Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Adv Healthc Mater ; 10(11): e2100059, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33890419

RESUMEN

Oxidative stress, which is one of the main harmful mechanisms of pathologies including ischemic stroke, contributes to both neurons and endothelial cell damages, leading to vascular lesions. Although many antioxidants are tested in preclinical studies, no treatment is currently available for stroke patients. Since cerium oxide nanoparticles (CNPs) exhibit remarkable antioxidant capacities, the objective is to develop an innovative coating to enhance CNPs biocompatibility without disrupting their antioxidant capacities or enhance their toxicity. This study reports the synthesis and characterization of functional polymers and their impact on the enzyme-like catalytic activity of CNPs. To study the toxicity and the antioxidant properties of CNPs for stroke and particularly endothelial damages, in vitro studies are conducted on a cerebral endothelial cell line (bEnd.3). Despite their internalization in bEnd.3 cells, coated CNPs are devoid of cytotoxicity. Microscopy studies report an intracellular localization of CNPs, more precisely in endosomes. All CNPs reduces glutamate-induced intracellular production of reactive oxygen species (ROS) in endothelial cells but one CNP significantly reduces both the production of mitochondrial superoxide anion and DNA oxidation. In vivo studies report a lack of toxicity in mice. This study therefore describes and identifies biocompatible CNPs with interesting antioxidant properties for ischemic stroke and related pathologies.


Asunto(s)
Cerio , Nanopartículas , Animales , Antioxidantes/farmacología , Cerio/toxicidad , Células Endoteliales , Humanos , Ratones , Polímeros
2.
Front Pharmacol ; 11: 623, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32477119

RESUMEN

Comorbidity between drug abuse and post-traumatic stress disorder (PTSD), a stress-related dysregulation of fear responses, is very high. While some drugs are known to increase fear and anxiety, there are only few data regarding interactions between voluntary drug consumption and fear memory. The spontaneous chronic consumption of either alcohol or cocaine under a 3-week free-choice progressive paradigm of alcohol (3/6/10%) or cocaine (0.2/0.4/0.6 mg/ml), was assessed in VGV transgenic mice, having full 5-HT2C receptor editing and displaying PTSD-like behaviors. The consequences of these drug consumptions on the potentiated contextual and cued fear conditioning responses of VGV mice were assessed. The effects of drugs on hippocampal brain-derived neurotrophic factor (Bdnf) mRNA were measured as its expression was previously found to be decreased in VGV mice. Chronic alcohol consumption was similar in WT and VGV mice. In the alcohol condition, fear acquisition was not different at the end of the learning session and cue-fear extinction was facilitated. Regarding cocaine, in contrast to WT mice, VGV mice did not increase their drug consumption along with increasing doses, an effect that might be related with enhanced drug stimuli discrimination via increased 5-HT2C receptors. Cocaine-intake VGV mice did not display the contextual fear generalization usually observed in control VGV mice. In addition, Bdnf expression was upregulated after either chronic alcohol or cocaine intake. Altogether, these results suggest that both chronic alcohol and cocaine voluntary oral consumptions can exert some therapeutic-like effects in a mutant model of PTSD predisposition.

3.
Behav Brain Res ; 360: 69-80, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30500429

RESUMEN

Efforts are still needed regarding the research of therapeutics for ischemic stroke. While in experimental studies the protective effect of pharmacological agents is often highlighted by a reduction of the lesion size evaluated in the short term (days), in clinical studies a functional recovery of patients suffering from stroke is expected on the long-term (months and years). Long-term functional preclinical studies are highly recommended to evaluate potential neuroprotective agents for stroke, rather than an assessment of the infarction size at a short time point. The present study thus aimed to select among various behavioral tests those able to highlight long-term deficits (3 months) after cerebral ischemia in mice. Permanent focal cerebral ischemia was carried out in male Swiss mice by intraluminal occlusion of the left middle cerebral artery (MCA). Fourteen behavioral tests were assessed from 7 days to 90 days after ischemia (locomotor activity, neurological score, exit circle test, grip and string tests, chimney test, adhesive removal test, pole test, beam-walking tests, elevated plus maze, marble burying test, forced swimming test, novel object recognition test). The present study clearly identified a battery of behavioral tests able to highlight deficits up to 3 months in our mouse model of permanent MCA occlusion (locomotor activity, neurological score, adhesive removal test, pole test, beam-walking tests, elevated plus maze, marble burying test, forced swimming test and novel object recognition test). This battery of behavioral tests highlighting long-term deficits is useful to study future neuroprotective strategies for stroke treatment.


Asunto(s)
Isquemia Encefálica/complicaciones , Trastornos Mentales/diagnóstico , Trastornos Mentales/etiología , Animales , Isquemia Encefálica/tratamiento farmacológico , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Locomoción/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Examen Neurológico , Oxígeno/uso terapéutico , Desempeño Psicomotor , Estadísticas no Paramétricas , Natación
4.
Mol Neurobiol ; 55(12): 9156-9168, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29651748

RESUMEN

Benefits from thrombolysis with recombinant tissue plasminogen activator (rt-PA) after ischemic stroke remain limited due to a narrow therapeutic window, low reperfusion rates, and increased risk of hemorrhagic transformations (HT). Experimental data showed that rt-PA enhances the post-ischemic activation of poly(ADP-ribose)polymerase (PARP) which in turn contributes to blood-brain barrier injury. The aim of the present study was to evaluate whether PJ34, a potent PARP inhibitor, improves poor reperfusion induced by delayed rt-PA administration, exerts vasculoprotective effects, and finally increases the therapeutic window of rt-PA. Stroke was induced by thrombin injection (0.75 UI in 1 µl) in the left middle cerebral artery (MCA) of male Swiss mice. Administration of rt-PA (0.9 mg kg-1) or saline was delayed for 4 h after ischemia onset. Saline or PJ34 (3 mg kg-1) was given intraperitoneally twice, just after thrombin injection and 3 h later, or once, 3 h after ischemia onset. Reperfusion was evaluated by laser Doppler, vascular inflammation by immunohistochemistry of vascular cell adhesion molecule-1 (VCAM-1) expression, and vasospasm by morphometric measurement of the MCA. Edema, cortical lesion, and sensorimotor deficit were evaluated. Treatment with PJ34 improved rt-PA-induced reperfusion and promoted vascular protection including reduction in vascular inflammation (decrease in VCAM-1 expression), HT, and MCA vasospasm. Additionally, the combined treatment significantly reduced brain edema, cortical lesion, and sensorimotor deficit. In conclusion, the combination of the PARP inhibitor PJ34 with rt-PA after cerebral ischemia may be of particular interest in order to improve thrombolysis with an extended therapeutic window.


Asunto(s)
Fármacos Neuroprotectores/uso terapéutico , Fenantrenos/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Reperfusión , Accidente Cerebrovascular/tratamiento farmacológico , Trombosis/tratamiento farmacológico , Animales , Edema/complicaciones , Edema/tratamiento farmacológico , Edema/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Hemorragia/complicaciones , Hemorragia/tratamiento farmacológico , Hemorragia/patología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Inflamación/patología , Masculino , Ratones , Fármacos Neuroprotectores/farmacología , Fenantrenos/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteolisis/efectos de los fármacos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/patología , Trombosis/complicaciones , Trombosis/patología , Activador de Tejido Plasminógeno/administración & dosificación , Activador de Tejido Plasminógeno/farmacología , Activador de Tejido Plasminógeno/uso terapéutico , Resultado del Tratamiento , Vasoespasmo Intracraneal/complicaciones , Vasoespasmo Intracraneal/tratamiento farmacológico , Vasoespasmo Intracraneal/patología
5.
J Neurol Sci ; 370: 187-195, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27772757

RESUMEN

Thrombolysis with recombinant tissue plasminogen activator (rt-PA) is currently the only approved pharmacological strategy for acute ischemic stroke. However, rt-PA exhibits vascular toxicity mainly due to endothelial damage. To investigate the mechanisms underlying rt-PA-induced endothelial alterations, we assessed the role of rt-PA in the generation of endothelial microparticles (EMPs), emerging biological markers and effectors of endothelial dysfunction. The mouse brain-derived endothelial cell line bEnd.3 was used. Cells were treated with rt-PA at 20, 40 or 80µg/ml for 15 or 24h, and EMPs were quantified in the culture media using Annexin-V staining coupled with flow cytometry. Rt-PA enhanced EMP release from bEnd.3 cells with a maximal increase at the 40µg/ml dose for 24h (+78% compared to controls). Using tranexamic acid and aprotinin we demonstrated that plasmin is responsible for rt-PA-induced EMP release. The p38 MAPK inhibitor SB203580 and the poly(ADP-ribose)polymerase (PARP) inhibitor PJ34 also reduced rt-PA-induced EMP production, suggesting that p38 MAPK and PARP are downstream intracellular effectors of rt-PA/plasmin. Rt-PA also altered through plasmin the morphology and the confluence of bEnd.3 cells. By contrast, these changes did not implicate p38 MAPK and PARP. This study demonstrates that rt-PA induces the production of microparticles by cerebral endothelial cells, through plasmin, p38 MAPK and PARP pathways. Determining the phenotype of these EMPs to clarify their role on the endothelium in ischemic conditions could thus be of particular interest.


Asunto(s)
Micropartículas Derivadas de Células/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Fibrinolisina/metabolismo , Fibrinolíticos/farmacología , Activador de Tejido Plasminógeno/farmacología , Animales , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Micropartículas Derivadas de Células/metabolismo , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Células Endoteliales/patología , Ratones , Plasminógeno/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Exp Neurol ; 248: 416-28, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23876515

RESUMEN

Recombinant tissue plasminogen activator (rt-PA) is the only pharmacological treatment approved for thrombolysis in patients suffering from ischemic stroke, but its administration aggravates the risk of hemorrhagic transformations. Experimental data demonstrated that rt-PA increases the activity of poly(ADP-ribose)polymerase (PARP). The aim of the present study was to investigate whether PJ34, a potent (PARP) inhibitor, protects the blood-brain barrier components from rt-PA toxicity. In our mouse model of cerebral ischemia, administration of rt-PA (10 mg/kg, i.v.) 6h after ischemia aggravated the post-ischemic degradation of ZO-1, claudin-5 and VE-cadherin, increased the hemorrhagic transformations (assessed by brain hemoglobin content and magnetic resonance imaging). Furthermore, rt-PA also aggravated ischemia-induced functional deficits. Combining PJ34 with rt-PA preserved the expression of ZO-1, claudin-5 and VE-cadherin, reduced the hemorrhagic transformations and improved the sensorimotor performances. In vitro studies also demonstrated that PJ34 crosses the blood-brain barrier and may thus exert its protective effect by acting on endothelial and/or parenchymal cells. Thus, co-treatment with a PARP inhibitor seems to be a promising strategy to reduce rt-PA-induced vascular toxicity after stroke.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Fenantrenos/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Accidente Cerebrovascular/tratamiento farmacológico , Activador de Tejido Plasminógeno/efectos adversos , Animales , Barrera Hematoencefálica/patología , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encéfalo/patología , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Ratones , Fenantrenos/farmacología , Accidente Cerebrovascular/patología , Activador de Tejido Plasminógeno/farmacología , Activador de Tejido Plasminógeno/uso terapéutico
7.
Fundam Clin Pharmacol ; 27(4): 393-401, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22436003

RESUMEN

Recombinant tissue-type plasminogen activator (rt-PA) is presently the only pharmacological treatment approved for thrombolysis in patients suffering from ischemic stroke. Although reperfusion of ischemic tissue is essential, the use of rt-PA is limited due to its narrow therapeutic window and risk of hemorrhagic transformations. Recent studies have shown that rt-PA amplifies the post-ischemic activation of the nuclear enzyme poly(ADP-ribose)polymerase (PARP). This enzyme has been shown to contribute to both the breakdown of the blood brain barrier and spontaneous hemorrhagic transformations after ischemia. We therefore examined the capacity of PJ34 (N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-2-(N,N-dimethylamino) acetamide hydrochloride), a potent inhibitor of PARP, to reduce the hemorrhagic transformations that occur after rt-PA in mice with permanent focal cerebral ischemia. Ischemia was produced by intraluminal occlusion of the left middle cerebral artery and treated with vehicle, rt-PA (10 mg/kg, i.v., 6 h after occlusion) or rt-PA plus PJ34 (3, 6 or 12 mg/kg, i.p., at ischemia onset and 4 h later). Hemorrhagic transformations, neurological examination, and infarct volumes were evaluated 48 h after the onset of ischemia. Delayed administration of rt-PA resulted in increased hemorrhagic transformations and aggravated the neurological deficit. Giving PJ34 (3 mg/kg) markedly reduced the hemorrhagic transformations, an effect not owing to a modification of matrix metalloprotease activity. Furthermore, PJ34 improved the neurological functions of rt-PA-treated ischemic mice. To conclude, the PARP inhibitor PJ34 makes rt-PA safer in experimental ischemic stroke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Hemorragia/tratamiento farmacológico , Fenantrenos/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Activador de Tejido Plasminógeno/metabolismo , Animales , Isquemia Encefálica/metabolismo , Inhibidores Enzimáticos/farmacología , Hemorragia/metabolismo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Arteria Cerebral Media/efectos de los fármacos , Arteria Cerebral Media/metabolismo , Proteínas Recombinantes/farmacología
8.
Crit Care Med ; 39(10): 2300-7, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21666443

RESUMEN

OBJECTIVES: Traumatic brain injury causes deleterious brain edema, leading to high mortality and morbidity. Brain edema exacerbates neurologic deficits and may be attributable to the breakdown of endothelial cell junction protein, leukocyte infiltration, and matrix metalloproteinase activation. These all contribute to loss of blood-brain barrier integrity. The pleiotropic effects of statins, hydroxymethylglutaryl-coenzyme A reductase inhibitors, may inhibit posttraumatic brain edema. We therefore investigated the effect of acute simvastatin on neurologic deficits, cerebral edema, and its origins. DESIGN: Randomized laboratory animal study. SETTINGS: University-affiliated research laboratory. SUBJECTS: Male Sprague-Dawley rats. INTERVENTIONS: Rats were subjected to lateral fluid percussion traumatic brain injury. Our preliminary dose-effect study indicated that 37.5 mg/kg simvastatin, administered orally 1 hr and 6 hrs after traumatic brain injury, has the greatest anti-edematous effect. This dose was used to study its effects on brain edema and on its mechanisms. MEASUREMENTS AND MAIN RESULTS: We first assessed the effects of simvastatin 24 hrs after traumatic brain injury on brain edema, brain claudin-5 expression, and the vascular endothelial-cadherin (pTyr731)/total vascular endothelial-cadherin ratio, matrix metalloproteinase-9 activity, intercellular adhesion molecule-1 expression, and polymorphonuclear neutrophil infiltration. We also evaluated blood-brain barrier permeability by measuring Evans blue and fluorescein sodium salt extravasation into the cerebral parenchyma. We then investigated whether simvastatin reduces neurologic deficits, edema, and blood-brain barrier permeability earlier than 24 hrs; these effects were evaluated 6 hrs after traumatic brain injury. The anti-edematous effect of simvastatin 24 hrs after traumatic brain injury was associated with increased claudin-5 and decreased intercellular adhesion molecule-1, polymorphonuclear neutrophil infiltration, and blood-brain barrier permeability, with no effect on matrix metalloproteinase-9 activity or vascular endothelial-cadherin phosphorylation. Earlier, 6-hrs after traumatic brain injury, simvastatin reduced neurologic deficits, cerebral edema, and blood-brain barrier permeability. CONCLUSIONS: Simvastatin could be a new therapy for reducing posttraumatic edema by preventing damage to tight junctions and neutrophil infiltration into the parenchyma, thus preserving blood-brain barrier integrity.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Lesiones Encefálicas/complicaciones , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Simvastatina/farmacología , Animales , Antígenos CD/biosíntesis , Barrera Hematoencefálica/metabolismo , Edema Encefálico/etiología , Edema Encefálico/patología , Cadherinas/biosíntesis , Claudina-5 , Células Endoteliales/metabolismo , Molécula 1 de Adhesión Intercelular/biosíntesis , Masculino , Metaloproteinasa 9 de la Matriz/biosíntesis , Proteínas de la Membrana/biosíntesis , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Ratas , Ratas Sprague-Dawley
9.
J Neurosci Methods ; 191(2): 180-90, 2010 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-20600312

RESUMEN

Although intracerebral haemorrhage (ICH) entails the highest rates of mortality and disability of all stroke subtypes, efficient neuroprotective therapy is still needed. As functional recovery is a major endpoint in clinical trials, preclinical studies must demonstrate the potential of drugs to improve the sensorimotor and cognitive function of animals. In addition, behavioural studies should be performed on the long-term in order to truly mimic clinical needs. The aim of our study was to characterise a model of intracerebral haemorrhage using both histology and long-term behaviour. ICH was induced in rats by an intrastriatal injection of collagenase. Histology was performed 24h, 7 days and 2 months after ICH. Among a set of sensorimotor tests, we discriminate those able to reveal long-term deficits (up to 2 months) after cerebral haemorrhage. Our five behavioural tests (a neurological score, an adhesive removal test, two beam-walking tests and ipsilateral circling induced by dexamphetamine) proved to be effective in revealing sensorimotor deficits up to 35 days or more after cerebral haemorrhage. In conclusion, these behavioural tests appear of particular interest to screen protective agents that may exhibit benefits in patients who suffer ICH.


Asunto(s)
Encéfalo/patología , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/patología , Hemorragia Cerebral/inducido químicamente , Colagenasas/toxicidad , Evaluación de la Discapacidad , Animales , Encéfalo/irrigación sanguínea , Encéfalo/fisiopatología , Arterias Cerebrales/fisiopatología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/fisiopatología , Dextroanfetamina , Modelos Animales de Enfermedad , Cojera Animal/diagnóstico , Cojera Animal/etiología , Cojera Animal/fisiopatología , Masculino , Examen Neurológico , Ratas , Ratas Sprague-Dawley , Tiempo
10.
J Clin Invest ; 119(1): 182-92, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19104149

RESUMEN

Parkinson disease (PD) is a neurodegenerative disorder characterized by a loss of dopamine-containing neurons. Mounting evidence suggests that dopaminergic cell death is influenced by the innate immune system. However, the pathogenic role of the adaptive immune system in PD remains enigmatic. Here we showed that CD8+ and CD4+ T cells but not B cells had invaded the brain in both postmortem human PD specimens and in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD during the course of neuronal degeneration. We further demonstrated that MPTP-induced dopaminergic cell death was markedly attenuated in the absence of mature T lymphocytes in 2 different immunodeficient mouse strains (Rag1-/- and Tcrb-/- mice). Importantly, similar attenuation of MPTP-induced dopaminergic cell death was seen in mice lacking CD4 as well as in Rag1-/- mice reconstituted with FasL-deficient splenocytes. However, mice lacking CD8 and Rag1-/- mice reconstituted with IFN-gamma-deficient splenocytes were not protected. These data indicate that T cell-mediated dopaminergic toxicity is almost exclusively arbitrated by CD4+ T cells and requires the expression of FasL but not IFNgamma. Further, our data may provide a rationale for targeting the adaptive arm of the immune system as a therapeutic strategy in PD.


Asunto(s)
Encéfalo , Linfocitos T CD4-Positivos/inmunología , Degeneración Nerviosa , Enfermedad de Parkinson , Trastornos Parkinsonianos , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/citología , Encéfalo/inmunología , Encéfalo/patología , Linfocitos T CD4-Positivos/citología , Muerte Celular/fisiología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Proteína Ligando Fas/genética , Proteína Ligando Fas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Sistema Inmunológico/fisiología , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/patología , Neuronas/citología , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/patología , Trastornos Parkinsonianos/inmunología , Trastornos Parkinsonianos/patología
11.
Eur J Pharmacol ; 588(1): 52-7, 2008 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-18468597

RESUMEN

Hemorrhagic transformation is an aggravating event that occurs in 15 to 43% of patients suffering from ischemic stroke. This phenomenon due to blood-brain barrier breakdown appears to be mediated in part by matrix metalloproteinases (MMPs) among which MMP-2 and MMP-9 could be particularly involved. Recent experimental studies demonstrated that post-ischemic MMP-9 overexpression is regulated by poly(ADP-ribose)polymerase (PARP). In this context, our study aimed to evaluate the effect of PJ34 (N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-2-(N,N-dimethylamino)acetamide), a potent PARP inhibitor, on MMP-2 and MMP-9 levels and on hemorrhagic transformations in a model of permanent focal cerebral ischemia in mice. PJ34 (6.25-12.5 mg/kg, i.p.) was given at the time of ischemia onset and 4 h later. Hemorrhagic transformations, divided into microscopic and macroscopic hemorrhages, were counted 48 h after ischemia on 12 coronal brain slices. Microscopic and macroscopic hemorrhages were respectively reduced by 38% and 69% with 6.25 mg/kg PJ34. The anti-hemorrhagic effect of PJ34 was associated with a 57% decrease in MMP-9 overexpression assessed by gelatin zymography. No increase in MMP-2 activity was observed after ischemia in our model. The vascular protection achieved by PJ34 was associated with a reduction in the motor deficit (P<0.05) and in infarct volume (-31%, P<0.01). In conclusion, our study demonstrates for the first time that PJ34 reduces hemorrhagic transformations after cerebral ischemia. Thus this PARP inhibitor exhibits both anti-hemorrhagic and neuroprotective effects that may be of valuable interest for the treatment of stroke.


Asunto(s)
Isquemia Encefálica/patología , Hemorragia Cerebral/prevención & control , Inhibidores Enzimáticos/farmacología , Fenantrenos/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/psicología , Hemorragia Cerebral/etiología , Hemorragia Cerebral/patología , Infarto Cerebral/patología , Masculino , Metaloproteasas/metabolismo , Ratones , Actividad Motora/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos
12.
Brain Res ; 987(1): 32-8, 2003 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-14499943

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/metabolismo , Corteza Cerebral/metabolismo , Infiltración Neutrófila , Neutrófilos/metabolismo , Estrés Oxidativo , Daño por Reperfusión/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/enzimología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/enzimología , Cuerpo Estriado/metabolismo , Masculino , Peroxidasa/metabolismo , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/enzimología , Vinblastina/farmacología
13.
Exp Neurol ; 182(2): 446-54, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12895455

RESUMEN

Polymorphonuclear leukocytes (PMNs) were reported to contribute to ischemia-reperfusion-induced brain damage. The present work examined whether PMN infiltration is deleterious in a severe model of transient focal cerebral ischemia and in which part PMNs contribute to oxidative stress and nitric oxide (NO) production. A 20-min occlusion of the left middle cerebral artery and both common carotid arteries was performed in rats. Infarction was maximal 24 h after reperfusion, while accumulation of PMNs in infarcted tissue was not significant before 48 h. Moreover, neutropenia induced by vinblastine (0.5 mg/kg iv) significantly decreased by 60-80% PMN infiltration 48 h after reperfusion but did not reduce the infarct volume. Thus PMNs do not contribute to cerebral injury in our model. Furthermore, decreased PMN infiltration modified neither oxidative stress evaluated by glutathione concentrations nor NO synthase activities 48 h after reperfusion. In conclusion, our results suggest that PMNs are not involved in severe cerebral ischemia and that anti-PMN strategies may be inefficient in some pathological conditions.


Asunto(s)
Infarto Cerebral/fisiopatología , Ataque Isquémico Transitorio/fisiopatología , Neutrófilos/metabolismo , Óxido Nítrico Sintasa/metabolismo , Estrés Oxidativo , Animales , Antineoplásicos Fitogénicos/farmacología , Calcio/metabolismo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Infarto Cerebral/etiología , Infarto Cerebral/patología , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Glutatión/metabolismo , Ataque Isquémico Transitorio/complicaciones , Ataque Isquémico Transitorio/patología , Masculino , Neutropenia/inducido químicamente , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II , Ratas , Ratas Sprague-Dawley , Vinblastina/farmacología
14.
Brain Res ; 958(1): 166-75, 2002 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-12468042

RESUMEN

Oxidative stress, inducible nitric oxide synthase (iNOS) and neutrophils all contribute to post-ischemic brain damage. This study has determined the time courses of these three phenomena after ischemia in parallel with histological and functional outcomes. Ischemia was produced in rats by occluding the left middle cerebral artery and both common carotid arteries for 20 min. Regional cerebral blood flow (rCBF) rapidly decreased to 20% of its preischemic value during occlusion and stabilized at 60% following reperfusion. The striatal infarction was maximal 15 h after reperfusion (50+/-3 mm(3)), whereas the cortical infarction reached its maximum at 48 h (183+/-10 mm(3)). This drastic decrease in rCBF followed by incomplete reperfusion and massive infarction is, thus, extremely severe. The cortical infarction was strongly correlated with the neurologic deficit and loss of body weight. Oxidative stress, evaluated by the decrease in glutathione concentrations, appeared in the striatum at 6 h after reperfusion and in the cortex at 15 h. Calcium-independent NOS activity, considered as inducible NOS activity, was significantly enhanced at 24 h in the striatum and at 48 h in the cortex. Myeloperoxidase activity, a marker of neutrophil infiltration, was significantly increased at 48 h in both the striatum and cortex. These time courses show that the delayed iNOS activity and neutrophil infiltration that occur after the maturation of infarction in severe ischemia may not contribute to ischemic brain damage. By contrast, early oxidative stress may well be implicated in cerebral injury.


Asunto(s)
Infarto Cerebral/enzimología , Quimiotaxis de Leucocito/inmunología , Ataque Isquémico Transitorio/enzimología , Neutrófilos/inmunología , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo/inmunología , Animales , Peso Corporal/fisiología , Calcio/metabolismo , Señalización del Calcio/fisiología , Infarto Cerebral/inmunología , Infarto Cerebral/fisiopatología , Circulación Cerebrovascular/fisiología , Modelos Animales de Enfermedad , Radicales Libres/metabolismo , Glutatión/metabolismo , Infarto de la Arteria Cerebral Media/enzimología , Infarto de la Arteria Cerebral Media/inmunología , Infarto de la Arteria Cerebral Media/fisiopatología , Ataque Isquémico Transitorio/inmunología , Ataque Isquémico Transitorio/fisiopatología , Masculino , Peroxidasa/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA