Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Neuroscience ; 243: 22-32, 2013 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-23562577

RESUMEN

Hemeoxygenase (HO) is an enzymatic system that degrades heme. HO-1 is an inducible isoform whereas HO-2 is constitutive. Stroke strongly induces HO-1 expression but the underlying mechanisms are not fully elucidated. Cytokines that are up-regulated after ischemia, like interleukin (IL)-10, can induce HO-1 gene expression, which is positively regulated by the transcriptional activator nuclear factor erythroid 2-related factor 2 (Nrf2) and negatively regulated by the transcriptional repressor breast cancer type 1 susceptibility protein (BRCA1) associated C-terminal helicase 1 (Bach-1). While Nrf2 is activated after ischemia and drugs promoting Nrf2 activation increase HO-1 and are beneficial, the involvement of Bach-1 is unknown. Here we investigated mechanisms involved in HO-1 induction and evaluated the effects of HO activity inhibition in mouse permanent middle cerebral artery occlusion (pMCAO). HO-1 was induced after ischemia in IL-10-deficient mice suggesting that post-ischemic HO-1 induction was IL-10-independent. Attenuation of Bach-1 gene repression after ischemia was associated to enhanced HO-1 induction. Administration of the HO activity inhibitor zinc proto-porphyrin IX (ZnPP) i.p. 24h before pMCAO exacerbated ischemia-induced tumor necrosis factor-α (TNF-α) and IL-1ß, nitro-oxidative stress, and the presence of neutrophils at 8h, and increased infarct volume at day 4. However, ZnPP did not worsen ischemic damage when given 30min before pMCAO. ZnPP induced HO-1 expression in the cerebral vasculature at 24h, when it was still detected by high-performance liquid chromatography (HPLC) in plasma. While ZnPP was not found in brain tissue extracts of controls, it could be detected after ischemia, supporting that a small fraction of the injected drug can reach the tissue following blood-brain barrier breakdown. The deleterious effect of inhibiting HO activity in ischemia became apparent in the presence of ZnPP-induced HO-1, which is known to exert effects independent of its enzymatic activity. In conclusion, HO-1 induction after ischemia was associated to down-regulation of transcriptional repressor Bach-1, and induction of HO-1 when HO enzymatic activity was inhibited was related to worst outcome after brain ischemia.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/biosíntesis , Isquemia Encefálica/enzimología , Hemo-Oxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Western Blotting , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/fisiología , Hemo-Oxigenasa 1/genética , Inflamación/enzimología , Inflamación/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Protoporfirinas/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
2.
Neuroscience ; 182: 208-16, 2011 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-21402129

RESUMEN

BACKGROUND AND PURPOSE: Positron emission tomography (PET) studies in humans have used (11)C-flumazenil (FMZ) to assess neuronal viability after stroke. Here we aimed to study whether (11)C-FMZ binding was sensitive to neuronal damage in the acute phase following ischemia/reperfusion in the rat brain. EXPERIMENTAL PROCEDURES: Transient (2 h followed by reperfusion) and permanent intraluminal middle cerebral artery occlusion was carried out. (11)C-FMZ binding was studied by PET up to 24 h after the onset of ischemia. Tissue infarction was evaluated post-mortem at 24 h. Immunohistochemistry against a neuronal nuclei specific protein (NeuN) was performed to assess neuronal injury. RESULTS: No decrease in (11)C-FMZ binding was detected in the ipsilateral cortex up to 24 h post-ischemia in the model of transient occlusion despite the fact that rats developed cortical and striatal infarction, and neuronal injury was clearly apparent at this time. In contrast, (11)C-FMZ binding was significantly depressed in the ipsilateral cortex at 24 h following permanent ischemia. CONCLUSIONS: This finding evidences that (11)C-FMZ binding is not sensitive to neuronal damage on the acute phase of ischemia/reperfusion in the rat brain.


Asunto(s)
Isquemia Encefálica/diagnóstico por imagen , Isquemia Encefálica/metabolismo , Degeneración Nerviosa/diagnóstico por imagen , Degeneración Nerviosa/metabolismo , Daño por Reperfusión/diagnóstico por imagen , Daño por Reperfusión/metabolismo , Enfermedad Aguda , Animales , Sitios de Unión/fisiología , Isquemia Encefálica/fisiopatología , Modelos Animales de Enfermedad , Flumazenil , Masculino , Degeneración Nerviosa/fisiopatología , Tomografía de Emisión de Positrones/métodos , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/fisiopatología
3.
Neuroscience ; 167(3): 872-9, 2010 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-20206237

RESUMEN

Chondroitin sulfate (CS) is a glucosaminoglycan (GAG) currently used for the treatment of osteoarthritis because of its antiinflammatory and antiapoptotic actions. Recent evidence has revealed that those peripheral effects of CS may also have therapeutic interest in diseases of the CNS. Since neuroinflammation has been implicated in different neuronal pathologies, this study was planned to investigate how CS could modulate the inflammatory response in the CNS by using rat astrocyte cultures stimulated with lipopolysaccharide (LPS). We have evaluated different proteins implicated in the nuclear factor kappa B (NFkappaB) and Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways employing RT-PCR, western blot and immunofluorescence techniques. At 10 microM, CS prevented translocation of p65 to the nucleus, reduced tumour necrosis factor alpha (TNF-alpha) mRNA and mitigated cyclooxygenase 2 (COX-2) and inducible nitric oxide synthase (iNOS) induction by LPS. However, it did not modify LPS-induced IP-10 and SOCS-1 mRNA, proteins that participate in the JAK/STAT pathway. The results of this study indicate that CS can potentially reduce neuroinflammation by inhibition of NFkappaB. Therefore endogenous GAGs could afford neuroimmunomodulatory actions under neurotoxic conditions.


Asunto(s)
Antiinflamatorios/farmacología , Astrocitos/efectos de los fármacos , Sulfatos de Condroitina/farmacología , Encefalitis/tratamiento farmacológico , Gliosis/tratamiento farmacológico , FN-kappa B/antagonistas & inhibidores , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Animales , Antiinflamatorios/uso terapéutico , Astrocitos/metabolismo , Células Cultivadas , Sulfatos de Condroitina/uso terapéutico , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Encefalitis/metabolismo , Encefalitis/fisiopatología , Gliosis/metabolismo , Gliosis/fisiopatología , Mediadores de Inflamación/farmacología , Janus Quinasa 1/efectos de los fármacos , Janus Quinasa 1/metabolismo , Lipopolisacáridos/farmacología , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Transcripción STAT/efectos de los fármacos , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor de Transcripción ReIA/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/genética
4.
Neuroscience ; 158(3): 1174-83, 2009 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-18619524

RESUMEN

UNLABELLED: Lymphocytes are major players in the development of innate and adaptive immune responses but their behavior in patients with acute stroke has received little attention. EXPERIMENTAL PROCEDURES: Using flow cytometry we identified total lymphocytes, T cells, helper T (Th) cells, cytotoxic T lymphocytes (CTL), natural killer (NK) cells, B cells, and regulatory T (Treg) cells in 46 consecutive patients with acute stroke within a median of 180 min of clinical onset, and at days 2, 7, and 90. Daily neurological score (National Institutes of Health Stroke Scale), diffusion-weighted imaging on brain magnetic resonance imaging, functional impairment, and stroke-associated infection (SAI) at day 7 were assessed. Apoptosis in lymphocyte subsets, tumor necrosis factor (TNF) -alpha/interleukin (IL) -4 production in stimulated Th and CTL, cluster of differentiation 86 (CD86) (B7-2) expression in B cells, cortisol and metanephrine in serum were measured. Multivariate analyses were used to evaluate SAI, and stroke outcome. RESULTS: Increased apoptosis and a fall of T, Th, CTL, B, and Treg cells were observed after stroke. Severer stroke on admission and SAI disclosed a greater decline of T, Th, and CTL cells. Increased cortisol and metanephrine was associated with severe stroke and SAI, and inversely correlated with T, and CTL. T cells, and CTL were correlated with infarct growth. Stroke but not SAI resulted in lower TNF-alpha production in Th cells. SAI showed the greatest fall of lymphocytes, T, Th, and CTL, but not B cells, or Treg. Poor outcome was associated with reduced levels of B cells, and increased expression of CD86 in B cells, but not with SAI. CONCLUSION: Lymphopenia and increased apoptosis of T, Th, CTL, Treg and B cells are early signatures after human stroke. A decreased cellular response after stroke is a marker of ongoing brain damage, the stress response, and a higher risk of infection. A lower humoral response is predictor of poorer long-term outcome.


Asunto(s)
Tolerancia Inmunológica/inmunología , Huésped Inmunocomprometido/inmunología , Linfocitos/inmunología , Linfopenia/inmunología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/inmunología , Enfermedad Aguda , Anciano , Anciano de 80 o más Años , Formación de Anticuerpos/inmunología , Apoptosis/inmunología , Biomarcadores/análisis , Citocinas/análisis , Citocinas/metabolismo , Femenino , Humanos , Hidrocortisona/análisis , Hidrocortisona/sangre , Recuento de Linfocitos , Linfocitos/citología , Linfopenia/diagnóstico , Linfopenia/fisiopatología , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Estudios Prospectivos , Estrés Fisiológico/inmunología , Accidente Cerebrovascular/fisiopatología
5.
Biochem Soc Trans ; 34(Pt 6): 1267-70, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17073799

RESUMEN

Stroke causes neuronal necrosis and generates inflammation. Pro-inflammatory molecules intervene in this process by triggering glial cell activation and leucocyte infiltration to the injured tissue. Cytokines are major mediators of the inflammatory response. Pro-inflammatory and anti-inflammatory cytokines are released in the ischaemic brain. Anti-inflammatory cytokines, such as interleukin-10, promote cell survival, whereas pro-inflammatory cytokines, such as TNFalpha (tumour necrosis factor alpha), can induce cell death. However, deleterious effects of certain cytokines can turn to beneficial actions, depending on particular features such as the concentration, time point and the very intricate network of intracellular signals that become activated and interact. A key player in the intracellular response to cytokines is the JAK (Janus kinase)/STAT (signal transducer and activator of transcription) pathway that induces alterations in the pattern of gene transcription. These changes are associated either with cell death or survival depending, among other things, on the specific proteins involved. STAT1 activation is related to cell death, whereas STAT3 activation is often associated with survival. Yet, it is clear that STAT activation must be tightly controlled, and for this reason the function of JAK/STAT modulators, such as SOCS (suppressors of cytokine signalling) and PIAS (protein inhibitor of activated STAT), and phosphatases is most relevant. Besides local effects in the ischaemic brain, cytokines are released to the circulation and affect the immune system. Unbalanced pro-inflammatory and anti-inflammatory plasma cytokine concentrations favouring an 'anti-inflammatory' state can decrease the immune response. Robust evidence now supports that stroke can induce an immunodepression syndrome, increasing the risk of infection. The contribution of individual cytokines and their intracellular signalling pathways to this response needs to be further investigated.


Asunto(s)
Isquemia Encefálica/fisiopatología , Inflamación/fisiopatología , Transducción de Señal/fisiología , Animales , Citocinas/fisiología , Homeostasis , Humanos , Quinasas Janus/fisiología , Factor de Transcripción STAT1/fisiología
6.
J Neurol Neurosurg Psychiatry ; 77(11): 1279-81, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17043295

RESUMEN

BACKGROUND AND PURPOSE: The pathophysiology of stroke-associated infection (SAI) is uncertain. The cytokine profile and peripheral white cell response were assessed in patients with or without SAI. METHODS: The incidence of SAI was assessed in 110 patients with ischaemic stroke allocated antibiotic prophylaxis or placebo within 24 h of clinical onset. Peripheral white cell counts, interleukin (IL)6, tumour necrosis factor (TNF)alpha and IL10 were measured in plasma. RESULTS: 17 (15%) patients developed infection and showed time-dependent increases of total white cell count, neutrophils, monocytes, lymphocytes, IL6 and IL10, whereas TNFalpha and the TNFalpha/IL10 ratio decreased. In logistic regression, IL10 (odds ratio (OR) 1.08, 95% confidence interval (CI) 1.01 to 1.16), monocyte count (OR 1.42, 95% CI 1.08 to 1.87) and National Institute for Health Stroke Survey score on admission (OR 1.17, 95% CI 1.05 to 1.31) were independent predictors of systemic infection. CONCLUSIONS: SAI is associated with stroke severity, excessive IL10-mediated response and an increased number of circulating monocytes. These results support the finding that acute ischaemic brain injury triggers a blood-borne anti-inflammatory response that decreases the antimicrobial drive of the immune system.


Asunto(s)
Isquemia Encefálica/complicaciones , Isquemia Encefálica/microbiología , Infecciones/etiología , Interleucina-10/sangre , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/microbiología , Anciano , Anciano de 80 o más Años , Profilaxis Antibiótica , Femenino , Humanos , Incidencia , Infecciones/epidemiología , Infecciones/inmunología , Masculino , Persona de Mediana Edad , Monocitos , Oportunidad Relativa , Factores de Riesgo
7.
J Neural Transm (Vienna) ; 113(12): 1837-45, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16752047

RESUMEN

Carboxyl-terminal fragments (CTs) of the amyloid precursor protein have been shown to be highly neurotoxic and are though to contribute to the neuropathology of Alzheimer's disease. We compared the effects of expressing CT99 in the human neuroblastoma MC65 with the effects of hydrogen peroxide on the parental SK-N-MC cells. CT99 and hydrogen peroxide generated a different pattern of free radicals and their toxic effects were differentially protected by a battery of antioxidants. Hydrogen peroxide caused a cell cycle arrest at phase S and apoptosis mediated through caspase-3 activation in a pattern similar to that described for amyloid-beta neurotoxicity. However, CT99 apoptosis appeared to be mediated through an unidentified mitochondrial pathway. Both oxidative injury types induced heme oxygenase-1 expression as a neuroprotective response. Overall we found a coincidence in the nonespecific stress oxidative effects of CT99 and hydrogen peroxide, but clear differences on their respective potencies and pathways of neurotoxicity.


Asunto(s)
Precursor de Proteína beta-Amiloide/toxicidad , Peróxido de Hidrógeno/toxicidad , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/fisiología , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Hemo-Oxigenasa 1/biosíntesis , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Fragmentos de Péptidos/toxicidad , Espectrometría de Fluorescencia , Superóxidos/metabolismo
8.
Stroke ; 36(7): 1495-500, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15961713

RESUMEN

BACKGROUND AND PURPOSE: Early infection after stroke is frequent but the clinical value of antibiotic prophylaxis in acute stroke has never been explored. OBJECTIVE AND METHODS: The Early Systemic Prophylaxis of Infection After Stroke (ESPIAS) is a randomized, double-blind, placebo-controlled study of antibiotic prophylaxis in patients older than 18 years with nonseptic ischemic or hemorrhagic stroke enrolled within 24 hours from clinical onset. Interventions included intravenous levofloxacin (500 mg/100 mL/d, for 3 days) or placebo (0.9% physiological serum) in addition to optimal care. A sample size of 240 patients was calculated to identify a 15% absolute risk reduction of the primary outcome measure, which was the incidence of infection at day 7 after stroke. Secondary outcome measures were neurological outcome and mortality at day 90. RESULTS: Based on a preplanned futility analysis, the study was interrupted prematurely when 136 patients had been included. Levofloxacin and placebo patients had a cumulative rate of infection of 6% and 6% (P=0.96) at day 1; 10% and 12% (P=0.83) at day 2; 12% and 15% (P=0.66) at day 3; 16% and 19% (P=0.82) at day 7; and 30% and 33% (P=0.70), at day 90. Using logistic regression, favorable outcome at day 90 was inversely associated with baseline National Institutes of Health Stroke Scale (OR, 0.72; 95% CI, 0.59 to 0.89; P=0.002) and allocation to levofloxacin (OR, 0.19; 95% CI, 0.04 to 0.87; P=0.03). CONCLUSIONS: Prophylactic administration of levofloxacin (500 mg/100 mL/day for 3 days) is not better than optimal care for the prevention of infections in patients with acute stroke.


Asunto(s)
Antibacterianos/uso terapéutico , Antiinfecciosos/uso terapéutico , Infecciones/patología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/microbiología , Accidente Cerebrovascular/terapia , Anciano , Temperatura Corporal , Encéfalo/patología , Isquemia Encefálica/terapia , Proteína C-Reactiva/metabolismo , Método Doble Ciego , Femenino , Humanos , Isquemia , Leucocitos/citología , Levofloxacino , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Ofloxacino/uso terapéutico , Placebos , Riesgo , Factores de Tiempo , Resultado del Tratamiento
9.
Cerebrovasc Dis ; 19(2): 91-5, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15608432

RESUMEN

BACKGROUND AND PURPOSE: The CC genotype of the -174 G/C interleukin (IL)-6 polymorphism has been associated with lacunar stroke. However, it remains unsettled whether this polymorphism is also associated with other ischemic stroke phenotypes. METHODS: The -174 G/C IL-6 polymorphism was genotyped in patients with lacunar stroke (n = 89), stroke due to large vessel disease (n = 82), cardioembolism (n = 53), stroke of undetermined cause (n = 49) and in white controls without any history of stroke (n = 105) by PCR and restriction enzyme analysis. Independent predictors of the -174 G/C IL-6 genotypes were assessed using multivariate logistic regression models adjusted for demographics, risk factors and disease state. RESULTS: The prevalence of the CC genotype was 8.5% in large vessel disease, 7.5% in embolism, 19.1% in lacunar stroke, 14.3% in stroke of undetermined cause and 8.6% in controls. The CC genotype was independently associated with lacunar stroke only (adjusted OR 3.22, 95% CI 9.09-1.12). Contrarily, there were no significant differences in genotype and allele distribution in the remainder of ischemic stroke phenotypes. Pooling of patients with nonlacunar stroke did not show any independent association with the CC genotype as compared with controls (OR 1.01, 95% CI 2.77-0.36). CONCLUSIONS: The unique association between the CC genotype of the -174 G/C IL-6 polymorphism and lacunar stroke suggests a particular susceptibility of small deep penetrators of cerebral arteries to IL-6-mediated inflammatory damage.


Asunto(s)
Isquemia Encefálica/genética , Interleucina-6/genética , Polimorfismo Genético/genética , Accidente Cerebrovascular/genética , Anciano , Estudios de Casos y Controles , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Regiones Promotoras Genéticas/genética
11.
Neuropathol Appl Neurobiol ; 29(5): 472-81, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14507339

RESUMEN

Transient focal ischaemia by middle cerebral artery occlusion (MCAO) may produce cell death, but the mechanisms leading to cell death differ in the infarct core and in the penumbra, the immediate zone surrounding the infarct core. In the present study, transient focal ischaemia to adult rats was produced by intraluminal occlusion of the middle cerebral artery for 1 h followed by 0 h (n=6), 1 h (n=10), 4 h (n=8), 6 h (n=2) and 12 h (n=3) of reperfusion. The present model of ischaemia causes a large cortico-striatal infarct extending through the mediolateral cortex and dorsolateral striatum at 12 h. The expression and subcellular distribution of several proteins involved in apoptosis have been examined in the penumbra and in the infarct core by using combined methods of immunohistochemistry, cell subfractionation and Western blotting. Transient focal ischaemia by MCAO results in activation of complex signal pathways for cell death in the penumbra. Increased expression of Bcl-2 and Bax, but not of Bcl-x, occurs in the penumbra at the time when Bax translocates from the cytosol to the mitochondria, cytochrome c is released to the cytoplasm and active caspase-3 is expressed. Bax translocation, cytochrome c release and active caspase-3 are observed at 4 h, but not at 1 h, following reperfusion, and together indicate activation of the caspase-dependent pathway of apoptosis in the penumbra. In contrast, reduced Bax expression but not Bax translocation and cytochrome c release occurs in the infarct core, thus suggesting apoptosis signals restricted to the penumbra. In addition, increased expression of an apoptosis-inducing factor in the cytoplasm and nuclei of selected cells shows, for the first time, activation of the caspase-independent mitochondrial pathway in the penumbra following transient focal ischaemia and reperfusion.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Caspasas/metabolismo , Neuronas/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Apoptosis , Factor Inductor de la Apoptosis , Western Blotting , Isquemia Encefálica/etiología , Citocromos c/metabolismo , Citosol/metabolismo , Activación Enzimática/fisiología , Flavoproteínas/biosíntesis , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Proteínas de la Membrana/biosíntesis , Mitocondrias/metabolismo , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ratas , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Factores de Tiempo , Proteína X Asociada a bcl-2 , Proteína bcl-X
12.
Acta Neuropathol ; 105(5): 425-37, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12677442

RESUMEN

Focal ischemia induced by middle cerebral artery occlusion (MCAO) to adult rats results in necrosis at the infarct core and activation of complex signal pathways for cell death and cell survival in the penumbra. Upstream from the cell death promoters and executioners are several kinases that, once activated by phosphorylation, may activate several transcription factor substrates involved in cell death and cell survival. In the present study we examined, by immunohistochemistry, the expression of phosphorylated (active) mitogen-activated protein kinase, extracellular signal-regulated kinase (MAPK/ERK), stress-activated protein kinase (SAPK), c-Jun N-terminal kinase (JNK) and p-38 kinase at early stages (1-4 h) following 1 h of MCAO in the rat. The expression of phosphorylation-dependent, active transcription substrates of these kinases, including cyclic AMP-responsive element-binding protein (CREB) Alk-1, ATF-2, c-Myc and c-Jun was examined at early stages following reperfusion. Increased nuclear phosphorylated SAPK/JNK (SAPK/JNK-P) and c-Jun-PSer63, and reduced CREB-P, occurred in the infarct core at 1 h following reperfusion, suggesting increased phosphorylated SAPK/JNK and c-JunSer63, together with decreased phospho-CREB associated with cell death in the infarct core. However, increased cytoplasmic expression of MAPK/ERK-P, SAPK/JNK-P, p38-P, CREB-P, Elk-1-P, c-Myc-P, ATF-2-P and c-Jun-P occurred in the region bordering the infarct core (penumbra) at 4 h following reperfusion. This indicates that different signals converge in the cytoplasm of neurons located at the borders of the infarct at 4 h following reperfusion, revealing the struggle of death promoters and life facilitators at the penumbra. Whether phosphorylated kinases and specific substrates participate in promoting cell death or survival in the penumbra probably depends on additional factors and on the interaction with other proteins.


Asunto(s)
Isquemia Encefálica/enzimología , Proteínas Portadoras/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factores de Transcripción Activadores , Animales , Proteínas Sanguíneas/metabolismo , Isquemia Encefálica/metabolismo , Cuerpo Estriado/citología , Cuerpo Estriado/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/metabolismo , MAP Quinasa Quinasa 4 , Masculino , Fosforilación , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Factores de Transcripción/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
13.
Neuroreport ; 12(15): 3381-4, 2001 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-11711890

RESUMEN

We studied whether pro-survival Akt was activated after transient focal cerebral ischemia and whether it inhibited pro-apoptotic Bad. Phosphorylation of Akt (serine-473) was enhanced in cortex after 1-hour ischemia, and also after 1h and 6 h of reperfusion, but it returned back to that in controls by 24 h. After this first wave of Akt activation, a second increase was observed between 4 and 7 days. In striatum, only the late Akt activation was seen. In contrast to Akt, no Bad phosphorylation (serine-136) was detected after ischemia. Therefore, injury spontaneously activated Akt, but this did not suppress Bad signalling. It is proposed that further pharmacological activation of Akt shortly after ischemia might promote cell survival, whereas Akt activation at longer time points is involved with glial reactivity.


Asunto(s)
Apoptosis/fisiología , Isquemia Encefálica/enzimología , Proteínas Portadoras/metabolismo , Supervivencia Celular/fisiología , Corteza Cerebral/enzimología , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Daño por Reperfusión/enzimología , Secuencia de Aminoácidos/fisiología , Animales , Isquemia Encefálica/fisiopatología , Corteza Cerebral/fisiopatología , Proteínas del Choque Térmico HSP72 , Proteínas de Choque Térmico/metabolismo , Masculino , Neostriado/enzimología , Neostriado/fisiopatología , Fosforilación , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/fisiopatología , Serina/metabolismo , Transducción de Señal/fisiología , Factores de Tiempo , Proteína Letal Asociada a bcl
14.
Neurobiol Dis ; 8(5): 834-46, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11592852

RESUMEN

Matrix metalloproteinases (MMPs) degrade the extracellular matrix and carry out key functions during development and after injury. By means of zymography, Western blot and immunohistochemistry, we studied MMP-2 (gelatinase A) and MMP-9 (gelatinase B) in rat brain after focal cerebral ischemia. The control rat brain showed constitutive MMP-2 and, to a lesser extent, MMP-9, which were mainly present as prozymogens. MMP-2 protein was located in the cell body of neurons, glia, and endothelium, whereas MMP-9 was associated to neurons and myelinated fibre tracts. Ischemia greatly increased MMP activation in two temporal waves, in the first one, MMP-9 protein was induced from 4 h to 4 days, and also a small and short-lasting increase in MMP-2 was detected at 4 h. The second wave showed a massive increase in MMP-2 protein expression and activation by day 4, which was compatible with abundant MMP-2 in reactive microglia/macrophages. Our results are compatible with progressive induction of MMP-9 proform, likely in neurons, shortly after ischemia. For MMP-2, the results suggest a discrete production immediately after reperfusion, while a very enhanced expression and activation of MMP-2 attributable to microglia/macrophages occurs on day 4, and it might contribute to the phagocytic action of these reactive cells.


Asunto(s)
Encéfalo/enzimología , Ataque Isquémico Transitorio/enzimología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Barrera Hematoencefálica , Western Blotting , Endotelio Vascular/enzimología , Activación Enzimática , Inducción Enzimática , Precursores Enzimáticos/metabolismo , Técnicas para Inmunoenzimas , Macrófagos/enzimología , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Fibras Nerviosas Mielínicas/enzimología , Proteínas del Tejido Nervioso/genética , Neuroglía/enzimología , Neuronas/enzimología , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/enzimología , Factores de Tiempo
15.
J Cereb Blood Flow Metab ; 21(9): 1097-104, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11524614

RESUMEN

Growth factors promote cell growth and survival and protect the brain from developing injury after ischemia. In this article, the authors examined whether transforming growth factor-alpha (TGF-alpha) was protective in transient focal ischemia and whether alteration of cerebral circulation was involved. Rats received intraventricular TGF-alpha (50 ng, either split into 2 doses given 30 minutes before and 30 minutes after middle cerebral artery occlusion (MCAO), or 1 dose given 30 minutes after MCAO) or vehicle. Rats were subjected to 1-hour intraluminal MCAO and cerebral blood flow was recorded continuously by laser-Doppler flowmetry. Infarct volume was measured 1 and 4 days later. The effects of TGF-alpha on arterial tone were assessed in isolated rabbit basilar and common carotid arteries. Transforming growth factor-alpha before and after ischemia reduced infarct volume by 70% at 1 day and 50% at 4 days. Transforming growth factor-alpha given only after ischemia also did reduce infarct volume by 70% at 1 day and 80% at 4 days. The protective effect was more marked in cortex than in striatum. Transforming growth factor-alpha did not change cortical microvascular perfusion and did not modify arterial passive tone nor agonist-induced active tone. It can be concluded that TGF-alpha reduces infarct volume, even when the factor is exclusively administered at reperfusion, and that this effect is not mediated by changes in microvascular perfusion or cerebral arteries. It is therefore suggested that TGF-alpha has a protective effect against neuronal cell death after transient focal ischemia.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Circulación Cerebrovascular/efectos de los fármacos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Factor de Crecimiento Transformador alfa/farmacología , Animales , Arteria Basilar/efectos de los fármacos , Arterias Carótidas/efectos de los fármacos , Técnicas In Vitro , Masculino , Microcirculación/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Conejos , Ratas , Ratas Sprague-Dawley , Vasoconstricción/efectos de los fármacos
16.
Hippocampus ; 11(2): 146-56, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11345121

RESUMEN

The role of gene induction (expression of HSP72 and c-JUN proteins) and delayed ischemic cell death (in situ labeling of DNA fragmentation) have been investigated in the goat hippocampus after transient global cerebral ischemia. The animals were subjected to 20-min ischemia (bilateral occlusion of the external carotid arteries plus bilateral jugular vein compression) and allowed to reperfuse for 2 h, and then 1, 3, and 7 days. Histological signs of cell loss were not found in the hippocampus at 2 h, 1 day, or 3 days of reperfusion. However, such an ischemic insult produced extensive, selective, and delayed degeneration in the hippocampus, as 68% of the neurons in CA1 had died at 7 days, but cell loss was not detected in CA3 and dentate gyrus fields. Concomitantly, a high percentage of TUNEL-positive CA1 neurons (60+/-9%, mean +/- SEM) was seen at 7 days, but not at the earlier time points. Mild induction of HSP72 was detected in the goat hippocampus after ischemia. The maximum percentage of HSP72-positive neurons (10-15%) was shown at 3 days of reperfusion and was concentrated mainly in the CA3 field, subiculum, and hilus, rather than in the CA1 field, whereas HSP72 expression was hardly detected at 7 days. At this later time point, scattered induction of nuclear c-JUN was found in a few neurons. The results show that: 1) postischemic delayed neuronal death selectively affects the CA1 field in the goat hippocampus, a phenomenon which seems to take longer to develop than in previously reported rodent models; and 2) postischemic expression of c-JUN does not appear to be related to cell death or survival, while the inability of most CA1 neurons to express HSP72 could contribute to neuronal death.


Asunto(s)
Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Fragmentación del ADN , Cabras/fisiología , Proteínas de Choque Térmico/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Animales , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Muerte Celular/fisiología , Femenino , Proteínas del Choque Térmico HSP72 , Neuronas/fisiología , Factores de Tiempo , Distribución Tisular
17.
Biochem Biophys Res Commun ; 278(3): 803-7, 2000 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-11095988

RESUMEN

Matrix metalloproteinases degrade the extracellular matrix and are involved in a variety of diseases, including inflammatory diseases of the central nervous system. Here we estimated the content of gelatinase in rat brain under control conditions and 4 h after transient focal ischemia using gelatinolytic extraction and zymographic analysis. We also examined the expression of the MMP-9 and MMP-2 proteins by Western blot. Using the zymographic apparent gelatinase activity we estimated that brain gelatinase content was 0.44 ng/mg of protein. Ischemia induced a 1.7-fold increase at 4 h, thus showing an early MMP response to the ischemic injury. The main increase was seen for the MMP-9 proform, which was accompanied by enhanced MMP-9 protein expression. We suggest that basal cerebral MMP-9 and MMP-2 activities are involved in the maintenance of the extracellular matrix and prevent substrate accumulation, while enhanced postischemic MMP activity before cell death may contribute to edema formation and blood-brain barrier breakdown.


Asunto(s)
Encéfalo/enzimología , Gelatinasas/metabolismo , Ataque Isquémico Transitorio/enzimología , Animales , Colagenasas/metabolismo , Activación Enzimática , Precursores Enzimáticos/metabolismo , Gelatinasas/análisis , Cinética , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratas , Ratas Sprague-Dawley
18.
Neurobiol Dis ; 7(4): 343-61, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10964606

RESUMEN

Injection of MPP(+) into the substantia nigra causes extensive necrosis and anterograde degeneration of pars compacta dopaminergic neurons. We studied secondary effects in the ipsilateral striatum by examining dopaminergic terminals, signs of neuronal damage, and glial reactivity at 1, 2, 3, and 7 days after injection of MPP(+) into the substantia nigra. Dopaminergic terminals and uptake sites were evaluated with [(3)H]GBR-12935 binding and tyrosine hydroxylase immunoreactivity. Glial reaction was examined with markers of astrocytes and microglia. Stereology was used to evaluate any changes in neuronal density. Tyrosine hydroxylase immunoreactivity and [(3)H]GBR-12935 binding markedly decreased (74%) from days 2 to 7. Loss of dopaminergic terminals in the ipsilateral striatum was accompanied by an intense astroglial and, to a lesser extent, microglial reaction. However, no signs of cell damage, neuronal loss, or disruption of the blood-brain barrier were found in the striatum. Resident astroglial and microglial cells showed a morphological shift and notable changes in protein expression typical of glial reactivity, yet the presence of macrophage-like cells was not detected. This study shows that injection of MPP(+) in the substantia nigra causes a secondary reaction within the ipsilateral striatum involving the transformation of quiescent glia to reactive glia. It is suggested that stimuli derived from damaged dopaminergic terminals within the striatum are able to activate resident glia and that this glial transformation may promote repair and regeneration.


Asunto(s)
1-Metil-4-fenilpiridinio/farmacología , Cuerpo Estriado/efectos de los fármacos , Herbicidas/farmacología , Neuroglía/efectos de los fármacos , Sustancia Negra/efectos de los fármacos , Animales , Antineoplásicos/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Cuerpo Estriado/metabolismo , Proteína Ácida Fibrilar de la Glía/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Isoquinolinas/metabolismo , Masculino , Neuroglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Sustancia Negra/metabolismo
19.
Glia ; 30(3): 253-70, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10756075

RESUMEN

JAK/STAT is one of the pathways bearing signals from the cell membrane to the nucleus in response to extracellular growth factors and cytokines. In the present study, we examined the cellular distribution of Jak1 and Stat3, and activation of the JAK/STAT pathway following transient focal cerebral ischemia in the rat. Jak1 was mainly seen in white matter astrocytes and in certain neurons. Notably, large pyramidal neurons of cortical layer V showed the highest neuronal Jak1 expression within cerebral cortex and, in addition, expressed Stat3 indicating that the JAK/STAT pathway is involved in signaling in the corticofugal projection system. Shortly following ischemia, Jak1 immunoreactive astrocytes located in the ipsilateral neighbouring white matter and ischemic cortex and striatum showed nuclear translocation of Stat3. These features were maintained in large reactive astrocytes that surrounded the infarct from 3 to 7 days. At these later times, the abundant reactive microglia/macrophages were strongly immunoreactive to Stat3 and, to a lesser extent, Jak1. Two main protein complexes showing DNA binding activity at the sis-inducible element site were found under basal conditions, followed by changes in this pattern following ischemia concomitant with neuronal cell loss and activation of glia. This study showed basal cerebral activity of JAK/STAT signaling pathway, involving Jak1 and Stat3 proteins, and selective activation following ischemia. It is suggested that the kinase activity of Jak1 mediates nuclear translocation of Stat3 in astrocytes, and that this signaling pathway is involved in the astroglial response to focal cerebral ischemia.


Asunto(s)
Astrocitos/metabolismo , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transactivadores/metabolismo , Animales , Astrocitos/patología , Encéfalo/patología , Isquemia Encefálica/patología , Inmunohistoquímica , Janus Quinasa 1 , Masculino , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3
20.
J Neurosci Res ; 59(6): 797-805, 2000 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-10700017

RESUMEN

The effects of nitric oxide synthase (NOS) inhibitors, N(omega)-nitro-L-arginine and 7-nitroindazole, and the NOS substrate L-arginine on kainic acid (KA)-induced microglial reactivity and stress response were studied in the hippocampus 7 and 1 days after KA, respectively. Density of peripheral-type benzodiazepine receptors was measured as an index of microglial reactivity. Histological damage in hippocampus was evaluated at 7 days by neuronal counting. KA increased the maximal number of binding sites (B(max)) versus controls. Administration of either 7-nitroindazole (25 mg/kg) or N(omega)-nitro-L-arginine (20 and 50 mg/kg) 24 hr before KA, further increased B(max). This later effect was abolished by L-arginine (1 g/kg), which given 24 hr before KA decreased B(max) to control values. Also, KA-induced HSP72 stress response was attenuated by pre-treatment with L-arginine. Histological evaluation showed reduced cell numbers in the pyramidal cell layer of the hippocampus in groups receiving KA, either alone or in combination with 7-nitroindazole. Administration of L-arginine before KA attenuated neuronal loss in CA3 but not CA1. A clear protective effect was observed, however, in CA1 and CA3, in rats receiving both L-arginine plus 7-nitroindazole before KA. The results show that the combination of a NO substrate with a NOS inhibitor reduces the neurotoxic effects of KA in the rat hippocampus. This study suggests that extremely fine regulation of NO levels in the different neural cell types can modulate excitotoxicity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/toxicidad , Hipocampo/efectos de los fármacos , Ácido Kaínico/toxicidad , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Animales , Antineoplásicos/farmacología , Western Blotting , Proteínas del Choque Térmico HSP72 , Proteínas de Choque Térmico/análisis , Proteínas de Choque Térmico/efectos de los fármacos , Hipocampo/metabolismo , Indazoles/farmacología , Péptidos y Proteínas de Señalización Intracelular , Isoquinolinas/farmacología , Masculino , Óxido Nítrico/agonistas , Óxido Nítrico/antagonistas & inhibidores , Nitroarginina/farmacología , Proteínas Serina-Treonina Quinasas/análisis , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA