enhanced CB chemosensory responses to acute hypoxia in male Sprague-Dawley rats exposed to CIH (5%O2, 12 times/h per 8h) for 7, 14 and 21 days. Exposure to CIH for 7 days resulted in a sustained potentiation of CB chemosensory responses to acute hypoxia, which persisted until 21 days of CIH. The chemosensory potentiation was paralleled by an increased 3-nitrotyrosine expression in the CB. On the contrary, CIH produced a transient 2-fold increase of ET-1 immunoreactivity at 7 days, a decrease in eNOS immunoreactivity, and a delayed but progressive increase of TNF-α, IL-1ß and iNOS immunoreactivity, which was not associated with changes in systemic plasma levels or immune cell invasion within the CB. Thus, present results suggest that the local expression of chemosensory modulators and pro-inflammatory cytokines in the CB may have different temporal contribution to the CB chemosensory potentiation induced by CIH.


Assuntos
Corpo Carotídeo/metabolismo , Citocinas/biossíntese , Endotelina-1/biossíntese , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/patologia , Mediadores da Inflamação/metabolismo , Óxido Nítrico Sintase Tipo III/biossíntese , Óxido Nítrico Sintase Tipo II/biossíntese , Animais , Corpo Carotídeo/enzimologia , Corpo Carotídeo/patologia , Citocinas/sangue , Modelos Animais de Doenças , Endotelina-1/sangue , Hipóxia Encefálica/enzimologia , Inflamação/sangue , Inflamação/enzimologia , Inflamação/metabolismo , Mediadores da Inflamação/sangue , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Regulação para Cima/fisiologia
18.
Int J Neurosci ; 121(5): 279-88, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21348795

RESUMO

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.


Assuntos
Doença da Altitude/tratamento farmacológico , Inibidores da Colinesterase/farmacologia , Hipóxia Encefálica/tratamento farmacológico , Transtornos da Memória/tratamento farmacológico , Memória/efeitos dos fármacos , Acetilcolina/agonistas , Acetilcolina/fisiologia , Pressão do Ar , Doença da Altitude/complicações , Doença da Altitude/enzimologia , Animais , Modelos Animais de Doenças , Galantamina/farmacologia , Hipóxia Encefálica/complicações , Hipóxia Encefálica/enzimologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Transtornos da Memória/enzimologia , Transtornos da Memória/etiologia , Fisostigmina/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Resultado do Tratamento
19.
J Neurosci ; 29(15): 4903-10, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19369559

RESUMO

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.


Assuntos
Corpo Carotídeo/enzimologia , Hipóxia Encefálica/enzimologia , NADPH Oxidases/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Corpo Carotídeo/metabolismo , Doença Crônica , Hipóxia Encefálica/metabolismo , Hipóxia Encefálica/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vias Neurais/enzimologia , Vias Neurais/metabolismo , Proteína Quinase C/fisiologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Receptores 5-HT2 de Serotonina/fisiologia , Serotonina/fisiologia
20.
Cell Cycle ; 8(8): 1271-8, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19305142

RESUMO

In acute thromboembolic stroke, neurological damage is due to ischemia-induced apoptotic death of neuronal cells and the surrounding vascular network. Here, we demonstrate that the BH4 domain of the anti-apoptotic protein, Bcl-x(L), attached to the membrane transport peptide, TAT, reduces stroke injury after intracerebroventricular infusion into immature rats subjected to carotid artery ligation and additional exposure to hypoxia. The injected TAT-BH4 entered neuron bodies, maintained brain architecture, protected neuronal and endothelial cells from apoptosis and promoted neuronal stem cell recruitment. In vitro, TAT-BH4 enhanced the survival of endothelial cells exposed to H(2)O(2), increased neuronal differentiation, and induced axonal remodelling of adult neuronal stem cells. These findings indicate that TAT-BH4 administration protects against acute hypoxia/ischemia injury in the brain by preventing endothelial and neuron cell apoptosis and by inducing neuronal plasticity.


Assuntos
Apoptose/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/prevenção & controle , Membrana Celular/metabolismo , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Animais , Isquemia Encefálica/enzimologia , Isquemia Encefálica/patologia , Inibidores de Caspase , Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipóxia Encefálica/tratamento farmacológico , Hipóxia Encefálica/enzimologia , Hipóxia Encefálica/patologia , Hipóxia Encefálica/prevenção & controle , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...