Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Bioengineered ; 12(2): 9790-9805, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34672892

RESUMO

Hypoxic-ischemic encephalopathy (HIE) is recognized as the main cause of neonatal death, and efficient treatment strategies remain limited. This study aims to investigate the mechanism of sevoflurane (SF) post-treatment in alleviating HIE in rats. The HIE rat model and oxygen-glucose deprivation (OGD) cell model were established, and adeno-associated virus (AAV)-histone-lysine N-methyltransferase EHMT2 (G9a) was transfected after SF treatment. The learning and memory ability and the levels of nerve growth factor (NGF)/brain-derived neurotrophic factor (BDNF) were evaluated and determined. The levels of G9a/histone H3 lysine 9 dimethylation (H3K9me2) and the enrichment level of H3K9me2 in the promoter region of BDNF gene were analyzed. After SF post-treatment, the neurons in cerebral cortex, the learning and memory skills and the contents of NGF/BDNF were increased, while the apoptosis and G9a/H3K9me2 levels were reduced. After overexpression of G9a in vitro/vivo, the enrichment levels of H3K9me2 in the promoter region of BDNF were increased, the levels of BDNF were decreased, the neurons were damaged and the learning and memory abilities of HIE rats were impaired. The conclusion is that SF post-treatment can promote the expression of BDNF by inhibiting H3K9me2 on the BDNF gene promoter and inhibiting G9a, thus alleviating HIE in rats.


Assuntos
Córtex Cerebral/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/biossíntese , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Neurônios/enzimologia , Sevoflurano/farmacologia , Animais , Hipóxia-Isquemia Encefálica/enzimologia , Masculino , Ratos , Ratos Sprague-Dawley
2.
PLoS One ; 13(7): e0200933, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30044838

RESUMO

Birth asphyxia resulting in brain hypoxia-ischemia (H-I) can cause neonatal death or lead to persistent brain damage. Recent investigations have shown that group II metabotropic glutamate receptor (mGluR2/3) activation can provide neuroprotection against H-I but the mechanism of this effect is not clear. The aim of this study was to investigate whether mGluR2/3 agonists applied a short time after H-I reduce brain damage in an experimental model of birth asphyxia, and whether a decrease in oxidative stress plays a role in neuroprotection. Neonatal H-I in 7-day-old rats was used as an experimental model of birth asphyxia. Rats were injected intra peritoneally with mGluR2 (LY 379268) or mGluR3 (NAAG) agonists 1 h or 6 h after H-I (5 mg/kg). The weight deficit of the ischemic brain hemisphere, radical oxygen species (ROS) content levels, antioxidant enzymes activity and the concentrations of reduced glutathione (GSH) were measured. Both agonists reduced weight loss in the ischemic hemisphere and mitigated neuronal degeneration in the CA1 hippocampal region and cerebral cortex. Both agonists reduced the elevated levels of ROS in the ipsilateral hemisphere observed after H-I and prevented an increase in antioxidant enzymes activity in the injured hemisphere restoring them to control levels. A decrease in GSH level was also restored after agonists application. The results show that the activation of mGluR2 and mGluR3 a short time after H-I triggers neuroprotective mechanisms that act through the inhibition of oxidative stress and ROS production. The prevention of ROS production by the inhibition of glutamate release and decrease in its extracellular concentration is likely the main mechanism involved in the observed neuroprotection.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/agonistas , Animais , Animais Recém-Nascidos , Antioxidantes/metabolismo , Encéfalo/enzimologia , Feminino , Glutationa/metabolismo , Hipóxia-Isquemia Encefálica/enzimologia , Masculino , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
3.
Biomed Pharmacother ; 84: 535-543, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27693962

RESUMO

BACKGROUND: Isoflurane postconditioning (ISPOC) plays a neuroprotection role in the brain. Previous studies confirmed that isoflurane postconditioning can provide better protection than preconditioning in acute hypoxic-ischemic brain damage, such as acute craniocerebral trauma and ischemic stroke. Numerous studies have reported that activin A can protect rat's brain from cell injury. However, whether activin A and its downstream ERK1/2 were involved in isoflurane postconditioning-induced neuroprotection is unknown. METHODS: A total of 80 healthy Sprague-Dawley rats weighing 50-70g were randomly divided into 10 groups of 8: normal control, oxygen and glucose deprivation (OGD), 1.5% ISPOC, 3.0% ISPOC, 4.5% ISPOC, blocker of activin A (SB431542), blocker of ERK1/2 (U0126), 3.0% ISPOC+SB431542, 3.0% ISPOC+U0126, and vehicle (dimethyl sulfoxide(DMSO)) group. Blockers (SB431542 and U0126) were used in each concentration of isoflurane before OGD. Hematoxylin-eosin staining, 2,3,5-triphenyl tetrazolium chloride staining, and propidium iodide (PI) staining were conducted to assess the reliability in the brain slices. Immunofluorescence, Western blot, and quantitative real-time PCR(Q-PCR) were performed to validate the protein expression levels of activin A, Smad2/3, P-Smad2/3, ERK1/2, and phosphorylation ERK1/2 (P-ERK1/2). RESULTS: The number of damaged neurons and mean fluorescence intensity(MFI) of PI staining increased, but formazan generation, expression levels of activin A and P-ERK1/2 protein, and mRNA synthesis level of activin A decreased in the OGD group compared with the normal control group (p<0.05). The number of damaged neurons and MFI of PI staining decreased, but formazan production, expression levels of activin A, P-Smad2/3, and P-ERK1/2, and mRNA synthesis level of activin A increased significantly in the 1.5% ISPOC and 3.0% ISPOC groups (p<0.05) compared with the OGD group. The result in the 4.5% ISPOC group, was completely opposite to the 1.5% ISPOC and 3.0% ISPOC groups. The number of damage neuron and MFI of PI staining increased, but formazan production, expression levels of activin A, P-Smad2/3, and P-ERK1/2, and mRNA synthesis level of activin A decreased in the 4.5% ISPOC group. However, the expression levels of activin A, P-Smad2/3, and P-ERK1/2, and mRNA synthesis level of activin A in the 4.5% ISPOC group were higher than the OGD group (p<0.05). The other results were compared between the SB431542 group/the U0126 group and 3.0% ISPOC group. The MFI of PI staining increased, but the expression levels of activin A, P-Smad2/3, and P-ERK1/2 decreased (p<0.05). The expression level of ERK1/2 protein in all groups exhibited no change (p>0.05). CONCLUSION: Results of this study showed that 3.0% concentration of isoflurane postconditioning provided better neuroprotection than 1.5% and 4.5% concentrations of isoflurane. Activin A/Smad 2/3 and activin A/ERK1/2 signaling pathway may be involved in ISPOC-induced neuroprotection.


Assuntos
Hipocampo/efeitos dos fármacos , Hipóxia-Isquemia Encefálica/prevenção & controle , Subunidades beta de Inibinas/metabolismo , Isoflurano/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Citoproteção , Relação Dose-Resposta a Droga , Glucose/deficiência , Hipocampo/enzimologia , Hipocampo/patologia , Hipóxia-Isquemia Encefálica/enzimologia , Hipóxia-Isquemia Encefálica/patologia , Técnicas In Vitro , Subunidades beta de Inibinas/genética , Degeneração Neural , Neurônios/enzimologia , Neurônios/patologia , Fosforilação , Ratos Sprague-Dawley , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fatores de Tempo
4.
Am J Physiol Cell Physiol ; 310(11): C903-10, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27009876

RESUMO

Astrocytes, the most numerous cells in the human brain, play a central role in the metabolic homeostasis following hypoxic injury. Caveolin-1 (Cav-1), a transmembrane scaffolding protein, has been shown to converge prosurvival signaling in the central nerve system. The present study aimed to investigate the role of Cav-1 in the hypoxia-induced astrocyte injury. We also examined how Cav-1 alleviates apoptotic astrocyte death. To this end, primary astrocytes were exposed to oxygen-glucose deprivation (OGD) for 6 h and a subsequent 24-h reoxygenation to mimic hypoxic injury. OGD significantly reduced Cav-1 expression. Downregulation of Cav-1 using Cav-1 small interfering RNA dramatically worsened astrocyte cell damage and impaired cellular glutamate uptake after OGD, whereas overexpression of Cav-1 with Cav-1 scaffolding domain peptide attenuated OGD-induced cell apoptosis. Mechanistically, the expressions of Ras-GTP, phospho-Raf, and phospho-ERK were sequestered in Cav-1 small interfering RNA-treated astrocytes, yet were stimulated after supplementation with caveolin peptide. MEK/ERK inhibitor U0126 remarkably blocked the Cav-1-induced counteraction against apoptosis following hypoxia, indicating Ras/Raf/ERK pathway is required for the Cav-1's prosurvival role. Together, these findings support Cav-1 as a checkpoint for the in hypoxia-induced astrocyte apoptosis and warrant further studies targeting Cav-1 to treat hypoxic-ischemic brain injury.


Assuntos
Apoptose , Astrócitos/enzimologia , Encéfalo/enzimologia , Caveolina 1/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipóxia-Isquemia Encefálica/enzimologia , Quinases raf/metabolismo , Proteínas ras/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Caveolina 1/genética , Hipóxia Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Glucose/deficiência , Ácido Glutâmico/metabolismo , Hipóxia-Isquemia Encefálica/genética , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/prevenção & controle , Fosforilação , Cultura Primária de Células , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Ratos Sprague-Dawley , Transdução de Sinais , Transfecção
5.
CNS Neurol Disord Drug Targets ; 15(1): 64-72, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26553162

RESUMO

Recent findings have demonstrated a dual effect of the folic acid (FA) supplementation on the nervous system of rats. We found that FA treatment prevented memory impairment and Na(+), K(+)- ATPase inhibition in the striatum and cortex in adult rats that suffered neonatal hypoxia-ischemia (HI). However, spatial memory deficit has been associated with FA supplementation. In the present study we investigated the role of FA supplementation on spatial memory and Na(+), K(+)-ATPase activity in the hippocampus, as well as on morphologic alterations in adolescent rats submitted to neonatal HI. Wistar rats of both sexes at postnatal day (PND) 7 were submitted to Levine-Rice HI procedure. Intraperitoneal doses of FA were administered immediately before HI and repeated daily until the maximum PND 40. Hippocampal volume and striatum area were estimated and Na(+), K(+)-ATPase activity in the hippocampus was measured at PND 31. Also, the performance of the animals in the water maze was assessed and Na(+), K(+)-ATPase activity measured again at PND 52. Interestingly, HI and FA resulted in spatial memory deficits in the Morris water maze and the Na(+), K(+)-ATPase activity was impaired at PND 31 in HI rats which received FA. Additionally, Na(+), K(+)-ATPase activity in adulthood showed a decrease after HI and a recovery in supplemented animals. Hippocampal and striatal atrophy were partially reversed by FA. To conclude, the present results support the hypothesis that FA supplementation during development contributes to memory deficits caused by HI and Na(+), K(+)-ATPase failure in adolescent rats, although, in adulthood, FA has been effective in reversing enzymatic activity in the hippocampus.


Assuntos
Ácido Fólico/toxicidade , Hipocampo/enzimologia , Hipóxia-Isquemia Encefálica/enzimologia , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipóxia-Isquemia Encefálica/patologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/patologia , Ratos , Ratos Wistar , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores
6.
Int J Clin Exp Pathol ; 8(6): 6960-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26261584

RESUMO

The specific and available markers proteins of neonatal hypoxic-ischemic encephalopathy (HIE) injury are correlated with disease severity and the disability in childhood. Exploring the mechanism of HIE is very helpful to the targeted therapeutic approach in clinical. This study aims to explore the cell death-related proteins or biomarkers that plays roles in the HIE injury. In this study, 15 patients were included the 487 autopsies patients performed at the Department of Pathology. The lactate dehydrogenase (LDH) assay was used to detect the cell viability of NGF-differentiated PC12 cell. TUNEL assay was employed to examine the apoptotic cells in embedded slides samples. Three ER stress-related protein, including ATF6, p-Perk and IRE-1 were investigated using Western blot assay for the ER stress examination. The apoptosis associated caspase-12 and CHOP protein were detected by Western blot. The results indicated that LDH activity of living cells during hypoxia was significantly enhanced to 45% and 64% after 8 hours and 24 hours. The TUNEL results showed that plenty of the PC12 cells became the positive staining cells when treated with 0.1% O2 hypoxia. ER stress UPR pathway protein, cleaved ATF6, was increased significantly when treated with 0.1% O2 compared with the cells treated with 20% O2. Furthermore, the caspase 12 activation was triggered when the cells treated with the 0.1% O2. In conclusion, apoptosis is served as an important factor that triggers the HIE brain injury through cleaving the ATF6 and caspase-12 ER stress-related protein.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Apoptose , Encéfalo/enzimologia , Caspase 12/metabolismo , Estresse do Retículo Endoplasmático , Retículo Endoplasmático/enzimologia , Hipóxia-Isquemia Encefálica/enzimologia , Animais , Autopsia , Biomarcadores/metabolismo , Encéfalo/patologia , Hipóxia Celular , Sobrevivência Celular , Retículo Endoplasmático/patologia , Feminino , Humanos , Hipóxia-Isquemia Encefálica/mortalidade , Hipóxia-Isquemia Encefálica/patologia , Recém-Nascido , L-Lactato Desidrogenase/metabolismo , Masculino , Neurônios/enzimologia , Neurônios/patologia , Células PC12 , Ratos , Transdução de Sinais , Fatores de Tempo
7.
Biochim Biophys Acta ; 1850(6): 1274-85, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25735211

RESUMO

BACKGROUND: Thioredoxin (Trx) family proteins are crucial mediators of cell functions via regulation of the thiol redox state of various key proteins and the levels of the intracellular second messenger hydrogen peroxide. Their expression, localization and functions are altered in various pathologies. Here, we have analyzed the impact of Trx family proteins in neuronal development and recovery, following hypoxia/ischemia and reperfusion. METHODS: We have analyzed the regulation and potential functions of Trx family proteins during hypoxia/ischemia and reoxygenation of the developing brain in both an animal and a cellular model of perinatal asphyxia. We have analyzed the distribution of 14 Trx family and related proteins in the cerebellum, striatum, and hippocampus, three areas of the rat brain that are especially susceptible to hypoxia. Using SH-SY5Y cells subjected to hypoxia and reoxygenation, we have analyzed the functions of some redoxins suggested by the animal experiment. RESULTS AND CONCLUSIONS: We have described/discovered a complex, cell-type and tissue-specific expression pattern following the hypoxia/ischemia and reoxygenation. Particularly, Grx2 and Trx1 showed distinct changes during tissue recovery following hypoxia/ischemia and reoxygenation. Silencing of these proteins in SH-SY5Y cells subjected to hypoxia-reoxygenation confirmed that these proteins are required to maintain the normal neuronal phenotype. GENERAL SIGNIFICANCE: These findings demonstrate the significance of redox signaling in cellular pathways. Grx2 and Trx1 contribute significantly to neuronal integrity and could be clinically relevant in neuronal damage following perinatal asphyxia and other neuronal disorders.


Assuntos
Asfixia Neonatal/enzimologia , Encéfalo/enzimologia , Glutarredoxinas/metabolismo , Hipóxia-Isquemia Encefálica/enzimologia , Neurônios/enzimologia , Tiorredoxinas/metabolismo , Animais , Asfixia Neonatal/patologia , Encéfalo/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Glutarredoxinas/genética , Humanos , Hipóxia-Isquemia Encefálica/patologia , Masculino , Neurônios/patologia , Oxirredução , Oxigênio/metabolismo , Fenótipo , Interferência de RNA , Ratos Sprague-Dawley , Transdução de Sinais , Tiorredoxinas/genética , Fatores de Tempo , Transfecção
8.
Mol Cell Biochem ; 403(1-2): 169-77, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25720338

RESUMO

Hypoxic-ischemic (HI) injury perinatal brain is a major contributor to morbidity and mortality to infants and children. Adenosine may play a role in the pathophysiology of HI, since it modulates the inflammatory process and the release of several neurotransmitters. Thus, the aim of this study was to identify the isoforms of adenosine deaminase (ADA) responsible for the enzymatic activity as well as the adenosine kinase (ADK) and A1 adenosine receptor (A1R) expression in the cerebral cortex eight days after HI. Myeloperoxidase (MPO) and N-acetyl-glucosaminidase (NAG) were assessed as inflammation markers. ADA activity was analyzed, in the presence or absence of a specific ADA1 inhibitor, erythro-9-(2-hydroxy-3-nonyl) adenine. The ADA1 activity (92.6%) was significantly higher than ADA2 (7.4%) activity in the cerebral cortex eight days after HI. A1Rs and ADK protein expression showed decreased 8 days after insult. Interestingly, the ADA1, MPO, and NAG activities were correlated positively. In view of this, we conclude that the inhibitor of ADA1, in in vitro conditions, was effective in decreasing the ADA activity, and that mainly ADA1 isoform is responsible for the increase in the ADA activity 8 days after HI insult. Therefore, HI neonatal was able to alter the ADK and A1R expression. Thus, due to the importance of adenosine signaling in the regulation of inflammatory and immune process and the crucial role of ADA in the postischemic homeostase of adenosine as well as during inflammatory process, we suggest that ADA1 inhibitors may play an important role in the regulation of events that follow the HI insult, favoring the increase in the adenosine in the sites of tissue injury. Together, these results highlight a role of the purinergic signaling cascade in the pathophysiology of HI neonatal.


Assuntos
Adenosina Desaminase/metabolismo , Encéfalo/patologia , Hipóxia-Isquemia Encefálica/enzimologia , Hipóxia-Isquemia Encefálica/patologia , Inflamação/patologia , Purinas/metabolismo , Acetilglucosaminidase/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Adenosina Quinase/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Córtex Cerebral/enzimologia , Córtex Cerebral/patologia , Isoenzimas/metabolismo , Masculino , Peroxidase/metabolismo , Ratos Wistar , Receptor A1 de Adenosina/metabolismo
9.
Neuroscience ; 291: 118-27, 2015 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-25617656

RESUMO

In animal models, environmental enrichment (EE) has been found to be an efficient treatment for alleviating the consequences of neonatal hypoxia-ischemia (HI). However the potential for this therapeutic strategy and the mechanisms involved are not yet clear. The aim of present study is to investigate behavioral performance in the ox-maze test and Na+,K+-ATPase, catalase (CAT) and glutathione peroxidase (GPx) activities in the hippocampus of rats that suffered neonatal HI and were stimulated in an enriched environment. Seven-day-old rats were submitted to the HI procedure and divided into four groups: control maintained in standard environment (CTSE), control submitted to EE (CTEE), HI in standard environment (HISE) and HI in EE (HIEE). Animals were stimulated with EE for 9 weeks (1 h/day for 6 days/week) and then behavioral and biochemical parameters were evaluated. Present results indicate learning and memory in the ox-maze task were impaired in HI rats and this effect was recovered after EE. Hypoxic-ischemic event did not alter the Na+,K+-ATPase activity in the right hippocampus (ipsilateral to arterial occlusion). However, on the contralateral hemisphere, HI caused a decrease in this enzyme activity that was recovered by EE. The activities of GPx and CAT were not changed by HI in any group evaluated. In conclusion, EE was effective in recovering learning and memory impairment in the ox-maze task and Na+,K+-ATPase activity in the hippocampus caused by HI. The present data provide further support for the therapeutic potential of environmental stimulation after neonatal HI in rats.


Assuntos
Meio Ambiente , Hipocampo/enzimologia , Hipóxia-Isquemia Encefálica/terapia , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/terapia , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Animais Recém-Nascidos , Catalase/metabolismo , Modelos Animais de Doenças , Glutationa Peroxidase/metabolismo , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/enzimologia , Deficiências da Aprendizagem/enzimologia , Deficiências da Aprendizagem/etiologia , Deficiências da Aprendizagem/terapia , Transtornos da Memória/enzimologia , Transtornos da Memória/etiologia , Distribuição Aleatória , Ratos Wistar , Resultado do Tratamento
10.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 45(5): 767-71, 2014 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-25341336

RESUMO

OBJECTIVE: To examine the changes in expression of phosphatase and tensin homolog deleted on chromosome ten (PTEN) protein, p-PTEN protein and Bim (Bcl-2 interacting mediator of cell death) mRNA in the cortex of neonate rat brains with hypoxic-ischemic brain damage (HIBD) and to explore the mechanisms of neuroprotective effects of PTEN inhibition. METHODS: One hundred and twenty-eight neonate (10 days) SD rats were divided into four groups: hypoxia-ischemia (HI), sham control (Sham), bisperoxovanadium (bpv), and normal saline (NS) group. Rats in the HI group had their right common carotid arteries (CCA) exposed and ligated, and were then exposed to hypoxia in a chamber filled with 8% oxygen (balanced with nitrogen) for 2.5 h. Rats in the sham control group had their right CCA surgically exposed without ligation and exposure to hypoxia. Rats in the bpv treated group received intraperitoneal injections of bpv, 30 min before HI was induced. Instead of bpv, rats in the NS-treated group received intraperitoneal injections of NS. Cerebral cortex samples of the rats were collected 0.5 h and 24 h after hypoxia. Western blot was used to detect the protein expression of PTEN, p-PTEN and Bim. Real-Time PCR was used to detect the level of Bim mRNA. TUNEL staining was used to detect apoptotic cells. RESULTS: No significant changes of PTEN protein were observed in the rats exposed to HI. However, p-PTEN protein decreased in the rats exposed to HI (0.5 h and 24 h) compared with those exposed to sham surgery (P < 0.01). Compared with the sham controls, Bim mRNA and protein increased in the rats exposed to HI (0.5 h, P < 0.01) and then returned to the baseline level 24 h after HI. No significant changes of PTEN protein were observed in the bpv-treated rats. However, p-PTEN protein increased and Bim mRNA and protein decreased in the bpv-treated rats (0.5 h and 24 h, P < 0.01) compared with those in the HI group and NS-treated group. TUNEL positive cells also reduced in the bpv-treated rats (24 h, P < 0.01) compared with those in the HI group and NS-treated group. CONCLUSION: PTEN activities increase in the brains of neonate rats with hypoxic-ischemic brain damage. PTEN activity inhibition can decrease the level of pro-apoptotic protein Bim mRNA, leading to reduction of neuronal apoptosis.


Assuntos
Apoptose , Hipóxia-Isquemia Encefálica/enzimologia , Neurônios/citologia , PTEN Fosfo-Hidrolase/metabolismo , Animais , Animais Recém-Nascidos , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 11 Semelhante a Bcl-2 , Encéfalo/patologia , Hipóxia-Isquemia Encefálica/patologia , Proteínas de Membrana/metabolismo , Neurônios/enzimologia , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro , Ratos , Ratos Sprague-Dawley
11.
Brain Res ; 1589: 68-77, 2014 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-25304361

RESUMO

Prior treatment with dexamethasone (Dex) provides neuroprotection against hypoxia ischemia (HI) in newborn rats. Recent studies have shown that the phosphatidylinositol-3-kinase/Akt (PI3K/Akt) pathway plays an important role in the neuroprotection. The objective of this study is to evaluate the role of the PI3K/Akt pathway in the Dex-induced neuroprotection against subsequent HI brain injury. Seven-day-old rat pups had the right carotid artery permanently ligated followed by 160min of hypoxia (8% oxygen). Rat pups received i.p. injection of either saline or Dex (0.25mg/kg) at 24 and 4h before HI exposure. To quantify the effects of a PI3K/Akt inhibitor, wortmannin (1µl of 1µg/µl) or vehicle was injected intracerebroventricularly in the right hemisphere on postnatal day 6 at 30min prior to the first dose of Dex or saline treatment. Dex pretreatment significantly reduced the brain injury following HI which was quantified by the decrease in cleaved caspase-3 protein as well as cleaved caspase-3 and TUNEL positive cells at 24h and percent loss of ipsilateral hemisphere weight at 22d after HI, while wortmannin partially reversed these effects. We conclude that Dex provides robust neuroprotection against subsequent HI in newborn rats in part via activation of PI3/Akt pathway.


Assuntos
Dexametasona/farmacologia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/enzimologia , Fármacos Neuroprotetores/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Androstadienos/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/patologia , Artérias Carótidas , Caspase 3/metabolismo , Modelos Animais de Doenças , Feminino , Hipóxia-Isquemia Encefálica/patologia , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Distribuição Aleatória , Ratos Sprague-Dawley , Wortmanina
12.
PLoS One ; 9(2): e88962, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24551199

RESUMO

Perinatal hypoxic ischemia (H-I) causes brain damage and long-term neurological impairments, leading to motor dysfunctions and cerebral palsy. Many studies have demonstrated that the TrkB-ERK1/2 signaling pathway plays a key role in mediating the protective effect of brain-derived neurotrophic factor (BDNF) following perinatal H-I brain injury in experimental animals. In the present study, we explored the neuroprotective effects of the TrkB-specific agonist monoclonal antibody 29D7 on H-I brain injury in neonatal rats. First, we found that intracerebroventricular (icv) administration of 29D7 in normal P7 rats markedly increased the levels of phosphorylated ERK1/2 and phosphorylated AKT in neurons up to 24 h. Second, P7 rats received icv administration of 29D7 and subjected to H-I injury induced by unilateral carotid artery ligation and exposure to hypoxia (8% oxygen). We found that 29D7, to a similar extent to BDNF, significantly inhibited activation of caspase-3, a biochemical hallmark of apoptosis, following H-I injury. Third, we found that this 29D7-mediated neuroprotective action persisted at least up to 5 weeks post-H-I injury as assessed by brain tissue loss, implicating long-term neurotrophic effects rather than an acute delay of cell death. Moreover, the long-term neuroprotective effect of 29D7 was tightly correlated with sensorimotor functional recovery as assessed by a tape-removal test, while 29D7 did not significantly improve rotarod performance. Taken together, these findings demonstrate that pretreatment with the TrkB-selective agonist 29D7 significantly increases neuronal survival and behavioral recovery following neonatal hypoxic-ischemic brain injury.


Assuntos
Anticorpos/farmacologia , Hipóxia-Isquemia Encefálica/patologia , Atividade Motora/efeitos dos fármacos , Receptor trkB/agonistas , Células Receptoras Sensoriais/patologia , Animais , Animais Recém-Nascidos , Anticorpos/administração & dosagem , Temperatura Corporal/efeitos dos fármacos , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/enzimologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Injeções Intraventriculares , Masculino , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor trkB/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/enzimologia
13.
Brain Res ; 1507: 105-14, 2013 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-23466455

RESUMO

Hypoxia-ischemia on 3-day-old rats (HIP3) allows the investigation of HI damage in the immature brain. HIP3 is characterized for neurological disabilities caused by white matter injury. This study investigates the relationship between animals' sex and injured hemisphere on HIP3 consequences. Male and female Wistar rats had their right or left common carotid artery occluded under halotane anesthesia and exposed to 8% O2 for 1.5 h. Control rats received sham surgery and exposure to 1.5 h of room air in isolation of their mothers. Sex and injured hemisphere influence in Na+/K+ -ATPase activity 24h after lesion: females and the right brain hemispheres showed decreased enzymatic activity after HIP3. Cognitive impairment was observed in step-down inhibitory avoidance, in which females HIP3 left injured were the most damaged. Histological analysis showed a trend to white matter damage in females left injured without hemispherical nor hippocampal volume decrease in HIP3 rats at postnatal day 21. However, at PND90, hemisphere and sex effects were noted in hemispherical volume and myelination: left brain hemisphere and the females evidenced higher histological damage. Our results points to an increased resistance of male rats and right brain hemisphere to support the impairment caused in Na+/K+ -ATPase activity early after HIP3, and evidencing more discrete behavioral impairments and histological damage at adulthood. Present data adds new evidence of distinct effects of brain lateralization and sex vulnerability on biochemical, behavioral and histological parameters after hypoxia-ischemia.


Assuntos
Encéfalo/patologia , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/psicologia , Animais , Animais Recém-Nascidos , Aprendizagem da Esquiva/fisiologia , Encéfalo/enzimologia , Lesões das Artérias Carótidas/enzimologia , Lesões das Artérias Carótidas/patologia , Lesões das Artérias Carótidas/psicologia , Feminino , Lateralidade Funcional/fisiologia , Hipóxia-Isquemia Encefálica/enzimologia , Masculino , Atividade Motora/fisiologia , Fibras Nervosas Mielinizadas/patologia , Ratos , Ratos Wistar , Fatores Sexuais , ATPase Trocadora de Sódio-Potássio/análise
14.
Eur J Neurosci ; 37(5): 816-30, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23279039

RESUMO

Cathepsin C (CC) (EC 3.4.14.1, dipeptidyl peptidase I) is a lysosomal cysteine protease that is required for the activation of several granule-associated serine proteases in vivo. CC has been shown to be constitutively expressed in various tissues, but the enzyme is hardly detectable in central nervous system (CNS) tissues. In the present study, we investigated the regional and cellular distribution of CC in normal, aging and pathological mouse brains. Immunoblotting failed to detect CC protein in whole brain tissues of normal mice, as previously described. However, low proteolytic activity of CC was detected in a brain region-dependent manner, and granular immunohistochemical signals were found in neuronal perikarya of particular brain regions, including the accessory olfactory bulb, the septum, CA2 of the hippocampus, a part of the cerebral cortex, the medial geniculate, and the inferior colliculus. In aged mice, the number of CC-positive neurons increased to some extent. The protein level of CC and its proteolytic activity showed significant increases in particular brain regions of mouse models with pathological conditions--the thalamus in cathepsin D-deficient mice, the hippocampus of ipsilateral brain hemispheres after hypoxic-ischemic brain injury, and peri-damaged portions of brains after penetrating injury. In such pathological conditions, the majority of the cells that were strongly immunopositive for CC were activated microglia. These lines of evidence suggest that CC is involved in normal neuronal function in certain brain regions, and also participates in inflammatory processes accompanying pathogenesis in the CNS.


Assuntos
Lesões Encefálicas/enzimologia , Encéfalo/enzimologia , Catepsina C/metabolismo , Hipóxia-Isquemia Encefálica/enzimologia , Fatores Etários , Animais , Encéfalo/patologia , Catepsina C/genética , Catepsina D/deficiência , Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Neurônios/metabolismo , Especificidade de Órgãos , Proteólise , Regulação para Cima
15.
J Neurochem ; 124(5): 735-46, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23199256

RESUMO

Vascular endothelial growth factor (VEGF), an angiogenic factor induced by hypoxia, also exerts direct effects on neural tissues. VEGF up-regulation after hypoxia coincides with expression of its two tyrosine kinase receptors Flt-1(VEGFR-1) and Flk-1 (KDR/VEGFR-2), which are the key mediators of physiological angiogenesis. We have recently shown that hypoxic-preconditioning (PC) leading to tolerance to hypoxia-ischemia in neonatal piglet brain resulted in increased expression of VEGF. In this study, we used a hypoxic-preconditioning model of ischemic tolerance to analyze the expression and cellular distribution of VEGF receptors and phosphorylation of cAMP-response element-binding protein (CREB) in newborn piglet brain. The response of Flt-1 and Flk-1 mRNA to PC alone was biphasic with peaks early (6 h) and late (1 week) after PC. The mRNA expression of Flt-1 and Flk-1 in piglets preconditioned 24 h prior to hypoxia-ischemia was significantly higher than non-preconditioned piglets and remained up-regulated up to 7 days. Furthermore, PC prior to hypoxia-ischemia significantly increased the protein levels of Flt-1 and Flk-1 compared with hypoxia-ischemia in a time-dependent manner. Double-immunolabeling indicated that both Flt-1 and Flk-1 are expressed in neurons and endothelial cells with a similar time course of expression following PC and that PC leads to the growth of new vessels. Finally, our data demonstrate that PC significantly phosphorylated and activated cAMP-response element-binding protein in nucleus. These results suggest that mechanism(s) initiated by PC can induce VEGF receptor up-regulation in newborn brain and that VEGF-VEGF receptor-coupled signal transduction pathways could contribute to the establishment of tolerance following hypoxia-ischemia.


Assuntos
Hipóxia-Isquemia Encefálica/enzimologia , Precondicionamento Isquêmico , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Proteína de Ligação a CREB/metabolismo , Imuno-Histoquímica , Fosforilação , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Suínos
16.
Exp Neurol ; 236(1): 50-7, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22531298

RESUMO

Research into neonatal ischemic brain damage is impeded by the lack of a complete understanding of the initial hemodynamic mechanisms resulting in a lesion, particularly that of NO-mediated vascular mechanisms. In a neonatal stroke rat model, we recently show that collateral recruitment contributes to infarct size variability. Non-specific and selective NO synthase (NOS) inhibition was evaluated on cerebral blood-flow changes and outcome in a P7 rat model of arterial occlusion (left middle cerebral artery electrocoagulation with 50 min occlusion of both common carotid arteries). Blood-flow changes were measured by using ultrasound imaging with sequential Doppler recordings in both internal carotid arteries and basilar trunk. Cortical perfusion was measured by using laser Doppler flowmetry. We showed that global NOS inhibition significantly reduced collateral support and cortical perfusion (collateral failure), and worsened the ischemic injury in both gender. Conversely, endothelial NOS inhibition increased blood-flows and aggravated volume lesion in males, whereas in females blood-flows did not change and infarct lesion was significantly reduced. These changes were associated with decreased phosphorylation of neuronal NOS at Ser(847) in males and increased phosphorylation in females at 24h, respectively. Neuronal NOS inhibition also increased blood-flows in males but not in females, and did not significantly change infarct volumes compared to their respective PBS-treated controls. In conclusion, both nNOS and eNOS appear to play a key role in modulating arterial blood flow during ischemia mainly in male pups with subsequent modifications in infarct lesion.


Assuntos
Infarto Encefálico/enzimologia , Circulação Cerebrovascular/fisiologia , Hipóxia-Isquemia Encefálica/enzimologia , Leucomalácia Periventricular/enzimologia , Óxido Nítrico Sintase Tipo III/fisiologia , Óxido Nítrico Sintase Tipo I/fisiologia , Animais , Animais Recém-Nascidos , Infarto Encefálico/patologia , Infarto Encefálico/fisiopatologia , Circulação Cerebrovascular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Humanos , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Recém-Nascido , Leucomalácia Periventricular/patologia , Leucomalácia Periventricular/fisiopatologia , Masculino , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/química , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/química , Ratos , Ratos Wistar
17.
Braz. j. med. biol. res ; 45(1): 13-19, Jan. 2012. ilus, tab
Artigo em Inglês | LILACS | ID: lil-610548

RESUMO

Neonatal Sprague-Dawley rats were randomly divided into normal control, mild hypoxia-ischemia (HI), and severe HI groups (N = 10 in each group at each time) on postnatal day 7 (P7) to study the effect of mild and severe HI on anxiety-like behavior and the expression of tyrosine hydroxylase (TH) in the substantia nigra (SN). The mild and severe HI groups were exposed to hypoxia (8 percent O2/92 percent N2) for 90 and 150 min, respectively. The elevated plus-maze (EPM) test was performed to assess anxiety-like behavior by measuring time spent in the open arms (OAT) and OAT percent, and immunohistochemistry was used to determine the expression of TH in the SN at P14, P21, and P28. OAT and OAT percent in the EPM were significantly increased in both the mild (1.88-, 1.99-, and 2.04-fold, and 1.94-, 1.51-, and 1.46-fold) and severe HI groups (1.69-, 1.68-, and 1.87-fold, and 1.83-, 1.43-, and 1.39-fold, respectively; P < 0.05). The percent of TH-positive cells occupying the SN area was significantly and similarly decreased in both the mild (17.7, 40.2, and 47.2 percent) and severe HI groups (16.3, 32.2, and 43.8 percent, respectively; P < 0.05). The decrease in the number of TH-positive cells in the SN and the level of protein expression were closely associated (Pearson correlation analysis: r = 0.991, P = 0.000 in the mild HI group and r = 0.974, P = 0.000 in the severe HI group) with the impaired anxiety-like behaviors. We conclude that neonatal HI results in decreased anxiety-like behavior during the juvenile period of Sprague-Dawley rats, which is associated with the decreased activity of TH in the SN. The impairment of anxiety and the expression of TH are not likely to be dependent on the severity of HI.


Assuntos
Animais , Feminino , Ratos , Ansiedade/metabolismo , Comportamento Animal/fisiologia , Hipóxia-Isquemia Encefálica/metabolismo , Neurônios/enzimologia , Substância Negra/enzimologia , /metabolismo , Animais Recém-Nascidos , Ansiedade/enzimologia , Hipóxia-Isquemia Encefálica/enzimologia , Imuno-Histoquímica , Ratos Sprague-Dawley , Índice de Gravidade de Doença , /análise
18.
Apoptosis ; 17(5): 463-74, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22183510

RESUMO

Growing lines of evidence suggests that high mobility group box-1 (HMGB1) plays an important role for promoting inflammation and apoptosis in brain ischemia. Previously, we demonstrated that inducers of heme oxygenase-1 (HO-1) significantly reduce HMGB1 release in inflammatory conditions in vitro and in vivo. Thus, we tested our hypothesis that higenamine protects brain injury by inhibition of middle cerebral artery occlusion (MCAO)-mediated HMGB1 release in vivo, and glucose/glucose oxidase (GOX)-induced apoptosis in C6 cells in vitro due to HO-1 induction. Higenamine increased HO-1 expression in C6 cells in both hypoxia and normoxia, in which the former was much more significant than the latter. Higenamine increased Nrf-2 luciferase activity, translocated Nrf-2 to nucleus, and increased phosphorylation of Akt in C6 cells. Consistent with this, LY 294002, a PI3K inhibitor, inhibited HO-1 induction by higenamine and apoptosis induced by glucose/GOX in C6 cells was prevented by higenamine, which effect was reversed by LY 294002. Importantly, administration of higenamine (i.p) significantly reduced brain infarct size, mortality rate, MPO activity and tissue expression of HMGB1 in MCAO rats. In addition, recombinant high mobility group box 1 induced apoptosis in C6 cells by increasing ratio of Bax/bcl-2 and cleaved caspase c, which was inhibited by higenamine, and all of these effects were reversed by co-treatment with ZnPPIX. Therefore, we conclude that higenamine, at least in part, protects brain cells against hypoxic damages by up-regulation of HO-1. Thus, higenamine may be beneficial for the use of ischemic injuries such as stroke.


Assuntos
Alcaloides/farmacologia , Indução Enzimática/efeitos dos fármacos , Proteína HMGB1/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais , Tetra-Hidroisoquinolinas/farmacologia , Alcaloides/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Hipóxia Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cromonas/farmacologia , Proteína HMGB1/farmacologia , Hipóxia-Isquemia Encefálica/enzimologia , Hipóxia-Isquemia Encefálica/patologia , Infarto da Artéria Cerebral Média , Masculino , Morfolinas/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Peroxidase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Tetra-Hidroisoquinolinas/uso terapêutico
19.
Anesth Analg ; 114(1): 200-4, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22075021

RESUMO

BACKGROUND: Cerebral hypoxia/ischemia during infant congenital heart surgery is not uncommon and may induce devastating neurologic disabilities persistent over the lifespan. Hypoxia/ischemia-induced cerebrovascular dysfunction is thought to be an important contributor to neurological damage. No pharmacological agents have been found to prevent this. Mitogen activated protein kinase (MAPK), including extracellular signal regulated kinase (ERK), c-Jun-N-terminal kinase, and p38, is thought to contribute to ischemic preconditioning. We investigated whether pretreatment with salvinorin A, the only natural nonopioid κ receptor agonist, could preserve autoregulation of the pial artery via MAPK. METHODS: The response of the pial artery to hypotension and hypercapnia was monitored in piglets equipped with a closed cranial window before and after hypoxia and ischemia in the presence or absence of U0126, an inhibitor for the protein kinase upstream of ERK, sp600125, an inhibitor of c-Jun-N-terminal kinase or sb203580, an inhibitor of p38. Salvinorin A (10 µg/kg IV) was administered 30 minutes before hypoxia/ischemia in salvinorin-treated animals. Cerebrospinal fluid samples were collected before and 30 minutes after salvinorin A administration for the measurement of MAPK. Data (n = 5) were analyzed by repeated-measures analysis of variance. RESULTS: Pial artery dilation to hypercapnia and hypotension was blunted after hypoxia/ ischemia but preserved well by pretreatment with salvinorin A. U0126, but not sp600125 or sb203580, abolished the preservative effects of salvinorin A on cerebral vascular autoregulation to hypotension and hypercapnia. The ratio of pERK/ERK in cerebrospinal fluid increased significantly in salvinorin-treated animals, which was inhibited by U0126. CONCLUSIONS: Salvinorin A pretreatment preserves autoregulation of the pial artery to hypotension and hypercapnia after hypoxia/ischemia via ERK in a piglet model.


Assuntos
Circulação Cerebrovascular/efeitos dos fármacos , Diterpenos Clerodânicos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Pia-Máter/irrigação sanguínea , Vasodilatadores/farmacologia , Animais , Modelos Animais de Doenças , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/líquido cefalorraquidiano , Homeostase , Hipercapnia/tratamento farmacológico , Hipercapnia/enzimologia , Hipercapnia/fisiopatologia , Hipotensão/tratamento farmacológico , Hipotensão/enzimologia , Hipotensão/fisiopatologia , Hipóxia-Isquemia Encefálica/líquido cefalorraquidiano , Hipóxia-Isquemia Encefálica/enzimologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Inibidores de Proteínas Quinases/farmacologia , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/metabolismo , Suínos , Vasodilatação/efeitos dos fármacos
20.
J Perinat Med ; 39(4): 441-4, 2011 07.
Artigo em Inglês | MEDLINE | ID: mdl-21410409

RESUMO

Several experimental models on adult and newborn animals showed that in cerebral hypoxic-ischemic conditions similar to clinical states the main source of the excessive production of free oxygen radicals is the highly activated xanthine oxidase (XO) enzyme reaction. Long before this data were available, it became known that the main role of allopurinol (AP) is the inhibition of XO. On the basis of these results, many therapeutic trials with AP were performed both in experimental and clinical studies of ischemia and reperfusion. However, it has been shown that only preventive administration of AP has favorable effects. The explanation for the poor results of AP treatment in human fetal brain damage (FBD) cases is that the drug was applied postnatally. The clinical studies performed in healthy laboring mothers whose deliveries were complicated with FBD showed that placental transfer after prenatal administration of AP may be effective in protecting newborns at increased risk of hypoxic-ischemic cerebral damage. Further controlled trials are required to determine if the prophylactic use of the drug might prevent hypoxic-ischemic injuries when the drug is administered immediately prior to impending fetal hypoxia, or even in deliveries at risk of developing hypoxia.


Assuntos
Alopurinol/uso terapêutico , Lesões Encefálicas/prevenção & controle , Hipóxia Fetal/tratamento farmacológico , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Xantina Oxidase/antagonistas & inibidores , Alopurinol/administração & dosagem , Animais , Animais Recém-Nascidos , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/enzimologia , Lesões Encefálicas/etiologia , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/uso terapêutico , Feminino , Hipóxia Fetal/complicações , Hipóxia Fetal/enzimologia , Humanos , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/enzimologia , Recém-Nascido , Gravidez , Traumatismo por Reperfusão/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA