Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Mol Psychiatry ; 20(7): 880-888, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25385367

RESUMEN

Inflammation in the central nervous system (CNS) and disruption of its immune privilege are major contributors to the pathogenesis of multiple sclerosis (MS) and of its rodent counterpart, experimental autoimmune encephalomyelitis (EAE). We have previously identified developmental endothelial locus-1 (Del-1) as an endogenous anti-inflammatory factor, which inhibits integrin-dependent leukocyte adhesion. Here we show that Del-1 contributes to the immune privilege status of the CNS. Intriguingly, Del-1 expression decreased in chronic-active MS lesions and in the inflamed CNS in the course of EAE. Del-1-deficiency was associated with increased EAE severity, accompanied by increased demyelination and axonal loss. As compared with control mice, Del-1(-/-) mice displayed enhanced disruption of the blood-brain barrier and increased infiltration of neutrophil granulocytes in the spinal cord in the course of EAE, accompanied by elevated levels of inflammatory cytokines, including interleukin-17 (IL-17). The augmented levels of IL-17 in Del-1-deficiency derived predominantly from infiltrated CD8(+) T cells. Increased EAE severity and neutrophil infiltration because of Del-1-deficiency was reversed in mice lacking both Del-1 and IL-17 receptor, indicating a crucial role for the IL-17/neutrophil inflammatory axis in EAE pathogenesis in Del-1(-/-) mice. Strikingly, systemic administration of Del-1-Fc ameliorated clinical relapse in relapsing-remitting EAE. Therefore, Del-1 is an endogenous homeostatic factor in the CNS protecting from neuroinflammation and demyelination. Our findings provide mechanistic underpinnings for the previous implication of Del-1 as a candidate MS susceptibility gene and suggest that Del-1-centered therapeutic approaches may be beneficial in neuroinflammatory and demyelinating disorders.


Asunto(s)
Axones/metabolismo , Barrera Hematoencefálica/metabolismo , Proteínas Portadoras/metabolismo , Vaina de Mielina/metabolismo , Neuroinmunomodulación/fisiología , Médula Espinal/metabolismo , Animales , Axones/efectos de los fármacos , Axones/patología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Proteínas de Unión al Calcio , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Proteínas Portadoras/genética , Moléculas de Adhesión Celular , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Granulocitos/patología , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Péptidos y Proteínas de Señalización Intercelular , Interleucina-17/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/patología , Neuroinmunomodulación/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/patología , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo , Índice de Severidad de la Enfermedad , Médula Espinal/efectos de los fármacos , Médula Espinal/patología
2.
Neurochem Res ; 40(5): 1013-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25777256

RESUMEN

p63 is a transcription factor of p53 gene family, which are involved in development, differentiation and cell response to stress; however, its roles in ischemic preconditioning (IPC) in the brain are not clear. In the present study, we investigated the effect of IPC on p63 immunoreactivity caused by 5 min of transient cerebral ischemia in gerbils. IPC was induced by subjecting the gerbils to 2 min of transie ischemia 1 day prior to 5 min of transient ischemia. The animals were randomly assigned to four groups (sham-operated-group, ischemia-operated-group, IPC plus (+)-sham-operated-group and IPC + ischemia-operated-group). The number of viable neurons in the stratum pyramidale of the hippocampal CA1 region (CA1) was significantly increased by IPC + ischemia-operated-group compared with that in the ischemia-operated-group 5 days after ischemic insult. We found that strong p63 immunoreactivity was detected in the CA1 pyramidal neurons in the sham-operated-group, and the immunoreactivity was decreased with time after ischemia-reperfusion. In addition, strong p63 immunoreactivity was newly expressed in microglial cells of the CA1 region from 2 days after ischemia-reperfusion. In all the IPC + sham-operated-groups, p63 immunoreactivity in the CA1 pyramidal neurons was similar to that in the sham-operated-group, and the immunoreactivity was well maintained in the IPC + ischemia-operated-groups after cerebral ischemia. In brief, our present findings show that IPC dramatically protected the reduction of p63 immunoreactivity in the pyramidal neurons of the CA1 region after ischemia-reperfusion, and this result suggests that the expression of p63 may be necessary for neurons to survive after transient cerebral ischemia.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/prevención & control , Precondicionamiento Isquémico/métodos , Fosfoproteínas/biosíntesis , Transactivadores/biosíntesis , Animales , Región CA1 Hipocampal/patología , Regulación de la Expresión Génica , Gerbillinae , Ataque Isquémico Transitorio/patología , Masculino
3.
Neurochem Res ; 39(8): 1553-63, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24880882

RESUMEN

The receptor for advanced glycation end products (RAGE) is a multi-ligand receptor of the immunoglobulin superfamily that has been implicated in multiple neuronal and inflammatory stress processes. In this study, we examined changes in RAGE immunoreactivity and its protein levels in the gerbil hippocampus (CA1-3 regions) after 5 min of transient global cerebral ischemia. The ischemic hippocampus was stained with cresyl violet, neuronal nuclei (a neuron-specific soluble nuclear antigen) antibody and Fluoro-Jade B (a marker for neuronal degeneration). 5 days after ischemia-reperfusion, delayed neuronal death occurred in the stratum pyramidale of the CA1 region. RAGE immunoreactivity was not detected in any regions of the CA1-3 regions of the sham-group; the immunoreactivity was markedly increased only in the CA1 region from 3 days after ischemia-reperfusion. On the other hand, RAGE immunoreactivity was newly expressed in astrocytes, not in microglia. Western blot analysis showed that RAGE protein level was highest at 5 days post-ischemia. In brief, both the RAGE immunoreactivity and protein level were distinctively increased in astrocytes in the ischemic CA1 region from 3 days after transient cerebral ischemia. These results indicate that the increase of RAGE expression in astrocytes after ischemia-reperfusion may be related to the ischemia-caused activation of astrocytes in the ischemic CA1 region.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Ataque Isquémico Transitorio/metabolismo , Receptores Inmunológicos/biosíntesis , Animales , Astrocitos/metabolismo , Astrocitos/patología , Región CA1 Hipocampal/patología , Regulación de la Expresión Génica , Gerbillinae , Ataque Isquémico Transitorio/patología , Masculino , Neuronas/metabolismo , Neuronas/patología , Receptor para Productos Finales de Glicación Avanzada
4.
Cell Mol Neurobiol ; 33(7): 991-1001, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23893372

RESUMEN

The extent of neuronal damage/death in some brain regions is highly correlated to duration time of transient ischemia. In the present study, we carried out neuronal degeneration/death and glial changes in the septum 4 days after 5, 10, 15, and 20 min of transient cerebral ischemia using gerbils. To examine neuronal damage, Fluoro-Jade B (F-J B, a marker for neuronal degeneration) histofluorescence staining was used. F-J B positive ((+)) cells were detected in the septo-hippocampal nucleus (SHN) of the septum only in the 20 min ischemia-group; the mean number of F-J B(+) neurons was 14.9 ± 2.5/400 µm(2) in a section. Gliosis of astrocytes and microglia was examined using anti-glial fibrillary acidic protein (GFAP) and anti-ionized calcium-binding adapter molecule 1 (Iba-1), respectively. In all the ischemia-groups, GFAP- and Iba-1-immunoreactive astrocytes and microglia, respectively, were increased in number, and apparently tended to be increased in their immunoreactivity. Especially, in the 20 min ischemia-group, the number and immunoreactivity of Iba-immunoreactive microglia was highest and strongest in the ischemic SHN 4 days after ischemia-reperfusion. In brief, our findings showed that neuronal damage/death in the SHN occurred and gliosis was apparently increased in the 20 min ischemia-group at 4 days after ischemia-reperfusion.


Asunto(s)
Isquemia Encefálica/patología , Fluoresceínas/metabolismo , Gerbillinae/metabolismo , Gliosis/metabolismo , Gliosis/patología , Neuronas/patología , Tabique del Cerebro/patología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Benzoxazinas , Isquemia Encefálica/complicaciones , Isquemia Encefálica/metabolismo , Técnica del Anticuerpo Fluorescente , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/complicaciones , Masculino , Proteínas de Microfilamentos/metabolismo , Microglía/metabolismo , Microglía/patología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Tabique del Cerebro/metabolismo , Coloración y Etiquetado
5.
Neurochem Res ; 38(1): 74-81, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22987057

RESUMEN

DNA methylation is a key epigenetic modification of DNA that is catalyzed by DNA methyltransferases (Dnmt). Increasing evidences suggest that DNA methylation in neurons regulates synaptic plasticity as well as neuronal network activity. In the present study, we investigated the changes in DNA methyltransferases 1 (Dnmt1) immunoreactivity and its protein levels in the gerbil hippocampal CA1 region after 5 min of transient global cerebral ischemia. CA1 pyramidal neurons were well stained with NeuN (a neuron-specific soluble nuclear antigen) antibody in the sham-group, Four days after ischemia-reperfusion (I-R), NeuN-positive ((+)) cells were significantly decreased in the stratum pyramidale (SP) of the CA1 region, and many Fluro-Jade B (a marker for neuronal degeneration)(+) cells were observed in the SP. Dnmt1 immunoreactivity was well detected in all the layers of the sham-group. Dnmt1 immunoreactivity was hardly detected only in the stratum pyramidale of the CA1 region from 4 days post-ischemia; however, at these times, Dnmt1 immunoreactivity was newly expressed in GABAergic interneurons or astrocytes in the ischemic CA1 region. In addition, the level of Dnmt1 was lowest at 4 days post-ischemia. In brief, both the Dnmt1 immunoreactivity and protein levels were distinctively decreased in the ischemic CA1 region 4 days after transient cerebral ischemia. These results indicate that the decrease of Dnmt1 expression at 4 days post-ischemia may be related to ischemia-induced delayed neuronal death.


Asunto(s)
Región CA1 Hipocampal/enzimología , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , Ataque Isquémico Transitorio/enzimología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Western Blotting , Muerte Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1 , Fluoresceínas , Técnica del Anticuerpo Fluorescente Indirecta , Colorantes Fluorescentes , Gerbillinae , Inmunohistoquímica , Interneuronas/efectos de los fármacos , Interneuronas/enzimología , Masculino , Células Piramidales/efectos de los fármacos , Células Piramidales/enzimología
6.
J Immunol ; 187(3): 1314-21, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21709153

RESUMEN

In intracerebral hemorrhage, microglia become rapidly activated and remove the deposited blood and cellular debris. To survive in a harmful hemorrhagic or posthemorrhagic condition, activated microglia must be equipped with appropriate self-defensive mechanism(s) to resist the toxicity of hemin, a component released from damaged RBCs. In the current study, we found that activation of microglia by pretreatment with LPS markedly reduced their vulnerability to hemin toxicity in vitro. Similarly, intracorpus callosum microinjection of LPS prior to hemin treatment reduced the brain tissue damage caused by hemin and increased microglial density in the penumbra in rats. LPS induced the expressions of inducible NO synthase (iNOS) and heme oxygenase (HO)-1, the rate-limiting enzyme in heme degradation in microglia. The preventive effect by LPS was significantly diminished by an iNOS inhibitor, L-N(6)-(1-iminoethyl)lysine, whereas it was mimicked by a NO donor, diethylamine-NONOate, both suggesting the crucial role of NO in the modulation of hemin-induced toxicity in activated microglia. We further found that NO reduced hemin toxicity via inhibition of hemin-induced activation of JNK and p38 MAPK pathways in microglia. Whereas HO-1 expression in LPS-stimulated microglia was markedly blocked by L-N(6)-(1-iminoethyl)lysine, the HO-1 inhibitor, tin protoporphyrin, increased iNOS expression and decreased the susceptibility of LPS-activated microglia to hemin toxicity. The data indicate that the mutual interaction between NO and HO-1 plays a critical role in modulating the adaptive response of activated microglia to hemin toxicity. Better understanding of the survival mechanism of activated microglia may provide a therapeutic strategy to attenuate the devastating intracerebral hemorrhagic injury.


Asunto(s)
Hemina/toxicidad , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Microglía/efectos de los fármacos , Microglía/enzimología , Óxido Nítrico/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Astrocitos/patología , Células Cultivadas , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Lipopolisacáridos/administración & dosificación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/inmunología , Masculino , Microglía/patología , Microinyecciones , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/patología , Óxido Nítrico/biosíntesis , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
7.
Am J Pathol ; 179(4): 2042-52, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21854743

RESUMEN

A3 adenosine receptor (A3AR) is recognized as a novel therapeutic target for ischemic injury; however, the mechanism underlying anti-ischemic protection by the A3AR agonist remains unclear. Here, we report that 2-chloro-N(6)-(3-iodobenzyl)-5'-N-methylcarbamoyl-4'-thioadenosine (LJ529), a selective A3AR agonist, reduces inflammatory responses that may contribute to ischemic cerebral injury. Postischemic treatment with LJ529 markedly reduced cerebral ischemic injury caused by 1.5-hour middle cerebral artery occlusion, followed by 24-hour reperfusion in rats. This effect was abolished by the simultaneous administration of the A3AR antagonist MRS1523, but not the A2AAR antagonist SCH58261. LJ529 prevented the infiltration/migration of microglia and monocytes occurring after middle cerebral artery occlusion and reperfusion, and also after injection of lipopolysaccharides into the corpus callosum. The reduced migration of microglia by LJ529 could be related with direct inhibition of chemotaxis and down-regulation of spatiotemporal expression of Rho GTPases (including Rac, Cdc42, and Rho), rather than by biologically relevant inhibition of inflammatory cytokine/chemokine release (eg, IL-1ß, TNF-α, and MCP-1) or by direct inhibition of excitotoxicity/oxidative stress (not affected by LJ529). The present findings indicate that postischemic activation of A3AR and the resultant reduction of inflammatory response should provide a promising therapeutic strategy for the treatment of ischemic stroke.


Asunto(s)
Agonistas del Receptor de Adenosina A3/farmacología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/prevención & control , Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Movimiento Celular/efectos de los fármacos , Inflamación/patología , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Infarto Encefálico/complicaciones , Infarto Encefálico/patología , Lesiones Encefálicas/patología , Isquemia Encefálica/complicaciones , Quimiocina CCL2/metabolismo , Glucosa/deficiencia , Inflamación/complicaciones , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Masculino , Microglía/efectos de los fármacos , Microglía/enzimología , Microglía/metabolismo , Microglía/patología , Monocitos/efectos de los fármacos , Monocitos/patología , N-Metilaspartato/toxicidad , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Tionucleósidos/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas de Unión al GTP rho/metabolismo
8.
Br J Pharmacol ; 177(4): 929-944, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31648364

RESUMEN

BACKGROUND AND PURPOSE: Bleeding is one of the most critical adverse effects of antithrombotic drugs, and many efforts have been made to discover novel antiplatelet agents without bleeding complications. Shear stress-induced platelet aggregation (SIPA), where the interaction of von Willebrand factor (vWF) and platelet glycoprotein (GP) Ib constitutes the initial step, is a promising target to overcome bleeding problems, as SIPA occurs only in pathological conditions. Here, we describe SP-8008, a novel modulator of vWF-GP Ib interactions and evaluated its antiplatelet/antithrombotic effects. EXPERIMENTAL APPROACH: Newly synthesized compounds were screened for antiplatelet effects in vitro, using human platelets exposed to high shear stress. Aggregation, intracellular calcium level, granule secretion, and integrin activation were assessed. Molecular modelling using virtual docking and flow cytometry were used to evaluate effects on vWF-GP Ib interactions. Antithrombotic effects in vivo were determined in rats, using arterial thrombosis and shear stress-specific thrombosis. Transection tail bleeding time was used to evaluate adverse effects. KEY RESULTS: SP-8008 was a potent inhibitor of SIPA, with IC50 of 1.44 ± 0.09 µM. SP-8008 effectively and broadly blocked shear stress-induced platelet activation events, without any significant toxicity. Importantly, SP-8008 was highly selective against SIPA, effectively interfering with vWF-GP Ib engagement. Most importantly, SP-8008 exerted significant antithrombotic effects in vivo in both shear stress-specific and arterial thrombosis, without prolonging bleeding time. CONCLUSIONS AND IMPLICATIONS: Our results demonstrated that SP-8008 can be a novel selective antiplatelet agent with improved safety profile.


Asunto(s)
Fibrinolíticos , Agregación Plaquetaria , Animales , Ácido Benzoico/farmacología , Fibrinolíticos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Complejo GPIb-IX de Glicoproteína Plaquetaria , Ratas , Factor de von Willebrand
9.
Mol Med Rep ; 17(1): 1625-1632, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29257227

RESUMEN

Neurofilaments (NFs) including neurofilament­200 kDa (NF­H), neurofilament­165 kDa (NF­M) and neurofilament­68 kDa (NF­L) are major protein constituents of the brain, and serve important roles in the regulation of axonal transport. NF alteration is a key feature in the pathogenesis of neurological disorders involving cognitive dysfunction. In the present study, cognitive impairments were investigated, via assessments using the Morris water maze and passive avoidance tests, in mice following chronic systemic treatment with 1 mg/kg scopolamine (SCO) for 4 weeks. SCO­induced cognitive impairments were significantly observed 1 week following the SCO treatment, and these cognitive deficits were maintained for 4 weeks. However, the NF immunoreactivities and levels were altered differently according to the hippocampal subregion following SCO treatment. NF­H immunoreactivity and levels were markedly altered in all hippocampal subregions, and were significantly increased 1 week following the SCO treatment; thereafter, the immunoreactivity and levels significantly decreased with time. NF­M immunoreactivity and levels gradually decreased in the hippocampus and were significantly decreased 4 weeks following SCO treatment. NF­L immunoreactivity and levels gradually decreased in the hippocampus, and were significantly decreased 2 and 4 weeks following SCO treatment. In conclusion, the results of the present study demonstrated that chronic systemic treatment with SCO induced cognitive impairment from 1 week following SCO treatment, and NF expression was diversely altered according to the hippocampal subregion from 1 week following SCO treatment. These results suggest that SCO­induced changes in NF expression may be associated with cognitive impairment.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Filamentos Intermedios/patología , Antagonistas Muscarínicos/uso terapéutico , Proteínas de Neurofilamentos/análisis , Escopolamina/uso terapéutico , Animales , Disfunción Cognitiva/patología , Hipocampo/patología , Masculino , Ratones , Ratones Endogámicos ICR
10.
Neural Regen Res ; 12(9): 1458-1464, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29089991

RESUMEN

Calbindin D-28K (CB), a Ca2+-binding protein, maintains Ca2+ homeostasis and protects neurons against various insults. Hyperthermia can exacerbate brain damage produced by ischemic insults. However, little is reported about the role of CB in the brain under hyperthermic condition during ischemic insults. We investigated the effects of transient global cerebral ischemia on CB immunoreactivity as well as neuronal damage in the hippocampal formation under hyperthermic condition using immunohistochemistry for neuronal nuclei (NeuN) and CB, and Fluoro-Jade B histofluorescence staining in gerbils. Hyperthermia (39.5 ± 0.2°C) was induced for 30 minutes before and during transient ischemia. Hyperthermic ischemia resulted in neuronal damage/death in the pyramidal layer of CA1-3 area and in the polymorphic layer of the dentate gyrus at 1, 2, 5 days after ischemia. In addition, hyperthermic ischemia significantly decreaced CB immunoreactivity in damaged or dying neurons at 1, 2, 5 days after ischemia. In brief, hyperthermic condition produced more extensive and severer neuronal damage/death, and reduced CB immunoreactivity in the hippocampus following transient global cerebral ischemia. Present findings indicate that the degree of reduced CB immunoreactivity might be related with various neuronal damage/death overtime and corresponding areas after ischemic insults.

11.
Mol Med Rep ; 16(2): 1627-1634, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28627606

RESUMEN

Ischemic preconditioning (IPC) is induced by exposure to brief durations of transient ischemia, which results in ischemic tolerance to a subsequent longer or lethal period of ischemia. In the present study, the effects of IPC (2 min of transient cerebral ischemia) were examined on immunoreactivity of platelet­derived growth factor (PDGF)­BB and on neuroprotection in the gerbil hippocampal CA1 region following lethal transient cerebral ischemia (LTCI; 5 min of transient cerebral ischemia). IPC was subjected to a 2­min sublethal ischemia and a LTCI was given 5­min transient ischemia. The animals in all of the groups were given recovery times of 1, 2 and 5 days and change in PDGF­BB immunoreactivity was examined as was the neuronal damage/death in the hippocampus induced by LTCI. LTCI induced a significant loss of pyramidal neurons in the hippocampal CA1 region 5 days after LTCI, and significantly decreased PDGF­BB immunoreactivity in the CA1 pyramidal neurons from day 1 after LTCI. Conversely, IPC effectively protected the CA1 pyramidal neurons from LTCI and increased PDGF­BB immunoreactivity in the CA1 pyramidal neurons post­LTCI. In conclusion, the results demonstrated that LTCI significantly altered PDGF­BB immunoreactivity in pyramidal neurons in the hippocampal CA1 region, whereas IPC increased the immunoreactivity. These findings indicated that PDGF­BB may be associated with IPC­mediated neuroprotection.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Gerbillinae/metabolismo , Ataque Isquémico Transitorio/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Animales , Becaplermina , Región CA1 Hipocampal/inmunología , Región CA1 Hipocampal/patología , Muerte Celular/fisiología , Hipocampo/metabolismo , Hipocampo/patología , Inmunohistoquímica , Ataque Isquémico Transitorio/patología , Precondicionamiento Isquémico/métodos , Locomoción , Masculino , Neuroprotección , Proteínas Proto-Oncogénicas c-sis/inmunología , Células Piramidales/inmunología , Células Piramidales/metabolismo , Células Piramidales/patología
12.
Brain Pathol ; 27(3): 276-291, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27117068

RESUMEN

Preconditioning by brief ischemic episode induces tolerance to a subsequent lethal ischemic insult, and it has been suggested that reactive oxygen species are involved in this phenomenon. Thioredoxin 2 (Trx2), a small protein with redox-regulating function, shows cytoprotective roles against oxidative stress. Here, we had focused on the role of Trx2 in ischemic preconditioning (IPC)-mediated neuroprotection against oxidative stress followed by a subsequent lethal transient cerebral ischemia. Animals used in this study were randomly assigned to six groups; sham-operated group, ischemia-operated group, IPC plus (+) sham-operated group, IPC + ischemia-operated group, IPC + auranofin (a TrxR2 inhibitor) + sham-operated group and IPC + auranofin + ischemia-operated group. IPC was subjected to a 2 minutes of sublethal transient ischemia 1 day prior to a 5 minutes of lethal transient ischemia. A significant loss of neurons was found in the stratum pyramidale (SP) of the hippocampal CA1 region (CA1) in the ischemia-operated-group 5 days after ischemia-reperfusion; in the IPC + ischemia-operated-group, pyramidal neurons in the SP were well protected. In the IPC + ischemia-operated-group, Trx2 and TrxR2 immunoreactivities in the SP and its protein level in the CA1 were not significantly changed compared with those in the sham-operated-group after ischemia-reperfusion. In addition, superoxide dismutase 2 (SOD2) expression, superoxide anion radical ( O2-) production, denatured cytochrome c expression and TUNEL-positive cells in the IPC + ischemia-operated-group were similar to those in the sham-operated-group. Conversely, the treatment of auranofin to the IPC + ischemia-operated-group significantly increased cell damage/death and abolished the IPC-induced effect on Trx2 and TrxR2 expressions. Furthermore, the inhibition of Trx2R nearly cancelled the beneficial effects of IPC on SOD2 expression, O2- production, denatured cytochrome c expression and TUNEL-positive cells. In brief, this study shows that IPC conferred neuroprotection against ischemic injury by maintaining Trx2 and suggests that the maintenance or enhancement of Trx2 expression by IPC may be a legitimate strategy for therapeutic intervention of cerebral ischemia.


Asunto(s)
Isquemia Encefálica/metabolismo , Región CA1 Hipocampal/metabolismo , Precondicionamiento Isquémico , Neuronas/metabolismo , Neuroprotección/fisiología , Tiorredoxinas/metabolismo , Animales , Auranofina/farmacología , Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/patología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Citocromos c/metabolismo , Inhibidores Enzimáticos/farmacología , Gerbillinae , Precondicionamiento Isquémico/métodos , Masculino , Neuronas/efectos de los fármacos , Neuronas/patología , Neuroprotección/efectos de los fármacos , Estrés Oxidativo/fisiología , Distribución Aleatoria , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Tiorredoxina Reductasa 2/antagonistas & inhibidores , Tiorredoxina Reductasa 2/metabolismo
13.
Mol Neurobiol ; 54(9): 6984-6998, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-27785755

RESUMEN

Ischemic preconditioning (IPC) provides neuroprotection against subsequent severe ischemic insults by specific mechanisms. We tested the hypothesis that IPC attenuates post-ischemic neuronal death in the gerbil hippocampal CA1 region (CA1) throughout hypoxia inducible factor-1α (HIF-1α) and its associated factors such as vascular endothelial growth factor (VEGF) and nuclear factor-kappa B (NF-κB). Lethal ischemia (LI) without IPC increased expressions of HIF-1α, VEGF, and p-IκB-α (/and translocation of NF-κB p65 into nucleus) in CA1 pyramidal neurons at 12 h and/or 1-day post-LI; thereafter, their expressions were decreased in the CA1 pyramidal neurons with time and newly expressed in non-pyramidal cells (pericytes), and the CA1 pyramidal neurons were dead at 5-day post-LI, and, at this point in time, their immunoreactivities were newly expressed in pericytes. In animals with IPC subjected to LI (IPC/LI)-group), CA1 pyramidal neurons were well protected, and expressions of HIF-1α, VEGF, and p-IκB-α (/and translocation of NF-κB p65 into nucleus) were significantly increased compared to the sham-group and maintained after LI. Whereas, treatment with 2ME2 (a HIF-1α inhibitor) into the IPC/LI-group did not preserve the IPC-mediated increases of HIF-1α, VEGF, and p-IκB-α (/and translocation of NF-κB p65 into nucleus) expressions and did not show IPC-mediated neuroprotection. In brief, IPC protected CA1 pyramidal neurons from LI by upregulation of HIF-1α, VEGF, and p-IκB-α expressions. This study suggests that IPC increases HIF-1α expression in CA1 pyramidal neurons, which enhances VEGF expression and NF-κB activation and that IPC may be a strategy for a therapeutic intervention of cerebral ischemic injury.


Asunto(s)
Región CA1 Hipocampal/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ataque Isquémico Transitorio/patología , Precondicionamiento Isquémico , FN-kappa B/metabolismo , Neuroprotección , Células Piramidales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , 2-Metoxiestradiol , Animales , Gerbillinae , Ataque Isquémico Transitorio/metabolismo , Masculino , Inhibidor NF-kappaB alfa/metabolismo , Neuronas/metabolismo
14.
Stroke ; 37(1): 216-22, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16322488

RESUMEN

BACKGROUND AND PURPOSE: In cerebral stroke, the overall mortality rate of older individuals is higher than that of younger individuals. We therefore investigated aging-related changes in brain tissue damage and immune response in response to intracerebral hemorrhage (ICH) in mice. METHODS: ICH was induced by microinjecting autologous whole blood (5 microL) into the striatum of 4- or 14-month-old senescence-accelerated prone (SAMP8) mice or senescence-accelerated resistant (SAMR1) mice. RESULTS: In all groups, neurological deficits occurred within 6 hours and gradually improved after the first day, but improvement was most delayed in 14-month-old SAMP8 mice. Isolectin B4-positive and amoeboid microglia/macrophages were abundantly distributed around and inside the hemorrhagic lesions in 14-month-old SAMP8 mice. In contrast, myeloperoxidase-immunoreactive neutrophils and reactive astrocytes with intensified glial fibrillary acidic protein-stained processes and swollen cytoplasm did not differ in number or distribution between SAMP8 and SAMR1 mice. Regardless of their age, the immunoreactivity of Mn-SOD, a major antioxidant enzyme in mitochondria, was much weaker in SAMP8 than in SAMR1 mice. The expression of inducible nitric oxide, however, was higher in old SAMP8 mice than in the other experimental groups. CONCLUSIONS: These results suggest that activated microglia/monocytes may aggravate intracerebral hemorrhagic damage in old SAMP8 mice. Further studies on the exact role of activated microglia/monocytes and the altered activities of antioxidant enzymes in old SAMP8 mice may provide useful information for ICH-induced brain injury in relation with aging.


Asunto(s)
Lesiones Encefálicas/patología , Encéfalo/patología , Hemorragia Cerebral/etiología , Hemorragia Cerebral/patología , Envejecimiento , Animales , Antioxidantes/metabolismo , Astrocitos/metabolismo , Peso Corporal , Lesiones Encefálicas/complicaciones , Senescencia Celular , Inmunohistoquímica , Macrófagos/metabolismo , Masculino , Ratones , Microglía/metabolismo , Neuronas/patología , Neutrófilos/metabolismo , Estrés Oxidativo , Oxígeno/metabolismo , Peroxidasa/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Factores de Tiempo
15.
Neurosci Res ; 54(2): 133-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16325294

RESUMEN

Cerebral paragonimiasis causes various neurological disorders including seizures, visual impairment and hemiplegia. The excretory-secretory product (ESP) released by Paragonimus westermani has a cysteine protease activity and plays important roles in its migration in the host tissue and modulation of host immune responses. To gain more insight into the pathogenesis of ESP in the brain, we investigated the inflammatory reaction and cerebral injury following microinjection of ESP into rat striatum. The size of injury was maximally observed 3 days after microinjection of ESP and then declined to control levels as astrocytes have repopulated the injury. ED1-positive monocytes and microglia were confluently found inside the injury. The mRNA expression of inducible nitric oxide synthase (iNOS) occurred as early as 9h after ESP injection and then declined to control levels within 1 day. The iNOS inhibitor aminoguanidine largely decreased the expression of iNOS but did not reduce the size of lesion caused by ESP. Interestingly, however, heat inactivation of ESP caused a decrease of injury formation with no altered expression of iNOS. The data indicate that ESP produces brain tissue injury by recruiting activated monocytes/microglia via heat-labile protease activity.


Asunto(s)
Encefalopatías/inducido químicamente , Proteínas del Helminto/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Neuroglía/efectos de los fármacos , Paragonimus/química , Animales , Encefalopatías/patología , Muerte Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Fluoresceínas , Colorantes Fluorescentes , Guanidinas/farmacología , Proteínas del Helminto/química , Inmunohistoquímica , Masculino , Microinyecciones , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Compuestos Orgánicos , ARN/biosíntesis , ARN/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
J Ethnopharmacol ; 186: 169-180, 2016 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-27036628

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Anti-inflammatory therapy has been intensively investigated as a potential strategy for treatment of cerebral stroke. However, despite many positive outcomes reported in animal studies, anti-inflammatory treatments have not proven successful in humans as yet. Although immunomodulatory activity and safety of Cordyceps species (Chinese caterpillar fungi) have been proven in clinical trials and traditional Asian prescriptions for inflammatory diseases, its anti-ischemic effect remains elusive. AIM OF THE STUDY: In the present study, therefore, we investigated the potential therapeutic efficacy of WIB801C, the standardized extract of Cordyceps militaris, for treatment of cerebral ischemic stroke. MATERIALS AND METHODS: The anti-chemotactic activity of WIB801C was assayed in cultured rat microglia/macrophages. Sprague-Dawley rats were subjected to ischemic stroke via either transient (1.5-h tMCAO and subsequent 24-h reperfusion) or permanent middle cerebral artery occlusion (pMCAO for 24-h without reperfusion). WIB801C was orally administered twice at 3- and 8-h (50mg/kg each) after the onset of MCAO. Infarct volume, edema, blood brain barrier and white matter damages, neurological deficits, and long-term survival rates were investigated. The infiltration of inflammatory cells into ischemic lesions was assayed by immunostaining. RESULTS: WIB801C significantly decreased migration of cultured microglia/macrophages. This anti-chemotactic activity of WIB-801C was not mediated via adenosine A3 receptors, although cordycepin, the major ingredient of WIB801C, is known as an adenosine receptor agonist. Post-ischemic treatment with WIB801C significantly reduced the infiltration of ED-1-and MPO-positive inflammatory cells into ischemic lesions in tMCAO rats. WIB801C-treated rats exhibited significantly decreased infarct volume and cerebral edema, less white matter and blood-brain barrier damages, and improved neurological deficits. WIB801C also improved survival rates over 34 days after ischemia onset. A significant reduction in infarct volume and neurobehavioral deficits by WIB801C was also observed in rats subjected to pMCAO. CONCLUSIONS: In summary, post-ischemic treatment of WIB801C reduced infiltration of inflammatory cells into ischemic lesions via inhibition of chemotaxis, which confers long-lasting histological and neurological protection in ischemic brain. WIB801C may be a promising anti-ischemic drug candidate with clinically relevant therapeutic time window and safety.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Cordyceps/química , Extractos Vegetales/farmacología , Animales , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Masculino , Microglía/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Ratas , Ratas Sprague-Dawley
17.
Iran J Basic Med Sci ; 19(6): 624-31, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27482343

RESUMEN

OBJECTIVES: In the present study, we investigated the effect of ischemic preconditioning (IPC) on c-myb immunoreactivity as well as neuronal damage/death after a subsequent lethal transient ischemia in gerbils. MATERIALS AND METHODS: IPC was subjected to a 2 min sublethal ischemia and a lethal transient ischemia was given 5 min transient ischemia. The animals in all of the groups were given recovery times of 1 day, 2 days and 5 days and we examined change in c-myb immunoreactivity as well as neuronal damage/death in the hippocampus induced by a lethal transient ischemia. RESULTS: A lethal transient ischemia induced a significant loss of cells in the stratum pyramidale (SP) of the hippocampal CA1 region at 5 days post-ischemia, and this insult showed that c-myb immunoreactivity in cells of the SP of the CA1 region was significantly decreased at 2 days post-ischemia and disappeared at 5 days post-ischemia. However, IPC effectively prevented the neuronal loss in the SP and showed that c-myb immunoreactivity was constitutively maintained in the SP after a lethal transient ischemia. CONCLUSION: Our results show that a lethal transient ischemia significantly decreased c-myb immunoreactivity in the SP of the CA1 region and that IPC well preserved c-myb immunoreactivity in the SP of the CA1 region. We suggest that the maintenance of c-myb might be related with IPC-mediated neuroprotection after a lethal ischemic insult.

18.
Biomol Ther (Seoul) ; 23(6): 531-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26535078

RESUMEN

Preceding infection or inflammation such as bacterial meningitis has been associated with poor outcomes after stroke. Previously, we reported that intracorpus callosum microinjection of lipopolysaccharides (LPS) strongly accelerated the ischemia/reperfusion-evoked brain tissue damage via recruiting inflammatory cells into the ischemic lesion. Simvastatin, 3-hydroxy-3-methylgultaryl (HMG)-CoA reductase inhibitor, has been shown to reduce inflammatory responses in vascular diseases. Thus, we investigated whether simvastatin could reduce the LPS-accelerated ischemic injury. Simvastatin (20 mg/kg) was orally administered to rats prior to cerebral ischemic insults (4 times at 72, 48, 25, and 1-h pre-ischemia). LPS was microinjected into rat corpus callosum 1 day before the ischemic injury. Treatment of simvastatin reduced the LPS-accelerated infarct size by 73%, and decreased the ischemia/reperfusion-induced expressions of pro-inflammatory mediators such as iNOS, COX-2 and IL-1ß in LPS-injected rat brains. However, simvastatin did not reduce the infiltration of microglial/macrophageal cells into the LPS-pretreated brain lesion. In vitro migration assay also showed that simvastatin did not inhibit the monocyte chemoattractant protein-1-evoked migration of microglial/macrophageal cells. Instead, simvastatin inhibited the nuclear translocation of NF-κB, a key signaling event in expressions of various proinflammatory mediators, by decreasing the degradation of IκB. The present results indicate that simvastatin may be beneficial particularly to the accelerated cerebral ischemic injury under inflammatory or infectious conditions.

19.
Neural Regen Res ; 10(10): 1604-11, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26692857

RESUMEN

Monocarboxylate transporters (MCTs), which carry monocarboxylates such as lactate across biological membranes, have been associated with cerebral ischemia/reperfusion process. In this study, we studied the effect of ischemic preconditioning (IPC) on MCT4 immunoreactivity after 5 minutes of transient cerebral ischemia in the gerbil. Animals were randomly designated to four groups (sham-operated group, ischemia only group, IPC + sham-operated group and IPC + ischemia group). A serious loss of neuron was found in the stratum pyramidale of the hippocampal CA1 region (CA1), not CA2/3, of the ischemia-only group at 5 days post-ischemia; however, in the IPC + ischemia groups, neurons in the stratum pyramidale of the CA1 were well protected. Weak MCT4 immunoreactivity was found in the stratum pyramidale of the CA1 in the sham-operated group. MCT4 immunoreactivity in the stratum pyramidale began to decrease at 2 days post-ischemia and was hardly detected at 5 days post-ischemia; at this time point, MCT4 immunoreactivity was newly expressed in astrocytes. In the IPC + sham-operated group, MCT4 immunoreactivity in the stratum pyramidale of the CA1 was increased compared with the sham-operated group, and, in the IPC + ischemia group, MCT4 immunoreactivity was also increased in the stratum pyramidale compared with the ischemia only group. Briefly, present findings show that IPC apparently protected CA1 pyramidal neurons and increased or maintained MCT4 expression in the stratum pyramidale of the CA1 after transient cerebral ischemia. Our findings suggest that MCT4 appears to play a significant role in the neuroprotective mechanism of IPC in the gerbil with transient cerebral ischemia.

20.
J Neurol Sci ; 358(1-2): 266-75, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26365286

RESUMEN

It is well known that neurons in the dentate gyrus (DG) of the hippocampus are resistant to short period of ischemia. Hyperthermia is a proven risk factor for cerebral ischemia and can produce more extensive brain damage related with mortality rates. The aim of this study was to examine the effect of hyperthermic conditioning (H) on neuronal death, gliosis and expressions of SODs as anti-oxidative enzymes in the gerbil DG following 5 min-transient cerebral ischemia. The animals were randomly assigned to 4 groups: 1) (N+sham)-group was given sham-operation with normothermia (N); 2) (N+ischemia)-group was given 5 min-transient ischemia with N; 3) (H+sham)-group was given sham-operation with H; and 4) (H+ischemia)-group was given 5 min-transient cerebral ischemia with H. H (39±0.5°C) was induced by subjecting the animals to a heating pad for 30 min before and during the operation. In the (N+ischemia)-groups, a significant neuronal death was observed in the polymorphic layer (PL) from 1 day after ischemia-reperfusion. In the (H+ischemia)-groups, neuronal death was also observed in the PL from 1day post-ischemia; the degree of the neuronal death was severer than that in the (N+ischemia)-groups. In addition, we examined the gliosis of astrocytes and microglia using anti-glial fibrillary acidic protein (GFAP) and anti- ionized calcium-binding adapter molecule 1 (Iba-1). GFAP(+) and Iba-1(+) glial cells were much more activated in the (H+ischemia)-groups than those in the (N+ischemia)-groups. On the other hand, immunoreactivities and levels of SOD1 rather than SOD2 were significantly lower in the (H+ischemia)-groups than those in the (N+ischemia)-groups. In brief, on the basis of our findings, we suggest that cerebral ischemic insult with hyperthermic conditioning brings up severer neuronal damage and gliosis in the polymorphic layer through reducing SOD1 expression rather than SOD2 expression in the DG.


Asunto(s)
Giro Dentado/patología , Fiebre/patología , Gliosis/patología , Ataque Isquémico Transitorio/patología , Neuronas/patología , Daño por Reperfusión/patología , Superóxido Dismutasa/metabolismo , Animales , Astrocitos/metabolismo , Muerte Celular , Modelos Animales de Enfermedad , Gerbillinae , Gliosis/etiología , Masculino , Microglía/metabolismo , Superóxido Dismutasa-1
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda