Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Stroke ; 50(11): 3228-3237, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31526124

RESUMEN

Background and Purpose- Recanalization of the occluded artery is a primary goal in stroke treatment. Unfortunately, endovascular treatment is not always available, and tPA (tissue-type plasminogen activator) therapy is limited by its narrow therapeutic window; importantly, the rate of early arterial recanalization after tPA administration is low, especially for platelet-rich thrombi. The mechanisms for this tPA resistance are not well known. Since neutrophil extracellular traps (NETs) have been implicated in this setting, our aim was to study whether NET pharmacological modulation can reverse tPA resistance and the role of TLR4 (Toll-like receptor 4), previously related to NET formation, in thrombosis. Methods- To this goal, we have used a mouse photothrombotic stroke model, which produces a fibrin-free thrombus composed primarily of aggregated platelets and thrombi obtained from human stroke patients. Results- Our results demonstrate that (1) administration of DNase-I, which promotes NETs lysis, but not of tPA, recanalizes the occluded vessel improving photothrombotic stroke outcome; (2) a preventive treatment with Cl-amidine, impeding NET formation, completely precludes thrombotic occlusion; (3) platelet TLR4 mediates NET formation after photothrombotic stroke; and (4) ex vivo fresh platelet-rich thrombi from ischemic stroke patients are effectively lysed by DNase-I. Conclusions- Hence, our data open new avenues for recanalization of platelet-rich thrombi after stroke, especially to overcome tPA resistance.


Asunto(s)
Desoxirribonucleasa I/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Trampas Extracelulares/metabolismo , Accidente Cerebrovascular , Trombosis , Activador de Tejido Plasminógeno/farmacología , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Transgénicos , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Trombosis/tratamiento farmacológico , Trombosis/metabolismo , Trombosis/patología , Receptor Toll-Like 4/metabolismo
2.
Mol Ther ; 26(8): 2047-2059, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29910175

RESUMEN

Since Toll-like receptor 4 (TLR4) mediates brain damage after stroke, development of TLR4 antagonists is a promising therapeutic strategy for this disease. Our aim was to generate TLR4-blocking DNA aptamers to be used for stroke treatment. From a random oligonucleotide pool, we identified two aptamers (ApTLR#1R, ApTLR#4F) with high affinity for human TLR4 by systematic evolution of ligands by exponential enrichment (SELEX). Optimized truncated forms (ApTLR#1RT, ApTLR#4FT) were obtained. Our data demonstrate specific binding of both aptamers to human TLR4 as well as a TLR4 antagonistic effect. ApTLR#4F and ApTLR#4FT showed a long-lasting protective effect against brain injury induced by middle cerebral artery occlusion (MCAO), an effect that was absent in TLR4-deficient mice. Similar effects were obtained in other MCAO models, including in rat. Additionally, efficacy of ApTLR#4FT in a model of brain ischemia-reperfusion in rat supports the use of this aptamer in patients undergoing artery recanalization induced by pharmacological or mechanical interventions. The absence of major toxicology aspects and the good safety profile of the aptamers further encourage their future clinical positioning for stroke therapy and possibly other diseases in which TLR4 plays a deleterious role.


Asunto(s)
Aptámeros de Nucleótidos/administración & dosificación , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Accidente Cerebrovascular/prevención & control , Receptor Toll-Like 4/metabolismo , Animales , Aptámeros de Nucleótidos/farmacología , Modelos Animales de Enfermedad , Humanos , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/etiología , Ratones , Ratas , Técnica SELEX de Producción de Aptámeros , Transducción de Señal , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo
3.
Rapid Commun Mass Spectrom ; 32(9): 721-729, 2018 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-29484723

RESUMEN

RATIONALE: Stroke is a leading cause of disability worldwide. Understanding the recovery process post-stroke is essential; however, longer-term recovery studies are lacking. In vivo positron emission tomography (PET) can image biological recovery processes, but is limited by spatial resolution and its targeted nature. Untargeted mass spectrometry imaging offers high spatial resolution, providing an ideal ex vivo tool for brain recovery imaging. METHODS: Magnetic resonance imaging (MRI) was used to image a rat brain 48 h after ischaemic stroke to locate the infarcted regions of the brain. PET was carried out 3 months post-stroke using the tracers [18 F]DPA-714 for TSPO and [18 F]IAM6067 for sigma-1 receptors to image neuroinflammation and neurodegeneration, respectively. The rat brain was flash-frozen immediately after PET scanning, and sectioned for matrix-assisted laser desorption/ionisation mass spectrometry (MALDI-MS) imaging. RESULTS: Three months post-stroke, PET imaging shows minimal detection of neurodegeneration and neuroinflammation, indicating that the brain has stabilised. However, MALDI-MS images reveal distinct differences in lipid distributions (e.g. phosphatidylcholine and sphingomyelin) between the scar and the healthy brain, suggesting that recovery processes are still in play. It is currently not known if the altered lipids in the scar will change on a longer time scale, or if they are stabilised products of the brain post-stroke. CONCLUSIONS: The data demonstrates the ability to combine MALD-MS with in vivo PET to image different aspects of stroke recovery.


Asunto(s)
Encéfalo/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Accidente Cerebrovascular/diagnóstico por imagen , Animales , Encéfalo/metabolismo , Encéfalo/patología , Lisofosfatidilcolinas/análisis , Imagen por Resonancia Magnética/métodos , Fosfatidilcolinas/análisis , Pirazoles , Pirimidinas , Ratas Wistar , Esfingomielinas/análisis , Accidente Cerebrovascular/patología , Factores de Tiempo
4.
Stroke ; 48(6): 1695-1699, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28428349

RESUMEN

BACKGROUND AND PURPOSE: Hemorrhagic transformation is the main complication of revascularization therapies after stroke. Toll-like receptor 4 (TLR4) is implicated in cerebral damage and inflammation in stroke. This study was designed to determine the role of TLR4 in hemorrhagic transformation development after tissue plasminogen activator (tPA) administration. METHODS: Mice expressing (TLR4+/+) or lacking functional TLR4 (TLR4-/-) were subjected to middle cerebral artery occlusion using an in situ thromboembolic model by thrombin injection into the middle cerebral artery, and tPA (10 mg/kg) was administered 20 minutes or 3 hours after ischemia. Infarct size, hemorrhages, IgG extravasation, matrix metalloproteinase 9 expression, and neutrophil infiltration were assessed 24 hours after ischemia. RESULTS: In TLR4+/+, early reperfusion (tPA at 20 minutes) resulted infarct volume, whereas late recanalization (tPA at 3 hours) did not modify lesion size and increased the rate of the most severe hemorrhages. In TLR4-/- mice, both early and late reperfusion did not modify lesion size. Importantly, late tPA administration did not result in worse hemorrhages and in an increased bleeding area as occurred in TLR4+/+ group. In TLR4-/- animals, late reperfusion produced a lesser increase in matrix metalloproteinase 9 expression when compared with TLR4+/+ animals. CONCLUSIONS: Our results demonstrate TLR4 involvement in hemorrhagic transformation induced by delayed tPA administration, very likely by increasing matrix metalloproteinase 9 expression.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Hemorragia Cerebral/metabolismo , Fibrinolíticos/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Activador de Tejido Plasminógeno/farmacología , Receptor Toll-Like 4/metabolismo , Animales , Isquemia Encefálica/etiología , Isquemia Encefálica/metabolismo , Hemorragia Cerebral/inducido químicamente , Infarto Cerebral/tratamiento farmacológico , Infarto Cerebral/metabolismo , Modelos Animales de Enfermedad , Fibrinolíticos/administración & dosificación , Infarto de la Arteria Cerebral Media/complicaciones , Embolia Intracraneal/complicaciones , Trombosis Intracraneal/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/metabolismo , Factores de Tiempo , Activador de Tejido Plasminógeno/administración & dosificación
5.
Stroke ; 48(1): 204-212, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27899748

RESUMEN

BACKGROUND AND PURPOSE: Stroke is a leading cause of adult disability characterized by physical, cognitive, and emotional disturbances. Unfortunately, pharmacological options are scarce. The cannabinoid type-2 receptor (CB2R) is neuroprotective in acute experimental stroke by anti-inflammatory mechanisms. However, its role in chronic stroke is still unknown. METHODS: Stroke was induced by permanent middle cerebral artery occlusion in mice; CB2R modulation was assessed by administering the CB2R agonist JWH133 ((6aR,10aR)-3-(1,1-dimethylbutyl)-6a,7,10,10a-tetrahydro-6,6,9-trimethyl-6H-dibenzo[b,d]pyran) or the CB2R antagonist SR144528 (N-[(1S)-endo-1,3,3-trimethylbicyclo-[2.2.1]-heptan-2-yl]-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)-pyrazole-3-carboxamide) once daily from day 3 to the end of the experiment or by CB2R genetic deletion. Analysis of immunofluorescence-labeled brain sections, 5-bromo-2´-deoxyuridine (BrdU) staining, fluorescence-activated cell sorter analysis of brain cell suspensions, and behavioral tests were performed. RESULTS: SR144528 decreased neuroblast migration toward the boundary of the infarct area when compared with vehicle-treated mice 14 days after middle cerebral artery occlusion. Consistently, mice on this pharmacological treatment, like mice with CB2R genetic deletion, displayed a lower number of new neurons (NeuN+/BrdU+ cells) in peri-infarct cortex 28 days after stroke when compared with vehicle-treated group, an effect accompanied by a worse sensorimotor performance in behavioral tests. The CB2R agonist did not affect neurogenesis or outcome in vivo, but increased the migration of neural progenitor cells in vitro; the CB2R antagonist alone did not affect in vitro migration. CONCLUSIONS: Our data support that CB2R is fundamental for driving neuroblast migration and suggest that an endocannabinoid tone is required for poststroke neurogenesis by promoting neuroblast migration toward the injured brain tissue, increasing the number of new cortical neurons and, conceivably, enhancing motor functional recovery after stroke.


Asunto(s)
Neurogénesis/fisiología , Receptor Cannabinoide CB2/fisiología , Recuperación de la Función/fisiología , Accidente Cerebrovascular/fisiopatología , Animales , Canfanos/farmacología , Cannabinoides/farmacología , Cannabinoides/uso terapéutico , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Pirazoles/farmacología , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/antagonistas & inhibidores , Recuperación de la Función/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Resultado del Tratamiento
6.
Brain Behav Immun ; 61: 117-126, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27856349

RESUMEN

Neuroprotective strategies for ischemic stroke have failed to translate from bench to bedside, possibly due to the lack of consideration of key clinical co-morbidities. Stroke and co-morbidities are associated with raised levels of the pro-inflammatory cytokine interleukin-1 (IL-1). Inhibition of IL-1 by the administration of interleukin-1 receptor antagonist (IL-1Ra) has shown to be neuroprotective after experimental cerebral ischemia. Stroke can also trigger a robust neuroreparative response following injury, yet many of these new born neurons fail to survive or integrate into pre-existing circuits. Thus, we explore here effects of IL-1Ra on post-stroke neurogenesis in young and aged/co-morbid rats. Aged lean, aged Corpulent (a model of atherosclerosis, obesity and insulin resistance) and young Wistar male rats were exposed to transient cerebral ischemia, received subcutaneous IL-1Ra 3 and 6h during reperfusion, and effects on stroke outcome and neurogenesis were analyzed. Our results show that administration of IL-1Ra improves stroke outcome in both young and aged/co-morbid rats. Furthermore, IL-1Ra not only increases stem cell proliferation, but also significantly enhances neuroblast migration and the number of newly born neurons after cerebral ischemia. Overall, our data demonstrate that systemic administration of IL-1Ra improves outcome and promotes neurogenesis after experimental stroke, further highlighting the therapeutic potential of this clinically approved drug.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Neurogénesis/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Movimiento Celular/efectos de los fármacos , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Masculino , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Wistar , Factores de Tiempo
7.
J Neuroinflammation ; 12: 87, 2015 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-25958332

RESUMEN

BACKGROUND: Aging is not just a risk factor of stroke, but it has also been associated with poor recovery. It is known that stroke-induced neurogenesis is reduced but maintained in the aged brain. However, there is no consensus on how neurogenesis is affected after stroke in aged animals. Our objective is to determine the role of aging on the process of neurogenesis after stroke. METHODS: We have studied neurogenesis by analyzing proliferation, migration, and formation of new neurons, as well as inflammatory parameters, in a model of cerebral ischemia induced by permanent occlusion of the middle cerebral artery in young- (2 to 3 months) and middle-aged mice (13 to 14 months). RESULTS: Aging increased both microglial proliferation, as shown by a higher number of BrdU(+) cells and BrdU/Iba1(+) cells in the ischemic boundary and neutrophil infiltration. Interestingly, aging increased the number of M1 monocytes and N1 neutrophils, consistent with pro-inflammatory phenotypes when compared with the alternative M2 and N2 phenotypes. Aging also inhibited (subventricular zone) SVZ cell proliferation by decreasing both the number of astrocyte-like type-B (prominin-1(+)/epidermal growth factor receptor (EGFR)(+)/nestin(+)/glial fibrillary acidic protein (GFAP)(+) cells) and type-C cells (prominin-1(+)/EGFR(+)/nestin(-)/Mash1(+) cells), and not affecting apoptosis, 1 day after stroke. Aging also inhibited migration of neuroblasts (DCX(+) cells), as indicated by an accumulation of neuroblasts at migratory zones 14 days after injury; consistently, aged mice presented a smaller number of differentiated interneurons (NeuN(+)/BrdU(+) and GAD67(+) cells) in the peri-infarct cortical area 14 days after stroke. CONCLUSIONS: Our data confirm that stroke-induced neurogenesis is maintained but reduced in aged animals. Importantly, we now demonstrate that aging not only inhibits proliferation of specific SVZ cell subtypes but also blocks migration of neuroblasts to the damaged area and decreases the number of new interneurons in the cortical peri-infarct area. Thus, our results highlight the importance of using aged animals for translation to clinical studies.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular/fisiología , Corteza Cerebral/patología , Infarto de la Arteria Cerebral Media/patología , Microglía/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Envejecimiento , Animales , Células Sanguíneas/patología , Células Sanguíneas/fisiología , Infarto Encefálico/etiología , Infarto Encefálico/patología , Bromodesoxiuridina/metabolismo , Proteínas de Unión al Calcio/metabolismo , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Proteína Doblecortina , Proteína Ácida Fibrilar de la Glía/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microglía/patología , Neuronas/patología , Infiltración Neutrófila/fisiología , Fosfopiruvato Hidratasa/metabolismo
8.
Ann Neurol ; 75(5): 670-83, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24644058

RESUMEN

OBJECTIVE: Bacterial infection contributes to diverse noninfectious diseases and worsens outcome after stroke. Streptococcus pneumoniae, the most common infection in patients at risk of stroke, is a major cause of prolonged hospitalization and death of stroke patients, but how infection impacts clinical outcome is not known. METHODS: We induced sustained pulmonary infection by a human S. pneumoniae isolate in naive and comorbid rodents to investigate the effect of infection on vascular and inflammatory responses prior to and after cerebral ischemia. RESULTS: S. pneumoniae infection triggered atherogenesis, led to systemic induction of interleukin (IL) 1, and profoundly exacerbated (50-90%) ischemic brain injury in rats and mice, a response that was more severe in combination with old age and atherosclerosis. Systemic blockade of IL-1 with IL-1 receptor antagonist (IL-1Ra) fully reversed infection-induced exacerbation of brain injury and functional impairment caused by cerebral ischemia. We show that infection-induced systemic inflammation mediates its effects via increasing platelet activation and microvascular coagulation in the brain after cerebral ischemia, as confirmed by reduced brain injury in response to blockade of platelet glycoprotein (GP) Ibα. IL-1 and platelet-mediated signals converge on microglia, as both IL-1Ra and GPIbα blockade reversed the production of IL-1α by microglia in response to cerebral ischemia in infected animals. INTERPRETATION: S. pneumoniae infection augments atherosclerosis and exacerbates ischemic brain injury via IL-1 and platelet-mediated systemic inflammation. These mechanisms may contribute to diverse cardio- and cerebrovascular pathologies in humans.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Interleucina-1/efectos adversos , Complejo GPIb-IX de Glicoproteína Plaquetaria/efectos adversos , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/patología , Streptococcus pneumoniae , Animales , Isquemia Encefálica/microbiología , Progresión de la Enfermedad , Humanos , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Interleucina-1/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/microbiología , Microglía/patología , Activación Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/antagonistas & inhibidores , Complejo GPIb-IX de Glicoproteína Plaquetaria/fisiología , Ratas , Ratas Wistar , Infecciones Estreptocócicas/microbiología
9.
FASEB J ; 28(11): 4710-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25063846

RESUMEN

Toll-like receptor 4 (TLR4) mediates brain damage after stroke. Now our objective is to determine TLR4 involvement in stroke-induced neurogenesis. Stroke was induced by permanent middle cerebral artery occlusion in wild-type and TLR4-deficient mice. Stereological and densitometric analysis of immunofluorescence-labeled brain sections and FACS analysis of cell suspensions were performed. Our results show that subventricular zone (SVZ) cell proliferation after stroke depends on infarct size. Second, when comparing brains with similar lesions, TLR4 attenuated SVZ proliferation, as shown by a decrease in prominin-1(+)/EGFR(+)/nestin(-) cells (type-C cells) at 1-2 d, and in BrdU(+) cells at 7 d, in TLR4(+/+) vs. TLR4(-/-) mice. Interestingly, 7 d after the infarct, neuroblasts in TLR4(+/+) mice migrated farther distances, reaching areas closer to the lesion than those in TLR4-deficient mice. However, at 14 d, TLR4-deficient mice presented a higher number of neuroblasts in all migratory zones than the TLR4(+/+) counterparts, which suggests that TLR4 deficiency delays neuroblast migration. Consistently, TLR4(+/+) mice showed an increased number of interneurons (NeuN(+)/BrdU(+)/GAD67(+) cells) in peri-infarct cortex 14-28 d after stroke. Our data indicate that, despite a negative effect on SVZ cell proliferation, TLR4 plays an important role in stroke-induced neurogenesis by promoting neuroblasts migration and increasing the number of new cortical neurons after stroke.


Asunto(s)
Isquemia Encefálica/metabolismo , Movimiento Celular/fisiología , Neurogénesis/fisiología , Neuronas/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Isquemia Encefálica/inmunología , Proliferación Celular/fisiología , Inmunidad Innata/fisiología , Masculino , Ratones Endogámicos C57BL , Receptor Toll-Like 4/inmunología
10.
Stroke ; 44(8): 2333-7, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23723308

RESUMEN

BACKGROUND AND PURPOSE: Sirtuin 1 (SIRT1) is a member of NAD+-dependent protein deacetylases implicated in a wide range of cellular functions and has beneficial properties in pathologies including ischemia/reperfusion processes and neurodegeneration. However, no direct evidence has been reported on the direct implication of SIRT1 in ischemic stroke. The aim of this study was to establish the role of SIRT1 in stroke using an experimental model in mice. METHODS: Wild-type and Sirt1-/- mice were subjected to permanent focal ischemia by permanent ligature. In another set of experiments, wild-type mice were treated intraperitoneally with vehicle, activator 3 (SIRT1 activator, 10 mg/kg), or sirtinol (SIRT1 inhibitor, 10 mg/kg) for 10 minutes, 24 hours, and 40 hours after ischemia. Brains were removed 48 hours after ischemia for determining the infarct volume. Neurological outcome was evaluated using the modified neurological severity score. RESULTS: Exposure to middle cerebral artery occlusion increased SIRT1 expression in neurons of the ipsilesional mouse brain cortex. Treatment of mice with activator 3 reduced infarct volume, whereas sirtinol increased ischemic injury. Sirt1-/- mice displayed larger infarct volumes after ischemia than their wild-type counterparts. In addition, SIRT1 inhibition/deletion was concomitant with increased acetylation of p53 and nuclear factor κB (p65). CONCLUSIONS: These results support the idea that SIRT1 plays an important role in neuroprotection against brain ischemia by deacetylation and subsequent inhibition of p53-induced and nuclear factor κB-induced inflammatory and apoptotic pathways.


Asunto(s)
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Sirtuina 1/fisiología , Sirtuinas/fisiología , Acetilación , Alelos , Animales , Proteínas Reguladoras de la Apoptosis/fisiología , Encéfalo/patología , Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Infarto de la Arteria Cerebral Media/complicaciones , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/fisiología , Ratones , Ratones Noqueados , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Distribución Aleatoria , Transducción de Señal/genética , Método Simple Ciego , Sirtuina 1/deficiencia , Sirtuina 1/genética , Sirtuinas/administración & dosificación , Sirtuinas/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/biosíntesis , Regulación hacia Arriba/fisiología
11.
J Neurochem ; 126(6): 819-26, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23600725

RESUMEN

CDP-choline has shown neuroprotective effects in cerebral ischemia. In humans, although a recent trial International Citicoline Trial on Acute Stroke (ICTUS) has shown that global recovery is similar in CDP-choline and placebo groups, CDP-choline was shown to be more beneficial in some patients, such as those with moderate stroke severity and not treated with t-PA. Several mechanisms have been proposed to explain the beneficial actions of CDP-choline. We have now studied the participation of Sirtuin1 (SIRT1) in the neuroprotective actions of CDP-choline. Fischer rats and Sirt1⁻/⁻ mice were subjected to permanent focal ischemia. CDP-choline (0.2 or 2 g/kg), sirtinol (a SIRT1 inhibitor; 10 mg/kg), and resveratrol (a SIRT1 activator; 2.5 mg/kg) were administered intraperitoneally. Brains were removed 24 and 48 h after ischemia for western blot analysis and infarct volume determination. Treatment with CDP-choline increased SIRT1 protein levels in brain concomitantly to neuroprotection. Treatment with sirtinol blocked the reduction in infarct volume caused by CDP-choline, whereas resveratrol elicited a strong synergistic neuroprotective effect with CDP-choline. CDP-choline failed to reduce infarct volume in Sirt1⁻/⁻ mice. Our present results demonstrate a robust effect of CDP-choline like SIRT1 activator by up-regulating its expression. Our findings suggest that therapeutic strategies to activate SIRT1 may be useful in the treatment of stroke. Sirtuin 1 (SIRT1) is implicated in a wide range of cellular functions. Regarding stroke, there is no direct evidence. We have demonstrated that citicoline increases SIRT1 protein levels in brain concomitantly to neuroprotection. Citicoline fails to reduce infarct volume in Sirt1⁻/⁻ mice. Our findings suggest that therapeutic strategies acting on SIRT1 may be useful in the treatment of stroke.


Asunto(s)
Citidina Difosfato Colina/farmacología , Fármacos Neuroprotectores , Nootrópicos/farmacología , Sirtuina 1/biosíntesis , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Benzamidas/farmacología , Western Blotting , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Células Cultivadas , Sinergismo Farmacológico , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones , Ratones Noqueados , Monocitos/metabolismo , Naftoles/farmacología , Neuronas/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Resveratrol , Sirtuina 1/antagonistas & inhibidores , Estilbenos/farmacología
12.
Br J Pharmacol ; 180(4): 459-478, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36181002

RESUMEN

BACKGROUND AND PURPOSE: Reperfusion therapy is the standard of care for ischaemic stroke; however, there is a need to identify new therapeutic targets able to ameliorate cerebral damage. Neutrophil ß1 adrenoceptors (ß1AR) have been linked to neutrophil migration during exacerbated inflammation. Given the central role of neutrophils in cerebral damage during stroke, we hypothesize that ß1AR blockade will improve stroke outcomes. EXPERIMENTAL APPROACH: Rats were subjected to middle cerebral artery occlusion-reperfusion to evaluate the effect on stroke of the selective ß1AR blocker metoprolol (12.5 mg·kg-1 ) when injected i.v. 10 min before reperfusion. KEY RESULTS: Magnetic resonance imaging and histopathology analysis showed that pre-reperfusion i.v. metoprolol reduced infarct size. This effect was accompanied by reduced cytotoxic oedema at 24 h and vasogenic oedema at 7 days. Metoprolol-treated rats showed reduced brain neutrophil infiltration and those which infiltrated displayed a high proportion of anti-inflammatory phenotype (N2, YM1+ ). Additional inflammatory models demonstrated that metoprolol specifically blocked neutrophil migration via ß1AR and excluded a significant effect on the glia compartment. Consistently, metoprolol did not protect the brain in neutrophil-depleted rats upon stroke. In patients suffering an ischaemic stroke, ß1AR blockade by metoprolol reduced circulating neutrophil-platelet co-aggregates. CONCLUSIONS AND IMPLICATIONS: Our findings describe that ß1AR blockade ameliorates cerebral damage by targeting neutrophils, identifying a novel therapeutic target to improve outcomes in patients with stroke. This therapeutic strategy is in the earliest stages of the translational pathway and should be further explored.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Ratas , Animales , Metoprolol/farmacología , Metoprolol/uso terapéutico , Metoprolol/metabolismo , Neutrófilos/metabolismo , Enfermedades Neuroinflamatorias , Isquemia Encefálica/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Receptores Adrenérgicos/metabolismo
13.
J Cereb Blood Flow Metab ; 41(7): 1692-1706, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34152893

RESUMEN

Stroke affects primarily aged and co-morbid people, aspects not properly considered to date. Since angiogenesis/vasculogenesis are key processes for stroke recovery, we purposed to determine how different co-morbidities affect the outcome and angiogenesis/vasculogenesis, using a rodent model of metabolic syndrome, and by dynamic enhanced-contrast imaging (DCE-MRI) to assess its non-invasive potential to determine these processes. Twenty/twenty-two month-old corpulent (JCR:LA-Cp/Cp), a model of metabolic syndrome and lean rats were used. After inducing the experimental ischemia by transient MCAO, angiogenesis was analyzed by histology, vasculogenesis by determination of endothelial progenitor cells in peripheral blood by flow cytometry and evaluating their pro-angiogenic properties in culture and the vascular function by DCE-MRI at 3, 7 and 28 days after tMCAO. Our results show an increased infarct volume, BBB damage and an impaired outcome in corpulent rats compared with their lean counterparts. Corpulent rats also displayed worse post-stroke angiogenesis/vasculogenesis, outcome that translated in an impaired vascular function determined by DCE-MRI. These data confirm that outcome and angiogenesis/vasculogenesis induced by stroke in old rats are negatively affected by the co-morbidities present in the corpulent genotype and also that DCE-MRI might be a technique useful for the non-invasive evaluation of vascular function and angiogenesis processes.


Asunto(s)
Medios de Contraste , Infarto de la Arteria Cerebral Media/complicaciones , Imagen por Resonancia Magnética/métodos , Síndrome Metabólico/fisiopatología , Neovascularización Patológica/patología , Accidente Cerebrovascular/complicaciones , Enfermedades Vasculares/patología , Animales , Modelos Animales de Enfermedad , Masculino , Neovascularización Patológica/etiología , Ratas , Enfermedades Vasculares/etiología
14.
J Neurosci ; 29(12): 3875-84, 2009 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-19321784

RESUMEN

Peroxisome proliferator-activated receptors gamma (PPARgamma) are nuclear receptors with essential roles as transcriptional regulators of glucose and lipid homeostasis. PPARgamma are also potent anti-inflammatory receptors, a property that contributes to the neuroprotective effects of PPARgamma agonists in experimental stroke. The mechanism of these beneficial actions, however, is not fully elucidated. Therefore, we have explored further the actions of the PPARgamma agonist rosiglitazone in experimental stroke induced by permanent middle cerebral artery occlusion (MCAO) in rodents. Rosiglitazone induced brain 5-lipoxygenase (5-LO) expression in ischemic rat brain, concomitantly with neuroprotection. Rosiglitazone also increased cerebral lipoxin A(4) (LXA(4)) levels and inhibited MCAO-induced production of leukotriene B4 (LTB(4)). Furthermore, pharmacological inhibition and/or genetic deletion of 5-LO inhibited rosiglitazone-induced neuroprotection and downregulation of inflammatory gene expression, LXA(4) synthesis and PPARgamma transcriptional activity in rodents. Finally, LXA(4) caused neuroprotection, which was partly inhibited by the PPARgamma antagonist T0070907, and increased PPARgamma transcriptional activity in isolated nuclei, showing for the first time that LXA(4) has PPARgamma agonistic actions. Altogether, our data illustrate that some effects of rosiglitazone are attributable to de novo synthesis of 5-LO, able to induce a switch from the synthesis of proinflammatory LTB(4) to the synthesis of the proresolving LXA(4). Our study suggests novel lines of study such as the interest of lipoxin-like anti-inflammatory drugs or the use of these molecules as prognostic and/or diagnostic markers for pathologies in which inflammation is involved, such as stroke.


Asunto(s)
Araquidonato 5-Lipooxigenasa/fisiología , Lipoxinas/biosíntesis , Fármacos Neuroprotectores/farmacología , PPAR gamma/agonistas , Accidente Cerebrovascular/metabolismo , Tiazolidinedionas/farmacología , Animales , Araquidonato 5-Lipooxigenasa/biosíntesis , Araquidonato 5-Lipooxigenasa/genética , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Infarto Encefálico/etiología , Infarto Encefálico/metabolismo , Infarto Encefálico/patología , Infarto de la Arteria Cerebral Media/complicaciones , Leucotrieno B4/biosíntesis , Ratones , PPAR gamma/fisiología , Ratas , Rosiglitazona , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/patología
15.
Stroke ; 41(12): 2956-64, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21115947

RESUMEN

BACKGROUND AND PURPOSE: The endocannabinoid system has been involved in the modulation of neural stem cells proliferation, survival and differentiation as well as in the generation of new oligodendrocyte progenitors in the postnatal brain. The present work aims to test the effect of the synthetic Type 1 and Type 2 cannabinoid receptor agonist WIN55212-2 on these processes in the context of neonatal rat brain hypoxia-ischemia (HI). METHODS: P7 Wistar rats were subjected to HI and treated either with WIN55212-2 (1 mg/kg) or vehicle twice daily for 7 days after HI and euthanized at 1, 2, 7, 14, or 28 days to explore white matter injury progression and the neurogenic response in the subventricular zone after HI. RESULTS: Our findings reveal that WIN55212-2 promotes remyelination of the injured external capsule, increasing the number of NG2+ early oligodendrocyte progenitors 7 days after HI in this area and the number of APC+ mature oligodendrocytes in the injured striatum 14 and 28 days after HI. WIN55212-2 also increases cell proliferation and protein expression of the neuroblast marker doublecortin in the subventricular zone 7 days after neonatal HI as well as the number of newly generated neuroblasts (5-bromodeoxyuridine+/doublecortin+ cells) in the ipsilateral striatum 14 days after HI. CONCLUSIONS: Our results suggest that the activation of the endocannabinoid system promotes white and gray matter recovery after neonatal HI injury.


Asunto(s)
Animales Recién Nacidos/fisiología , Benzoxazinas/uso terapéutico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Morfolinas/uso terapéutico , Naftalenos/uso terapéutico , Regeneración Nerviosa/efectos de los fármacos , Receptores de Cannabinoides/efectos de los fármacos , Animales , Antimetabolitos , Western Blotting , Encéfalo/patología , Bromodesoxiuridina , Recuento de Células , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Proteína Doblecortina , Femenino , Técnica del Anticuerpo Fluorescente , Hipoxia-Isquemia Encefálica/patología , Inmunohistoquímica , Oligodendroglía/efectos de los fármacos , Embarazo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
J Neurochem ; 109(1): 287-94, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19200341

RESUMEN

It has been demonstrated that a short ischemic event (ischemic preconditioning, IPC) results in a subsequent resistance to severe ischemia (ischemic tolerance, IT). We have recently demonstrated the role of innate immunity and in particular of toll-like receptor (TLR) 4 in brain ischemia. Several evidences suggest that TLR4 might also be involved in IT. Therefore, we have now used an in vivo model of IPC to investigate whether TLR4 is involved in IT. A 6-min temporary bilateral common carotid arteries occlusion was used for focal IPC and it was performed on TLR4-deficient mice (C57BL/10ScNJ) and animals that express TLR4 normally (C57BL/10ScSn). To assess the ability of IPC to induce IT, permanent middle cerebral artery occlusion was performed 48 h after IPC. Stroke outcome was evaluated by determination of infarct volume and assessment of neurological scores. IPC caused neuroprotection as shown by a reduction in infarct volume and better outcome in mice expressing TLR4 normally. TLR4-deficient mice showed less IPC-induced neuroprotection than wild-type animals. Western blot analysis of tumor necrosis factor alpha (TNF-alpha), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) showed an up-regulation in the expression of these proteins in both substrains of mice measured 18, 24 and 48 h after IPC, being higher in mice with TLR4. Similarly, nuclear factor-kappa B (NF-kappaB) activation was observed 18, 24 and 48 h after IPC, being more intense in TLR4-expressing mice. These data demonstrate that TLR4 signalling is involved in brain tolerance as shown by the difference in the percentage of neuroprotection produced by IPC between ScSn and ScNJ (60% vs. 18%). The higher expression of TNF-alpha, iNOS and cyclooxygenase-2 and NF-kappaB activation in mice expressing TLR4 is likely to participate in this endogenous neuroprotective effect.


Asunto(s)
Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/prevención & control , Precondicionamiento Isquémico , Receptor Toll-Like 4/fisiología , Animales , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Infarto de la Arteria Cerebral Media/enzimología , Precondicionamiento Isquémico/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/fisiología , Fármacos Neuroprotectores/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Transducción de Señal/genética , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
17.
Circulation ; 115(12): 1599-608, 2007 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-17372179

RESUMEN

BACKGROUND: Stroke is the second to third leading cause of death. Toll-like receptor 4 (TLR4) is a signaling receptor in innate immunity that is a specific immunologic response to systemic bacterial infection and cerebral injury. The role of TLR4 in brain ischemia has not been examined yet. We have therefore investigated whether cerebral ischemia and inflammation produced by permanent occlusion of the middle cerebral artery differ in mice that lack a functional TLR4 signaling pathway. METHODS AND RESULTS: Permanent occlusion of the middle cerebral artery was performed on 2 strains of TLR4-deficient mice (C3H/HeJ and C57BL/10ScNJ) and respective controls (C3H/HeN and C57BL/10ScSn). Stroke outcome was evaluated by determination of infarct volume and assessment of neurological scores. Brains were collected 24 hours and 7 days after stroke. When compared with control mice, TLR4-deficient mice had lower infarct volumes and better outcomes in neurological and behavioral tests. Mice that lacked TLR4 had minor expression of stroke-induced interferon regulatory factor-1, inducible nitric oxide synthase, and cyclooxygenase-2, mediators implicated in brain damage. The levels of interferon-beta and of the lipid peroxidation marker malondialdehyde were also lower in brains from TLR4-deficient mice than in those from control mice. In addition, the expression of matrix metalloproteinase-9, which is induced and mediates brain damage, was also reduced in TLR4-deficient mice after experimental stroke. CONCLUSIONS: TLR4-deficient mice have minor infarctions and less inflammatory response after an ischemic insult. These data demonstrate that TLR4 signaling and innate immunity are involved in brain damage and in inflammation triggered by ischemic injury.


Asunto(s)
Daño Encefálico Crónico/patología , Infarto de la Arteria Cerebral Media/metabolismo , Mediadores de Inflamación/metabolismo , Receptor Toll-Like 4/fisiología , Animales , Química Encefálica , Daño Encefálico Crónico/etiología , Daño Encefálico Crónico/metabolismo , Daño Encefálico Crónico/prevención & control , Infarto Cerebral/etiología , Infarto Cerebral/metabolismo , Infarto Cerebral/patología , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Dinoprostona/análisis , Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Encefalitis/metabolismo , Encefalitis/patología , Regulación de la Expresión Génica , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Factor 1 Regulador del Interferón/biosíntesis , Factor 1 Regulador del Interferón/genética , Interferones/biosíntesis , Interferones/genética , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Peroxidación de Lípido , Masculino , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Nitrobencenos/uso terapéutico , Estrés Oxidativo , Sulfonamidas/uso terapéutico , Receptor Toll-Like 4/deficiencia , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
18.
Stroke ; 39(4): 1314-20, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18309167

RESUMEN

BACKGROUND AND PURPOSE: Psychological stress causes an inflammatory response in the brain and is able to exacerbate brain damage caused by experimental stroke. We previously reported that subacute immobilization stress in mice worsens stroke outcome through mechanisms that involve inflammatory mechanisms, such as accumulation of oxidative/nitrosative mediators and expression of inducible nitric oxide synthase and cyclooxygenase-2 in the brain. Some of these inflammatory mediators could be regulated by innate immunity, the activation of which takes place in the brain and produces an inflammatory response mediated by toll-like receptors (TLRs). Recently, we described the implications of TLR4 in ischemic injury, but the role of TLR4 in stress has not yet been examined. We therefore investigated whether inflammation produced by immobilization stress differs in mice that lack a functional TLR4 signaling pathway. METHODS: We used an experimental paradigm consisting of the exposure of mice to repeated immobilization sessions (1 hour daily for 7 days) before permanent middle cerebral artery occlusion. RESULTS: We found that TLR4-deficient mice subjected to subacute stress had a better behavioral condition compared with normal mice (C3H/HeN) and that this effect was associated with a minor inflammatory response (cyclooxygenase-2 and inducible nitric oxide synthase expression) and lipid peroxidation (malondialdehyde levels) in brain tissue. Furthermore, previous exposure to stress was followed by a smaller infarct volume after permanent middle cerebral artery occlusion in TLR4-deficient mice than in mice that express TLR4 normally. CONCLUSIONS: Our results indicate that TLR4 is involved in the inflammatory response after subacute stress and its exacerbating effect on stroke. These data implicate the effects of innate immunity on inflammation and damage in the brain after stroke.


Asunto(s)
Isquemia Encefálica/inmunología , Encefalitis/inmunología , Estrés Psicológico/inmunología , Accidente Cerebrovascular/inmunología , Receptor Toll-Like 4/inmunología , Animales , Conducta Animal , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Corticosterona/sangre , Ciclooxigenasa 2/genética , Encefalitis/complicaciones , Encefalitis/patología , Expresión Génica/inmunología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Mutantes , Neuroinmunomodulación/fisiología , Óxido Nítrico Sintasa de Tipo II/genética , Peroxidasa/metabolismo , Recuperación de la Función , Restricción Física , Transducción de Señal/inmunología , Estrés Psicológico/complicaciones , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/patología , Receptor Toll-Like 4/genética
19.
J Cereb Blood Flow Metab ; 38(12): 2150-2164, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30129391

RESUMEN

Ischemic brain injury causes a local inflammatory response, involving the activation of resident brain cells such as microglia and the recruitment of infiltrating immune cells. Increasing evidence supports that plasticity of the myeloid cell lineage is determinant for the specific role of these cells on stroke outcome, from initiation and maintenance to resolution of post-ischemic inflammation. The aim of this review is to summarize some of the key characteristics of these cells and the mechanisms for their recruitment into the injured brain through interactions with platelets, endothelial cells and other leukocytes. Also, we discuss the existence of different leukocyte subsets in the ischemic tissue and, specifically, the impact of different myeloid phenotypes on stroke outcome, with special emphasis on neutrophils and their interplay with platelets. Knowledge of these cellular phenotypes and interactions may pave the way to new therapies able to promote protective immune responses and tissue repair after cerebral ischemia.


Asunto(s)
Células Mieloides/patología , Neuroinmunomodulación/fisiología , Accidente Cerebrovascular/patología , Animales , Humanos , Inflamación/inmunología , Inflamación/patología , Células Mieloides/inmunología , Accidente Cerebrovascular/inmunología
20.
J Cereb Blood Flow Metab ; 36(3): 596-605, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26661169

RESUMEN

Stroke represents a global challenge and is a leading cause of permanent disability worldwide. Despite much effort, translation of research findings to clinical benefit has not yet been successful. Failure of neuroprotection trials is considered, in part, due to the low quality of preclinical studies, low level of reproducibility across different laboratories and that stroke co-morbidities have not been fully considered in experimental models. More rigorous testing of new drug candidates in different experimental models of stroke and initiation of preclinical cross-laboratory studies have been suggested as ways to improve translation. However, to our knowledge, no drugs currently in clinical stroke trials have been investigated in preclinical cross-laboratory studies. The cytokine interleukin 1 is a key mediator of neuronal injury, and the naturally occurring interleukin 1 receptor antagonist has been reported as beneficial in experimental studies of stroke. In the present paper, we report on a preclinical cross-laboratory stroke trial designed to investigate the efficacy of interleukin 1 receptor antagonist in different research laboratories across Europe. Our results strongly support the therapeutic potential of interleukin 1 receptor antagonist in experimental stroke and provide further evidence that interleukin 1 receptor antagonist should be evaluated in more extensive clinical stroke trials.


Asunto(s)
Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Receptores de Interleucina-1/antagonistas & inhibidores , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/inmunología , Edema Encefálico/patología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/inmunología , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores de Interleucina-1/inmunología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA