Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Stroke ; 47(1): 262-6, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26564103

RESUMEN

BACKGROUND AND PURPOSE: Preconditioning with poly-l-lysine and carboxymethylcellulose (ICLC) provides robust neuroprotection from cerebral ischemia in a mouse stroke model. However, the receptor that mediates neuroprotection is unknown. As a synthetic double-stranded RNA, poly-ICLC may bind endosomal Toll-like receptor 3 or one of the cytosolic retinoic acid-inducible gene-I-like receptor family members, retinoic acid-inducible gene-I, or melanoma differentiation-associated protein 5. Activation of these receptors culminates in type I interferons (IFN-α/ß) induction-a response required for poly-ICLC-induced neuroprotection. In this study, we investigate the receptor required for poly-ICLC-induced neuroprotection. METHODS: Toll-like receptor 3, melanoma differentiation-associated protein 5-, and IFN-promoter stimulator 1-deficient mice were treated with poly-ICLC 24 hours before middle cerebral artery occlusion. Infarct volume was measured 24 hours after stroke to identify the receptor signaling pathways involved in protection. IFN-α/ß induction was measured in plasma samples collected 6 hours after poly-ICLC treatment. IFN-ß-deficient mice were used to test the requirement of IFN-ß for poly-ICLC-induced neuroprotection. Mice were treated with recombinant IFN-α-A to test the role of IFN-α as a potential mediator of neuroprotection. RESULTS: Poly-ICLC induction of both neuroprotection and systemic IFN-α/ß requires the cytosolic receptor melanoma differentiation-associated protein 5 and the adapter molecule IFN-promoter stimulator 1, whereas it is independent of Toll-like receptor 3. IFN-ß is not required for poly-ICLC-induced neuroprotection. IFN-α treatment protects against stroke. CONCLUSIONS: Poly-ICLC preconditioning is mediated by melanoma differentiation-associated protein 5 and its adaptor molecule IFN-promoter stimulator 1. This is the first evidence that a cytosolic receptor can mediate neuroprotection, providing a new target for the development of therapeutic agents to protect the brain from ischemic injury.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevención & control , ARN Helicasas DEAD-box/metabolismo , Precondicionamiento Isquémico/métodos , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/prevención & control , Animales , Carboximetilcelulosa de Sodio/análogos & derivados , Carboximetilcelulosa de Sodio/metabolismo , Carboximetilcelulosa de Sodio/uso terapéutico , Helicasa Inducida por Interferón IFIH1 , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Poli I-C/metabolismo , Poli I-C/uso terapéutico , Polilisina/análogos & derivados , Polilisina/metabolismo , Polilisina/uso terapéutico
2.
Acta Neurochir Suppl ; 121: 39-44, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26463920

RESUMEN

Stroke activates an inflammatory response that results in the infiltration of peripheral immune cells into the ischemic area, contributing to exacerbation of tissue damage. However, evidence indicates that inflammatory cell infiltration can also promote neuroprotection through regulatory immune cells that mitigate injury. These immune regulatory cells may also be important mediators of neuroprotection associated with preconditioning, a phenomenon whereby small exposure to a potential harmful stimulus is able to induce protection against a subsequent ischemic event. The elucidation of mechanisms that allow these immune cells to confer neuroprotection is critical to developing new therapeutic strategies against acute stroke. In the present review, we discuss the dual role of peripheral immune cells in stroke-related brain injury and neuroprotection. Furthermore, we report new data from our laboratory that supports the important role of peripheral cells and their interaction with the brain endothelium for the establishment of the protective phenotype in preconditioning.


Asunto(s)
Precondicionamiento Isquémico , Linfocitos/inmunología , Macrófagos/inmunología , Neuroprotección/inmunología , Neutrófilos/inmunología , Accidente Cerebrovascular/inmunología , Animales , Humanos
3.
Circulation ; 126(12): 1484-94, 2012 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-22879370

RESUMEN

BACKGROUND: The involvement of the complement system in brain injury has been scarcely investigated. Here, we document the pivotal role of mannose-binding lectin (MBL), one of the recognition molecules of the lectin complement pathway, in brain ischemic injury. METHODS AND RESULTS: Focal cerebral ischemia was induced in mice (by permanent or transient middle cerebral artery occlusion) and rats (by 3-vessel occlusion). We first observed that MBL is deposited on ischemic vessels up to 48 hours after injury and that functional MBL/MBL-associated serine protease 2 complexes are increased. Next, we demonstrated that (1) MBL(-/-) mice are protected from both transient and permanent ischemic injury; (2) Polyman2, the newly synthesized mannosylated molecule selected for its binding to MBL, improves neurological deficits and infarct volume when given up to 24 hours after ischemia in mice; (3) anti-MBL-A antibody improves neurological deficits and infarct volume when given up to 18 hours after ischemia, as assessed after 28 days in rats. CONCLUSIONS: Our data show an important role for MBL in the pathogenesis of brain ischemic injury and provide a strong support to the concept that MBL inhibition may be a relevant therapeutic target in humans, one with a wide therapeutic window of application.


Asunto(s)
Isquemia Encefálica/fisiopatología , Infarto de la Arteria Cerebral Media/fisiopatología , Lectina de Unión a Manosa/genética , Lectina de Unión a Manosa/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/genética , Edema Encefálico/fisiopatología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/genética , Modelos Animales de Enfermedad , Humanos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/genética , Masculino , Mananos/metabolismo , Mananos/farmacología , Lectina de Unión a Manosa/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratas , Ratas Endogámicas
4.
J Neurochem ; 122(6): 1219-29, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22804628

RESUMEN

The molecular mechanisms that lead to ischemic pre-conditioning are not completely understood, and proteins are important players. We compared the mouse brain cortex proteome from different ischemia sets: transient (7 min) middle cerebral artery occlusion (7'MCAo, pre-conditioning stimulus), permanent MCAo (pMCAo, severe ischemia), and pMCAo 4 days after 7'MCAo (7'MCAo/pMCAo, pre-conditioned model). Proteins were analyzed by two-dimensional electrophoresis coupled to liquid chromatography-tandem mass spectrometry. Overall, 28 proteins were expressed differentially from sham controls, and identified. The ischemic pre-conditioning stimulus alone up-regulated the stress protein heat-shock protein 70 (HSP70), possibly activated by the androgen receptor. Western blotting confirmed the increased expression of HSP70 and showed that androgen receptor expression paralleled that of HSP70. In the ischemic-tolerant group (7'MCAo/pMCAo), a number of proteins over-expressed after pMCAo returned to sham levels, seven proteins remained up-regulated as in pMCAo, and five proteins mainly involved in energy metabolism and mitochondrial electron transport and unchanged in pMCAo were down-regulated only in ischemic tolerance, suggesting a role in brain pre-conditioning. Astrocytes participated in ischemic-tolerance induction, as shown by the down-regulation of glutamine synthetase in the 7'MCAo/pMCAo group. The results suggest that metabolic down-regulation was a general feature of ischemic pre-conditioning, playing a pivotal role in neuroprotection.


Asunto(s)
Isquemia Encefálica/metabolismo , Corteza Cerebral/metabolismo , Regulación hacia Abajo/fisiología , Metabolismo Energético/fisiología , Precondicionamiento Isquémico/métodos , Proteómica/métodos , Animales , Isquemia Encefálica/terapia , Masculino , Ratones , Ratones Endogámicos C57BL
5.
J Neurochem ; 123 Suppl 2: 75-85, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23050645

RESUMEN

Preconditioning with a low dose of harmful stimulus prior to injury induces tolerance to a subsequent ischemic challenge resulting in neuroprotection against stroke. Experimental models of preconditioning primarily focus on neurons as the cellular target of cerebral protection, while less attention has been paid to the cerebrovascular compartment, whose role in the pathogenesis of ischemic brain injury is crucial. We have shown that preconditioning with polyinosinic polycytidylic acid (poly-ICLC) protects against cerebral ischemic damage. To delineate the mechanism of poly-ICLC protection, we investigated whether poly-ICLC preconditioning preserves the function of the blood-brain barrier (BBB) in response to ischemic injury. Using an in vitro BBB model, we found that poly-ICLC treatment prior to exposure to oxygen-glucose deprivation maintained the paracellular and transcellular transport across the endothelium and attenuated the drop in transendothelial electric resistance. We found that poly-ICLC treatment induced interferon (IFN) ß mRNA expression in astrocytes and microglia and that type I IFN signaling in brain microvascular endothelial cells was required for protection. Importantly, this implicates a potential mechanism underlying neuroprotection in our in vivo experimental stroke model, where type I IFN signaling is required for poly-ICLC-induced neuroprotection against ischemic injury. In conclusion, we are the first to show that preconditioning with poly-ICLC attenuates ischemia-induced BBB dysfunction. This mechanism is likely an important feature of poly-ICLC-mediated neuroprotection and highlights the therapeutic potential of targeting BBB signaling pathways to protect the brain against stroke.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Carboximetilcelulosa de Sodio/análogos & derivados , Infarto de la Arteria Cerebral Media/prevención & control , Factor 1 Regulador del Interferón/metabolismo , Precondicionamiento Isquémico/métodos , Fármacos Neuroprotectores/administración & dosificación , Poli I-C/administración & dosificación , Polilisina/análogos & derivados , Transducción de Señal/efectos de los fármacos , Análisis de Varianza , Animales , Animales Recién Nacidos , Barrera Hematoencefálica/metabolismo , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/etiología , Carboximetilcelulosa de Sodio/administración & dosificación , Carboximetilcelulosa de Sodio/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Glucosa/deficiencia , Hipoxia/tratamiento farmacológico , Hipoxia/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/genética , Factor 1 Regulador del Interferón/deficiencia , Interferón beta/genética , Interferón beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroglía/efectos de los fármacos , Poli I-C/farmacología , Polilisina/administración & dosificación , Polilisina/farmacología , ARN Mensajero/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/patología , Factores de Tiempo
6.
Stroke ; 42(5): 1445-53, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21474800

RESUMEN

BACKGROUND AND PURPOSE: The cerebrovascular contribution to ischemic preconditioning (IPC) has been scarcely explored. Using in vivo and in vitro approaches, we investigated the involvement of the blood-brain barrier and the role of its cellular components. METHODS: Seven-minute occlusion of the right middle cerebral artery, used as in vivo IPC stimulus 4 days before permanent occlusion of the right middle cerebral artery, significantly reduced brain infarct size (8.45±0.7 versus 13.61±0.08 mm3 measured 7 days after injury) and preserved blood-brain barrier function (Evans blue leakage, 0.54±0.1 versus 0.89±0.1 ng/mg). Assessment of neuronal, endothelial, and glial gene expression revealed that IPC specifically increased glial fibrillary acidic protein mRNA, thus showing selective astrocyte activation in IPC-protected mice. RESULTS: The blood-brain barrier was modeled by coculturing murine primary brain microvessel endothelial and astroglial cells. One-hour oxygen-glucose deprivation (OGD), delivered 24 hours before a 5-hour OGD, acted as an IPC stimulus, significantly attenuating the reduction in transendothelial electric resistance (199.17±11.7 versus 97.72±3.4 Ωcm2) and the increase in permeability coefficients for sodium fluorescein (0.98±0.11×10(-3) versus 1.8±0.36×10(-3) cm/min) and albumin (0.12±0.01×10(-3) versus 0.29±0.07×10(-3) cm/min) induced by severe OGD. IPC also prevented the 5-hour OGD-induced disorganization of the tight junction proteins ZO-1 and claudin-5. IPC on glial (but not endothelial) cells alone preserved transendothelial electric resistance, permeability coefficients, and ZO-1 localization after 5 hours of OGD. Astrocyte metabolic inhibition by fluorocitrate abolished IPC protection, confirming the critical role of astrocytes. IPC significantly increased glial fibrillary acidic protein, interleukin-6, vascular endothelial growth factor-a, and ciliary neurotrophic factor gene expression after OGD in glial cells, indicating that multiple pathways mediate the glial contribution to IPC. CONCLUSIONS: Our data show that the blood-brain barrier can be directly preconditioned and that astrocytes are major mediators of IPC protection.


Asunto(s)
Barrera Hematoencefálica/fisiología , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Precondicionamiento Isquémico , Neuroglía/fisiología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Isquemia Encefálica/metabolismo , Claudina-5 , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Proteína Ácida Fibrilar de la Glía , Infarto de la Arteria Cerebral Media , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/patología , Fosfoproteínas/metabolismo , Proteína de la Zonula Occludens-1
7.
Ann Neurol ; 66(3): 332-42, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19798727

RESUMEN

OBJECTIVE: C1 inhibitor (C1-INH) is an endogenous inhibitor of complement and kinin systems. We have explored the efficacy and the therapeutic window of the recently available human recombinant (rh) C1-INH on ischemic brain injury and investigated its mechanism of action in comparison with that of plasma-derived (pd) C1-INH. METHODS: rhC1-INH was administered intravenously to C57Bl/6 mice undergoing transient or permanent ischemia, and its protective effects were evaluated by measuring infarct volume and neurodegeneration. The binding profiles of rhC1-INH and pdC1-INH were assessed in vitro using surface plasmon resonance. Their localization in the ischemic brain tissue was determined by immunohistochemistry and confocal analysis. The functional consequences of rhC1-INH and pdC1-INH administration on complement activation were analyzed by enzyme-linked immunosorbent assay on plasma samples. RESULTS: rhC1-INH markedly reduced cerebral damage when administered up to 18 hours after transient ischemia and up to 6 hours after permanent ischemia, thus showing a surprisingly wide therapeutic window. In vitro rhC1-INH bound mannose-binding lectin (MBL), a key protein in the lectin complement pathway, with high affinity, whereas pdC1-INH, which has a different glycosylation pattern, did not. In the ischemic brain, rhC1-INH was confined to cerebral vessels, where it colocalized with MBL, whereas pdC1-INH diffused into the brain parenchyma. In addition, rhC1-INH was more active than pdC1-INH in inhibiting MBL-induced complement activation. INTERPRETATION: rhC1-INH showed a surprisingly wider time window of efficacy compared with the corresponding plasmatic protein. We propose that the superiority of rhC1-INH is due to its selective binding to MBL, which emerged as a novel target for stroke treatment.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Proteína Inhibidora del Complemento C1/uso terapéutico , Ataque Isquémico Transitorio/tratamiento farmacológico , Animales , Isquemia Encefálica/metabolismo , Activación de Complemento , Proteína Inhibidora del Complemento C1/farmacología , Vía Alternativa del Complemento , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Masculino , Lectina de Unión a Manosa/farmacología , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes
8.
J Neuropathol Exp Neurol ; 73(5): 378-86, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24709682

RESUMEN

Toll-like receptors (TLRs) are master regulators of innate immunity and play an integral role in the activation of inflammatory response during infections. In addition, TLRs influence the body's response to numerous forms of injury. Recent data have shown that TLRs play a modulating role in ischemic brain damage after stroke. Interestingly, their stimulation before ischemia induces a tolerant state that is neuroprotective. This phenomenon, referred to as TLR preconditioning, is the result of the reprogramming of TLR response to ischemic injury. This review addresses the role of TLRs in brain ischemia and the activation of endogenous neuroprotective pathways in the setting of preconditioning. We highlight the protective role of interferon-related response and the potential site of action for TLR preconditioning involving the blood-brain barrier. Pharmacologic modulation of TLR activation to promote protection against stroke is a promising approach for the development of prophylactic and immediate therapies targeting ischemic brain injury.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevención & control , Transducción de Señal/fisiología , Receptores Toll-Like/metabolismo , Animales , Humanos , Fármacos Neuroprotectores/metabolismo
9.
Transl Stroke Res ; 4(1): 89-103, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23504609

RESUMEN

Efforts to treat cardiovascular and cerebrovascular diseases often focus on the mitigation of ischemia-reperfusion (I/R) injury. Many treatments or "preconditioners" are known to provide substantial protection against the I/R injury when administered prior to the event. Brief periods of ischemia itself have been validated as a means to achieve neuroprotection in many experimental disease settings, in multiple organ systems, and in multiple species suggesting a common pathway leading to tolerance. In addition, pharmacological agents that act as potent preconditioners have been described. Experimental induction of neuroprotection using these various preconditioning paradigms has provided a unique window into the brain's endogenous protective mechanisms. Moreover, preconditioning agents themselves hold significant promise as clinical-stage therapies for prevention of I/R injury. The aim of this article is to explore several key steps involved in the preclinical validation of preconditioning agents prior to the conduct of clinical studies in humans. Drug development is difficult, expensive and relies on multi-factorial analysis of data from diverse disciplines. Importantly, there is no single path for the preclinical development of a novel therapeutic and no proven strategy to ensure success in clinical translation. Rather, the conduct of a diverse array of robust preclinical studies reduces the risk of clinical failure by varying degrees depending upon the relevance of preclinical models and drug pharmacology to humans. A strong sense of urgency and high tolerance of failure are often required to achieve success in the development of novel treatment paradigms for complex human conditions.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Precondicionamiento Isquémico , Fármacos Neuroprotectores/uso terapéutico , Investigación Biomédica Traslacional , Animales , Barrera Hematoencefálica/fisiología , Isquemia Encefálica/terapia , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Ratones , Ratas
10.
J Cereb Blood Flow Metab ; 31(9): 1919-29, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21468087

RESUMEN

We investigated the occurrence of endotoxin (lipopolysaccharide, LPS) preconditioning in traumatic brain injury (TBI), evaluating the time window of LPS-induced protection, its persistence, and the associated molecular mechanisms. Mice received 0.1 mg/kg LPS or saline intraperitoneally and subsequently TBI (by controlled cortical impact brain injury) at various time intervals. Mice receiving LPS 3, 5, or 7 days before TBI showed attenuated motor deficits at 1 week after injury compared with mice receiving saline. Those receiving LPS 5 days before injury had also a reduced contusion volume (7.9±1.3 versus 12±2.3 mm(3)) and decreased cell death. One month after injury, the protective effect of LPS on contusion volume (14.5±1.2 versus 18.2±1.2 mm(3)) and neurologic function was still present. Traumatic brain injury increased glial fibrillary acidic protein, CD11b, CD68, tumor necrosis factor-α, interleukin (IL)-10, and IL-6 mRNA expression 24 hours after injury. Lipopolysaccharide administered 5 (but not 9) days before injury increased the expression of CD11b (233%) and of interferon ß (500%) in uninjured mice, while it reduced the expression of CD68 (by 46%) and increased that of IL-6 (by 52%) in injured mice. Lipopolysaccharide preconditioning conferred a long-lasting neuroprotection after TBI, which was associated with a modulation of microglia/macrophages activity and cytokine production.


Asunto(s)
Lesiones Encefálicas/prevención & control , Encéfalo/efectos de los fármacos , Lipopolisacáridos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Cognición/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos
11.
Neurobiol Aging ; 28(9): 1388-95, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16846668

RESUMEN

Recent findings suggested a role for soluble amyloid-beta (Abeta) peptides in Alzheimer's disease associated cognitive decline. We investigated the action of soluble, monomeric Abeta(1-40) on CaM kinase II, a kinase involved in neuroplasticity and cognition. We treated organotypic hippocampal cultures short-term (up to 4h) and long-term (5 days) with Abeta(1-40) (1nM-5microM). Abeta did not induce cell damage, apoptosis or synaptic loss. Short-term treatment down-regulated enzymatic activity of the kinase, by reducing its Thr(286) phosphorylation. In contrast, long-term treatment (1nM-microM) markedly and significantly up-regulated enzymatic activity, with peak stimulation at 10nM (three-fold). Up-regulation of activity was associated with increased expression of the alpha-isoform of CaM kinase II, increased phosphorylation at Thr(286) (activator residue) and decreased phosphorylation at Thr(305-306) (inhibitory residues). We investigated the effect of glutamate on CaM kinase II following exposure to 1 or 10nM Abeta(1-40). As previously reported, glutamate increased CaM kinase II activity. However, the glutamate effect was not altered by pretreatment of slices with Abeta. Short- and long-term Abeta treatment showed opposite effects on CaM kinase II, suggesting that long-term changes are an adaptation to the kinase early down-regulation. The marked effect of Abeta(1-40) on the kinase suggests that semi-physiological and slowly raising peptide concentrations may have a significant impact on synaptic plasticity in the absence of synaptic loss or neuronal cell death.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Hipocampo/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Animales , Animales Recién Nacidos , Western Blotting , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Activación Enzimática/efectos de los fármacos , Técnicas de Cultivo de Órganos , Fosforilación/efectos de los fármacos , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Treonina/metabolismo , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
12.
J Pharmacol Exp Ther ; 318(2): 849-54, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16705080

RESUMEN

We have studied the effect of DX-88, a selective recombinant inhibitor of human plasma kallikrein, in transient or permanent focal brain ischemia (with or without reperfusion, respectively) induced in C57BL/6 mice. Twenty-four hours after transient ischemia, DX-88 administered at the beginning of ischemia (pre) induced a dose-dependent reduction of ischemic volume that, at the dose of 30 microg/mouse, reached 49% of the volume of saline-treated mice. At the same dose, DX-88 was also able to reduce brain swelling to 32%. Mice treated with DX-88 pre had significantly lower general and focal deficit score. Fluoro-Jade staining, a marker for neuronal degeneration, showed that DX-88-treated mice had a reduction in the number of degenerating cells, compared with saline-treated mice. Seven days after transient ischemia, the DX-88 protective effect was still present. When the inhibitor was injected at the end of ischemia (post), it was still able to reduce ischemic volume, brain swelling, and neurological deficits. DX-88 efficacy was lost when the inhibitor was given 30 min after the beginning of reperfusion (1 h post) or when reperfusion was not present (permanent occlusion model). This study shows that DX-88 has a strong neuroprotective effect in the early phases of brain ischemia preventing reperfusion injury and indicates that inhibition of plasma kallikrein may be a useful tool in the strategy aimed at reducing the detrimental effects linked to reperfusion.


Asunto(s)
Trastornos Cerebrovasculares/prevención & control , Calicreína Plasmática/antagonistas & inhibidores , Daño por Reperfusión/prevención & control , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Encéfalo/patología , Edema Encefálico/patología , Edema Encefálico/prevención & control , Recuento de Células , Fluoresceínas , Colorantes Fluorescentes , Infarto de la Arteria Cerebral Media/patología , Ataque Isquémico Transitorio/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/patología , Neuronas/patología , Compuestos Orgánicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA