Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
BMC Mol Biol ; 9: 62, 2008 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-18611280

RESUMO

BACKGROUND: Traumatic brain injury models are widely studied, especially through gene expression, either to further understand implied biological mechanisms or to assess the efficiency of potential therapies. A large number of biological pathways are affected in brain trauma models, whose elucidation might greatly benefit from transcriptomic studies. However the suitability of reference genes needed for quantitative RT-PCR experiments is missing for these models. RESULTS: We have compared five potential reference genes as well as total cDNA level monitored using Oligreen reagent in order to determine the best normalizing factors for quantitative RT-PCR expression studies in the early phase (0-48 h post-trauma (PT)) of a murine model of diffuse brain injury. The levels of 18S rRNA, and of transcripts of beta-actin, glyceraldehyde-3P-dehydrogenase (GAPDH), beta-microtubulin and S100beta were determined in the injured brain region of traumatized mice sacrificed at 30 min, 3 h, 6 h, 12 h, 24 h and 48 h post-trauma. The stability of the reference genes candidates and of total cDNA was evaluated by three different methods, leading to the following rankings as normalization factors, from the most suitable to the less: by using geNorm VBA applet, we obtained the following sequence: cDNA(Oligreen); GAPDH > 18S rRNA > S100beta > beta-microtubulin > beta-actin; by using NormFinder Excel Spreadsheet, we obtained the following sequence: GAPDH > cDNA(Oligreen) > S100beta > 18S rRNA > beta-actin > beta-microtubulin; by using a Confidence-Interval calculation, we obtained the following sequence: cDNA(Oligreen) > 18S rRNA; GAPDH > S100beta > beta-microtubulin > beta-actin. CONCLUSION: This work suggests that Oligreen cDNA measurements, 18S rRNA and GAPDH or a combination of them may be used to efficiently normalize qRT-PCR gene expression in mouse brain trauma injury, and that beta-actin and beta-microtubulin should be avoided. The potential of total cDNA as measured by Oligreen as a first-intention normalizing factor with a broad field of applications is highlighted. Pros and cons of the three methods of normalization factors selection are discussed. A generic time- and cost-effective procedure for normalization factor validation is proposed.


Assuntos
Lesões Encefálicas/genética , Perfilação da Expressão Gênica/normas , Reação em Cadeia da Polimerase Via Transcriptase Reversa/normas , Actinas/genética , Animais , DNA Complementar/análise , DNA Complementar/normas , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/genética , Camundongos , Modelos Animais , Fatores de Crescimento Neural/genética , RNA Ribossômico 18S/genética , Padrões de Referência , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/genética , Tubulina (Proteína)/genética , Estudos de Validação como Assunto
2.
J Neurosci ; 25(41): 9367-77, 2005 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-16221845

RESUMO

Acute brain injuries have been identified as a risk factor for developing Alzheimer's disease (AD). Because glutamate plays a pivotal role in these pathologies, we studied the influence of glutamate receptor activation on amyloid-beta (Abeta) production in primary cultures of cortical neurons. We found that sublethal NMDA receptor activation increased the production and secretion of Abeta. This effect was preceded by an increased expression of neuronal Kunitz protease inhibitory domain (KPI) containing amyloid-beta precursor protein (KPI-APP) followed by a shift from alpha-secretase to beta-secretase-mediated APP processing. This shift is a result of the inhibition of the alpha-secretase candidate tumor necrosis factor-alpha converting enzyme (TACE) when associated with neuronal KPI-APPs. This KPI-APP/TACE interaction was also present in AD brains. Thus, our findings reveal a cellular mechanism linking NMDA receptor activation to neuronal Abeta secretion. These results suggest that even mild deregulation of the glutamatergic neurotransmission may increase Abeta production and represent a causal risk factor for developing AD.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Endopeptidases/metabolismo , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/metabolismo , Secretases da Proteína Precursora do Amiloide , Peptídeos beta-Amiloides/genética , Animais , Ácido Aspártico Endopeptidases , Células Cultivadas , Endopeptidases/genética , Agonistas de Aminoácidos Excitatórios/farmacologia , Humanos , Camundongos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Receptores de N-Metil-D-Aspartato/fisiologia
3.
J Neurosci Methods ; 144(2): 183-91, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15910976

RESUMO

Neuroinflammation is one of the events occurring after acute brain injuries. The aim of the present report was to characterize a rat model to study acute neuroinflammation on the histopathological, biochemical and functional outcomes. Lipopolysaccharide (LPS), known as a strong immunostimulant, was directly injected into the hippocampus. The spatiotemporal evolution of inducible NOS (iNOS) and cell death was studied from 6 h to 7 days. A perfect time course correlation was observed between iNOS immunoreactivity and iNOS activity showing an acute, expansive and transient iNOS induction in the hippocampus with a peak at 24 h. It was associated with a marked increase in NO metabolite (NO(x)) levels, and a high level of myeloperoxidase (MPO) activity. This inflammation precedes a massive cellular loss including at least neurons and astrocytes, and a drop of constitutive NOS activity, restrictive to the ipsilateral hippocampus from 48 h after LPS injection. Moreover, sensorimotor function impairment occurred from 24 h to 7 days with a maximum at 24 h post-LPS injection. Therefore, we characterized an in vivo model of acute neuroinflammation and neurodegeneration, in relation with a neurological deficit, which may be a powerful tool for mechanistic studies and for further evaluation of the potential neuroprotective agents.


Assuntos
Modelos Animais de Doenças , Encefalite/fisiopatologia , Mediadores da Inflamação/farmacologia , Lipopolissacarídeos/farmacologia , Degeneração Neural/fisiopatologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Encefalite/induzido quimicamente , Encefalite/patologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/fisiopatologia , Masculino , Transtornos dos Movimentos/patologia , Transtornos dos Movimentos/fisiopatologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Peroxidase/metabolismo , Ratos , Ratos Sprague-Dawley , Transtornos de Sensação/induzido quimicamente , Transtornos de Sensação/patologia , Transtornos de Sensação/fisiopatologia , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Fatores de Tempo , Regulação para Cima/fisiologia
4.
Brain Res ; 987(1): 32-8, 2003 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-14499943

RESUMO

The present work examined whether polymorphonuclear neutrophil (PMN) infiltration contributes to cortical and striatal brain damage and oxidative stress in a model of transient focal cerebral ischemia. A 2-h occlusion of the left middle cerebral artery and ipsilateral common carotid artery was performed in rats. Administration of the neutropenic agent vinblastine (0.5 mg/kg, i.v.) resulted in a profound decrease in circulating PMNs which was associated with a 80% decrease in myeloperoxidase activity, a marker of PMN infiltration, in both the cortex and the striatum. In the cortex, vinblastine-treated animals exhibited a 44% decrease in the infarct volume and also reduced the oxidative stress (evaluated by the decrease in glutathione concentrations). By contrast, in the striatum, neutropenia modified neither the lesion size nor the oxidative stress. These results indicate that PMN contribution to postischemic injury and oxidative stress is dependent on the brain structure.


Assuntos
Isquemia Encefálica/metabolismo , Córtex Cerebral/metabolismo , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Estresse Oxidativo , Traumatismo por Reperfusão/metabolismo , Animais , Antineoplásicos Fitogênicos/farmacologia , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/enzimologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/enzimologia , Corpo Estriado/metabolismo , Masculino , Peroxidase/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/enzimologia , Vimblastina/farmacologia
5.
Brain Res ; 989(1): 58-66, 2003 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-14519512

RESUMO

Traumatic brain injury produces nitric oxide and reactive oxygen species. Peroxynitrite, resulting from the combination of nitric oxide and superoxide anions, triggers DNA strand breaks, leading to the activation of poly(ADP-ribose)polymerase-1. As excessive activation of this enzyme induces cell death, we examined the production of nitrosative stress, the activation of poly(ADP-ribose)polymerase-1, and the role of this enzyme in the outcomes of traumatic brain injury produced by fluid percussion in rats. Immunohistochemistry showed that 3-nitrotyrosine, an indicator of nitrosative stress, and poly(ADP-ribose), a marker of poly(ADP-ribose)polymerase-1 activation, were present as early as 30 min post-injury, and that persisted for 72 h. The poly(ADP-ribose)polymerase inhibitor, 3-aminobenzamide, at 10 and 30 mg/kg, significantly improved the neurological deficit, with a 60% reduction in the brain lesion volume and inhibition of poly(ADP-ribose)polymerase-1 activation. Thus, poly(ADP-ribose)polymerase-1 is involved in the neurological consequences of traumatic brain injury and may be a promising therapeutic target in clinical treatment of acute brain trauma.


Assuntos
Lesões Encefálicas/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Transdução de Sinais/fisiologia , Animais , Benzamidas/farmacologia , Benzamidas/uso terapêutico , Lesões Encefálicas/tratamento farmacológico , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Masculino , Inibidores de Poli(ADP-Ribose) Polimerases , Poli(ADP-Ribose) Polimerases/análise , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
6.
Eur J Pharmacol ; 483(2-3): 259-65, 2004 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-14729115

RESUMO

The present study examined caspase activation and its modulation by nitric oxide (NO) in a model of oxidative stress induced by injection of malonate (3 micromol), a mitochondrial toxin, into rat striatum. Caspase-3-like enzymatic activity was maximal 6 h after malonate while NO production evaluated by its metabolites nitrites and nitrates was increased at 3 h. The neuronal NO-synthase inhibitor 7-nitroindazole reduced malonate induced-NO production by 50% at 25 mg/kg and enhanced by 32% caspase activation. This result suggests that a moderate production of NO potentiates caspase activation, an effect counterbalanced by NO itself at higher concentrations. Accordingly, complete inhibition of NO production by 7-nitroindazole at 50 mg/kg did not modify malonate-induced caspase activity. Thus NO production by the neuronal isoform of NO-synthase is not the major event leading to caspase activation due to malonate. However, NO seems to have pro- and anti-caspase effects that neutralize each other.


Assuntos
Caspases/metabolismo , Corpo Estriado/efeitos dos fármacos , Malonatos/administração & dosagem , Óxido Nítrico/biossíntese , Animais , Inibidores de Caspase , Corpo Estriado/metabolismo , Indazóis/farmacologia , Masculino , Óxido Nítrico/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley
7.
Eur J Pharmacol ; 457(2-3): 137-46, 2002 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-12464359

RESUMO

The role of nitric oxide (NO) in the development of post-ischemic cerebral infarction has been extensively examined, but fewer studies have investigated its role in other outcomes. In the present study, we first determined the temporal evolution of infarct volume, NO production, neurological deficit and blood-brain barrier disruption in a model of transient focal cerebral ischemia in mice. We then examined the effect of the nonselective NO-synthase inhibitor N(omega)-nitro-L-arginine-methylester (L-NAME). L-NAME given at 3 mg/kg 3 h after ischemia reduced by 20% the infarct volume and abolished the increase in brain NO production evaluated by its metabolites (nitrites/nitrates) 48 h after ischemia. L-NAME with this protocol also reduced the neurological deficit evaluated by the grip test and decreased by 65% the extravasation of Evans blue, an index of blood-brain barrier breakdown. These protective activities of L-NAME suggest that NO has multiple deleterious effects in cerebral ischemia.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Isquemia Encefálica/complicações , Inibidores Enzimáticos/farmacologia , Infarto da Artéria Cerebral Média/prevenção & controle , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/biossíntese , Animais , Barreira Hematoencefálica/fisiologia , Encéfalo/enzimologia , Encéfalo/metabolismo , Química Encefálica/efeitos dos fármacos , Isquemia Encefálica/enzimologia , Isquemia Encefálica/fisiopatologia , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/etiologia , Masculino , Camundongos , Nitratos/análise , Óxido Nítrico Sintase/antagonistas & inibidores , Nitritos/análise
8.
Brain Res ; 1491: 78-87, 2013 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-23122881

RESUMO

The implication of cyclooxygenase (COX) type 2 in post-traumatic consequences is so far controversial. In experimental models of traumatic brain injury (TBI), genetic disruption or pharmacological inhibition of COX-2 has been shown to be neuroprotective, deleterious or without effect. Therefore, the aim of our study was to investigate the effect of COX-2 inhibition against neurological deficit and brain oedema after TBI that was induced by mechanical percussion in male Swiss mice. Despite the increased level and activity of COX-2, its inhibition either with nimesulide (12 mg/kg) or meloxicam (2mg/kg) modified neither the neurological score nor the brain water content that were evaluated at 6 and 24h after injury. Interestingly, the non-selective COX inhibition with indomethacin (5mg/kg) significantly promoted neurological recovery at 6 and 24h after trauma, without improving brain oedema. In conclusion, the present study yields considerable evidence that COX-2 may not solely constitute an interesting target for the treatment of TBI consequences. Our data point to a potentially deleterious role of COX-1 in the development of neurological impairment in brain-injured mice. However, the neuroprotective mechanism of indomethacin remains to be clarified.


Assuntos
Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Inibidores de Ciclo-Oxigenase/uso terapêutico , Traumatismos Cranianos Fechados/complicações , Traumatismos Cranianos Fechados/tratamento farmacológico , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/etiologia , 6-Cetoprostaglandina F1 alfa/biossíntese , Animais , Comportamento Animal/fisiologia , Western Blotting , Encéfalo/patologia , Edema Encefálico/patologia , Lesões Encefálicas/complicações , Lesões Encefálicas/tratamento farmacológico , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Comportamento Exploratório/fisiologia , Traumatismos Cranianos Fechados/enzimologia , Técnicas Imunoenzimáticas , Indometacina/uso terapêutico , Masculino , Camundongos , Doenças do Sistema Nervoso/patologia , Desempenho Psicomotor/fisiologia , Recuperação de Função Fisiológica , Especificidade por Substrato
9.
J Neurotrauma ; 28(10): 2135-43, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21770756

RESUMO

Traumatic brain injury (TBI) induces both focal and diffuse lesions that are concurrently responsible for the ensuing morbidity and mortality and for which no established treatment is available. It has been recently reported that an endogenous neuroprotector, the soluble form α of the amyloid precursor protein (sAPPα), exerts neuroprotective effects following TBI. However, the emergent post-traumatic neuroinflammatory environment compromises sAPPα production and may promote neuronal degeneration and consequent brain atrophy. Hence, the aim of this study was to examine the effects of the anti-inflammatory drug minocycline on sAPPα levels, as well as on long-term histological consequences post-TBI. The weight-drop model was used to induce TBI in mice. Minocycline or its vehicle were administered three times: at 5 min (90 mg/kg, i.p.) and at 3 and 9 h (45 mg/kg, i.p.) post-TBI. The levels of sAPPα, the extent of brain atrophy, and reactive gliosis were evaluated by ELISA, cresyl violet, and immunolabeling of GFAP and CD11b, respectively. Our results revealed a post-TBI sAPPα decrease that was significantly attenuated by minocycline. Additionally, corpus callosum and striatal atrophy, ventriculomegaly, astrogliosis, and microglial activation were observed at 3 months post-TBI. All the above consequences were significantly reduced by minocycline. In conclusion, inhibition of the acute phase of post-TBI neuroinflammation was associated with the sparing of sAPPα and the protection of brain tissue in the long-term, emphasizing the potential role of minocycline as an effective treatment for TBI.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Antibacterianos/farmacologia , Química Encefálica/efeitos dos fármacos , Lesões Encefálicas/tratamento farmacológico , Minociclina/farmacologia , Fármacos Neuroprotetores , Animais , Lesões Encefálicas/patologia , Antígeno CD11b/metabolismo , Ventrículos Cerebrais/patologia , Corpo Caloso/metabolismo , Corpo Caloso/patologia , Corpo Estriado/patologia , Ensaio de Imunoadsorção Enzimática , Gliose/patologia , Imuno-Histoquímica , Masculino , Camundongos
10.
J Neurotrauma ; 27(5): 911-21, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20166806

RESUMO

Traumatic brain injury (TBI) causes a wide spectrum of consequences, such as microglial activation, cerebral inflammation, and focal and diffuse brain injury, as well as functional impairment. In this study we aimed to investigate the effects of acute treatment with minocycline as an inhibitor of microglial activation on cerebral focal and diffuse lesions, and on the spontaneous locomotor activity following TBI. The weight-drop model was used to induce TBI in mice. Microglial activation and diffuse axonal injury (DAI) were detected by immunohistochemistry using CD11b and ss-amyloid precursor protein (ss-APP) immunolabeling, respectively. Focal injury was determined by the measurement of the brain lesion volume. Horizontal and vertical locomotor activities were measured for up to 12 weeks post-injury by an automated actimeter. Minocycline or vehicle were administered three times post-insult, at 5 min (90 mg/kg i.p.), 3 h, and 9 h post-TBI (45 mg/kg i.p.). Minocycline treatment attenuated microglial activation by 59% and reduced brain lesion volume by 58%, yet it did not affect DAI at 24 h post-TBI. More interestingly, minocycline significantly decreased TBI-induced locomotor hyperactivity at 48 h post-TBI, and its effect lasted for up to 8 weeks. Taken together, the results indicate that microglial activation appears to play an important role in the development of TBI-induced focal injury and the subsequent locomotor hyperactivity, and its short-term inhibition provides long-lasting functional recovery after TBI. These findings emphasize the fact that minocycline could be a promising new therapeutic strategy for head-injured patients.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Gliose/tratamento farmacológico , Traumatismos Cranianos Fechados/tratamento farmacológico , Microglia/efeitos dos fármacos , Microglia/metabolismo , Minociclina/farmacologia , Animais , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Seguimentos , Gliose/patologia , Traumatismos Cranianos Fechados/patologia , Hipercinese/tratamento farmacológico , Hipercinese/patologia , Hipercinese/prevenção & controle , Masculino , Camundongos , Microglia/patologia , Fármacos Neuroprotetores/farmacologia , Fatores de Tempo , Resultado do Tratamento
11.
Brain Res ; 1291: 122-32, 2009 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-19631631

RESUMO

One of the severe complications following traumatic brain injury (TBI) is cerebral edema and its effective treatment is of great interest to prevent further brain damage. This study investigated the effects of minocycline, known for its anti-inflammatory properties, on cerebral edema and its respective inflammatory markers by comparing different dose regimens, on oxidative stress and on neurological dysfunction following TBI. The weight drop model was used to induce TBI in mice. The brain water content was measured to evaluate cerebral edema. Inflammatory markers were detected by ELISA (IL-1beta), zymography and Western blot (MMP-9). The oxidative stress marker (glutathione levels) and neurological function were measured by Griffith technique and string test, respectively. Minocycline was administered i.p. once (5 min), twice (5 min and 3 h) or triple (5 min, 3 h and 9 h) following TBI. The first dose of minocycline only varied (45 or 90 mg/kg), whereas the following doses were all at 45 mg/kg. The single and double administrations of minocycline reduced the increase of inflammatory markers at 6 h post-TBI. Minocycline also reduced cerebral edema at this time point, only after double administration and at the high dose regimen, although with no effect on the TBI-induced oxidized glutathione increase. The anti-edematous effect of minocycline persisted up to 24 h, upon a triple administration, and accompanied by a neurological recovery. In conclusion, we reported an anti-edematous effect of minocycline after TBI in mice according to a specific treatment regimen. These findings emphasize that the beneficial effects of minocycline depend on the treatment regimen following a brain injury.


Assuntos
Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Lesões Encefálicas/complicações , Córtex Cerebral/metabolismo , Interleucina-1beta/metabolismo , Minociclina/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Análise de Variância , Animais , Anti-Inflamatórios/uso terapêutico , Western Blotting , Água Corporal , Edema Encefálico/metabolismo , Edema Encefálico/fisiopatologia , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/fisiopatologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiopatologia , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Exame Neurológico , Estresse Oxidativo/fisiologia
12.
Exp Neurol ; 184(2): 973-80, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14769390

RESUMO

Poly(ADP-ribose) polymerase (PARP) was shown to be detrimental in cerebral ischemia but the mechanisms whereby PARP is deleterious have yet to be determined. They may include a role in neutrophil infiltration known to aggravate ischemic damage. In this context, we investigated the effect of 3-aminobenzamide (3-AB), a PARP inhibitor, on brain damage and neutrophil infiltration after transient focal cerebral ischemia in mice. Ischemia was induced in male Swiss mice, anaesthetized with chloral hydrate (400 mg/kg, i.p.), by a 15-min-occlusion of the left middle cerebral artery using an intraluminal suture. Treatments with 3-AB were first administered intraperitoneally 15 min before reperfusion and endpoints measured at 24 h. Among the range of dosages studied (20-320 mg/kg), 40 mg/kg gave the maximal neuroprotection with a 30% decrease in the infarct volume and tended to improve the neurological score evaluated by a grip test. The same dosage was, however, devoid of effect when injection was delayed 2 or 6 h after reperfusion. Myeloperoxidase (MPO) activity used as an index of neutrophil infiltration showed that infiltration peaked 48 h after reperfusion in our model. At this time point, 3-AB (40 mg/kg given 15 min before reperfusion) markedly reduced the neutrophil infiltration, as evidenced by a 72%-decrease in MPO activity, and was still neuroprotective. Our results confirm that 3-AB reduces brain damage. Moreover, for the first time, a quantitative study shows that 3-AB decreases neutrophil infiltration elicited by cerebral ischemia.


Assuntos
Benzamidas/farmacologia , Infarto Encefálico/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Infiltração de Neutrófilos/efeitos dos fármacos , Animais , Infarto Encefálico/etiologia , Infarto Encefálico/patologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Ataque Isquêmico Transitório/complicações , Masculino , Camundongos , Peroxidase/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases , Reperfusão , Fatores de Tempo
13.
J Pharmacol Exp Ther ; 306(2): 588-94, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12730357

RESUMO

Nitric oxide (NO) and reactive oxygen species are both implicated in neuronal death due to cerebral ischemia. BN 80933, an original compound associating an inhibitor of neuronal NO synthase with an antioxidant, has been shown to reduce functional and histological damage in rat submitted to cerebral ischemia. The aim of the present study was to confirm these results in mice and to further examine the effects of BN 80933 on inflammatory response, including blood-brain barrier (BBB) disruption, brain edema, and neutrophil infiltration after transient middle cerebral artery occlusion (MCAO). Intravenous administration of BN 80933 at 3 and 10 mg/kg 3 h after MCAO significantly reduced by 26 to 36% the infarct volume evaluated 24 and 48 h after ischemia, and improved the neurological score. Furthermore, BN 80933 at both dosages decreased by 42 to 75% the extravasation of Evans blue in brain parenchyma observed 24 h after ischemia. This reduction in BBB disruption was associated with decreased brain edema as demonstrated by the 37% reduction in brain water content induced by BN 80933 at 3 mg/kg 24 h after MCAO. Neutrophil infiltration in brain parenchyma, evaluated by the myeloperoxidase activity, was also reduced by 45 to 56% in animals treated with BN 80933 at 3 and 10 mg/kg. Together, these results extend the protective capacity of BN 80933 against brain ischemic injury and confirm that BN 80933 represents a promising treatment for stroke.


Assuntos
Ataque Isquêmico Transitório/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Óxido Nítrico Sintase/antagonistas & inibidores , Pirazinas/uso terapêutico , Tiofenos/uso terapêutico , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Barreira Hematoencefálica/efeitos dos fármacos , Edema Encefálico/prevenção & controle , Infarto Encefálico/prevenção & controle , Infarto da Artéria Cerebral Média/enzimologia , Masculino , Camundongos , Doenças do Sistema Nervoso/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico Sintase Tipo I , Peroxidase/metabolismo , Pirazinas/farmacologia , Tiofenos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA