Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Neural Plast ; 2022: 7670629, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36160326

RESUMEN

Electroacupuncture (EA) therapy has been widely reported to alleviate neuropathic pain with few side effects in both clinical practice and animal studies worldwide. However, little is known about the comparison of the therapeutic efficacy among the diverse EA schemes used for neuropathic pain. The present study is aimed at investigating the therapeutic efficacy discrepancy between the single and combined-acupoint EA and to reveal the difference of mechanisms behind them. Electroacupuncture was given at both Zusanli (ST36) and Huantiao (GB30) in the combined group or ST36 alone in the single group. Paw withdrawal mechanical threshold (PWMT) was measured to determine the pain level. Electrophysiology was performed to detect the effects of EA on synaptic transmission in the spinal dorsal horn of the vGlut2-tdTomato mice. Spinal contents of endogenous opioids, endocannabinoids, and their receptors were examined. Inhibitors of CBR (cannabinoid receptor) and opioid receptors were used to study the roles of opioid and endocannabinoid system (ECS) in EA analgesia. We found that combined-acupoint acupuncture provide stronger analgesia than the single group did, and the former inhibited the synaptic transmission at the spinal level to a greater extent than later. Besides, the high-intensity stimulation at ST36 or normal stimulation at two sham acupoints did not mimic the similar efficacy of analgesia in the combined group. Acupuncture stimulation in single and combined groups both activated the endogenous opioid system. The ECS was only activated in the combined group. Naloxone totally blocked the analgesic effect of single-acupoint EA; however, it did not attenuate that of combined-acupoint EA unless coadministered with CBR antagonists. Hence, in the CCI-induced neuropathic pain model, combined-acupoint EA at ST36 and GB30 is more effective in analgesia than the single-acupoint EA at ST36. EA stimulation at GB30 alone neither provided a superior analgesic effect to EA treatment at ST36 nor altered the content of AEA, 2-AG, CB1 receptor, or CB2 receptor compared with the CCI group. Activation of the ECS is the main contributor of the better analgesia by the combined acupoint stimulation than that induced by single acupoint stimulation.


Asunto(s)
Electroacupuntura , Neuralgia , Puntos de Acupuntura , Analgésicos Opioides , Animales , Endocannabinoides , Ratones , Naloxona , Neuralgia/terapia , Receptor Cannabinoide CB1 , Receptor Cannabinoide CB2 , Receptores Opioides , Médula Espinal , Asta Dorsal de la Médula Espinal
2.
J Pain Res ; 12: 2663-2672, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31564958

RESUMEN

PURPOSE: Chemokine CX3CL1 and its receptor CX3CR1 in the lumbar spinal cord play crucial roles in pain processing. Electroacupuncture (EA) is recognized as an alternative therapy in pain treatment due to its efficacy and safety. However, the analgesic mechanism of EA remains unclear. The aim of this study was to investigate whether EA suppressed complete Freund's adjuvant (CFA)-induced pain via modulating CX3CL1-CX3CR1 pathway. MATERIALS AND METHODS: Inflammatory pain was induced by intraplantar injection of CFA to the left hind paw of Sprague-Dawley rats. EA with 2 Hz for 30 mins was given to bilateral Zusanli acupoints (ST36) on the first and third day after CFA injection. Mechanical allodynia and thermal hyperalgesia were tested with von Frey tests and Hargreaves tests, respectively. The expressions of CX3CL1, CX3CR1 and p38 mitogen-activated protein kinase (MAPK) were quantified with Western blots. The release of IL-1ß, IL-6 and TNF-α were evaluated with ELISA. Recombinant CX3CL1 or control IgG were then injected through intrathecal catheters in the EA-treated CFA model rats. The behavioral tests, p38 MAPK activation and cytokine release were then evaluated. RESULTS: EA significantly inhibited inflammatory pain induced by CFA for 3 days. Meanwhile, EA downregulated the expression of CX3CL1 but not CX3CR1 in the lumbar spinal cord of the CFA rats. Besides, activation of p38 MAPK and the release of pain-related cytokines (IL-1ß, IL-6 and TNF-α) were inhibited by EA. Intrathecal injection of CX3CL1 largely reversed the analgesic effect of EA treatment and re-activated p38 MAPK signaling, and resulted in pro-inflammatory cytokines increase in acupuncture-treated rats. CONCLUSION: Our findings indicate that EA alleviates inflammatory pain via modulating CX3CL1 signaling in lumbar spinal cord, revealing a potential mechanism of anti-nociception of EA in inflammatory pain.

3.
Sci Rep ; 5: 9490, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25830356

RESUMEN

We investigated whether glutamate receptor subunit 2 (GluR2) is involved in EA pretreatment-induced neuroprotection via cannabinoid CB1 receptors (CB1R) after global cerebral ischemia in mice. Two hours after electric acupuncture (EA) pretreatment, global cerebral ischemia (GCI) was induced by bilateral common carotid artery occlusion (BCCAO) for 20 min. The GluR2 expression was examined in the hippocampus after reperfusion. Cell survival, neuronal apoptosis, the Bax/Bcl-2 ratio and neurological scores were evaluated at 24 h after BCCAO in the presence or absence of the GluR2 inhibitor. Furthermore, the GluR2 was determined in the presence and absence of CB1R inhibitor. Our results showed EA pretreatment enhanced expression of GluR2 in the hippocampus 2 h after reperfusion. Moreover, EA pretreatment improved neurological outcome, promoted cell survival, inhibited neuronal apoptosis, and decreased the Bax/Bcl-2 ratio after reperfusion. GluR2 knockdown by GluR2 siRNA effectively reversed the beneficial effects of EA pretreatment. Furthermore, CB1R siRNA and two CB1R antagonists blocked the elevation of GluR2 expression by EA pretreatment, whereas the two CB1R agonists up-regulated GluR2 expression as EA pretreatment. In conclusion, GluR2 up-regulation is involved in neuroprotection of EA pretreatment against GCI through CB1R, suggesting that GluR2 may be a novel target for stroke intervention.


Asunto(s)
Electroacupuntura , Regulación de la Expresión Génica , Receptor Cannabinoide CB1/metabolismo , Receptores AMPA/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Ácidos Araquidónicos/farmacología , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/terapia , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Endocannabinoides/farmacología , Técnicas de Silenciamiento del Gen , Glicéridos/farmacología , Hipocampo/metabolismo , Ratones , Células Piramidales/metabolismo , Interferencia de ARN , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptores AMPA/deficiencia , Reperfusión , Factores de Tiempo , Regulación hacia Arriba
4.
J Neurosci ; 34(40): 13326-35, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25274812

RESUMEN

How general anesthetics cause loss of consciousness is unknown. Some evidence points toward effects on the neocortex causing "top-down" inhibition, whereas other findings suggest that these drugs act via subcortical mechanisms, possibly selectively stimulating networks promoting natural sleep. To determine whether some neuronal circuits are affected before others, we used Morlet wavelet analysis to obtain high temporal resolution in the time-varying power spectra of local field potentials recorded simultaneously in discrete brain regions at natural sleep onset and during anesthetic-induced loss of righting reflex in rats. Although we observed changes in the local field potentials that were anesthetic-specific, there were some common changes in high-frequency (20-40 Hz) oscillations (reductions in frequency and increases in power) that could be detected at, or before, sleep onset and anesthetic-induced loss of righting reflex. For propofol and natural sleep, these changes occur first in the thalamus before changes could be detected in the neocortex. With dexmedetomidine, the changes occurred simultaneously in the thalamus and neocortex. In addition, the phase relationships between the low-frequency (1-4 Hz) oscillations in thalamic nuclei and neocortical areas are essentially the same for natural sleep and following dexmedetomidine administration, but a sudden change in phase, attributable to an effect in the central medial thalamus, occurs at the point of dexmedetomidine loss of righting reflex. Our data are consistent with the central medial thalamus acting as a key hub through which general anesthesia and natural sleep are initiated.


Asunto(s)
Anestésicos Intravenosos/farmacología , Neocórtex/efectos de los fármacos , Vías Nerviosas/fisiología , Propofol/farmacología , Sueño/fisiología , Tálamo/efectos de los fármacos , Animales , Ondas Encefálicas/efectos de los fármacos , Estimulación Eléctrica , Electrodos Implantados , Electroencefalografía , Electromiografía , Neocórtex/fisiología , Vías Nerviosas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Análisis Espectral , Tálamo/fisiología
5.
Int J Biol Sci ; 10(4): 457-65, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24719563

RESUMEN

Stroke has severe consequences in postmenopausal women. As replacement therapy of estrogen have various adverse effects and the undermined outcomes. Genistein, a natural phytoestrogen, has been suggested to be a potential neuroprotective agent for such stroke patients. However, the role of genistein and its underlying mechanism in ovariectomized mice has not yet been evaluated. In the present study, ovariectomized mice were treated with genistein (10 mg/kg) or vehicle daily for two weeks before developing transient cerebral ischemia (middle cerebral artery occlusion). The neurological manifestation was evaluated, and infarct volumes were demonstrated by 2,3,5-triphenyltetrazolium chloride staining at 24 h after reperfusion. In addition, phosphorylation of extracellular signal-regulated kinase (ERK) was detected by Western blotting and immunofluorescence staining, and cellular apoptosis was evaluated in the ischemic penumbra. We found that treatment with genistein reduced infarct volumes, improved neurological outcomes and attenuated cellular apoptosis at 24 h after reperfusion. ERK1/2 showed increased phosphorylation by genistein treatment after reperfusion, and an ERK1/2 inhibitor U0126 abolished this protective effect of genistein in terms of infarct volumes, neurological scores and cellular apoptosis. Our findings indicate that treatment with genistein can reduce the severity of subsequent stroke episodes, and that this beneficial function is associated with ERK activation.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Genisteína/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Fitoestrógenos/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Butadienos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/genética , Femenino , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Nitrilos/farmacología , Fosforilación , Posmenopausia , Regulación hacia Arriba/efectos de los fármacos
6.
J Cereb Blood Flow Metab ; 33(3): 396-406, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23299244

RESUMEN

Our previous studies have shown that hyperbaric oxygen preconditioning (HBO-PC) induces tolerance to cerebral ischemia/reperfusion (I/R). This study aimed to investigate whether SirT1, a class III histone deacetylase, is involved in neuroprotection elicited by HBO-PC in animal and cell culture models of ischemia. Rats were subjected to middle cerebral artery occlusion for 120 minutes after HBO-PC (once a day for 5 days). Primary cultured cortical neurons were exposed to 2 hours of HBO-PC after 2 hours of oxygen-glucose deprivation (OGD). We showed that HBO-PC increased SirT1 protein and mRNA expression, promoted neurobehavioral score, reduced infarct volume, and improved morphology at 24 hours and 7 days after cerebral I/R. Neuroprotection of HBO-PC was attenuated by SirT1 inhibitor EX527 and SirT1 knockdown by short interfering RNA (siRNA), whereas it was mimicked by SirT1 activator resveratrol. Furthermore, HBO-PC enhanced SirT1 expression and cell viability and reduced lactate dehydrogenase release 24 hours after OGD/re-oxygenation. The neuroprotective effect of HBO-PC was emulated through upregulating SirT1 and, reversely, attenuated through downregulating SirT1. The modulation of SirT1 was made by adenovirus infection carrying SirT1 or SirT1 siRNA. Besides, SirT1 increased B-cell lymphoma 2 (Bcl-2) expression and decrease cleaved caspase 3. These results indicate that SirT1 mediates HBO-PC-induced tolerance to cerebral I/R through inhibition of apoptosis.


Asunto(s)
Apoptosis , Isquemia Encefálica/enzimología , Corteza Cerebral/enzimología , Oxigenoterapia Hiperbárica , Proteínas del Tejido Nervioso/biosíntesis , Sirtuina 1/biosíntesis , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Carbazoles/farmacología , Caspasa 3/genética , Caspasa 3/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Corteza Cerebral/patología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Proteínas del Tejido Nervioso/genética , Neuronas/enzimología , Neuronas/patología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
7.
J Neuroinflammation ; 9: 24, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22277256

RESUMEN

BACKGROUND: We have previously reported that electroacupuncture (EA) pretreatment induced tolerance against cerebral ischemic injury, but the mechanisms underlying this effect of EA are unknown. In this study, we assessed the effect of EA pretreatment on the expression of α7 nicotinic acetylcholine receptors (α7nAChR), using the ischemia-reperfusion model of focal cerebral ischemia in rats. Further, we investigated the role of high mobility group box 1 (HMGB1) in neuroprotection mediated by the α7nAChR and EA. METHODS: Rats were treated with EA at the acupoint "Baihui (GV 20)" 24 h before focal cerebral ischemia which was induced for 120 min by middle cerebral artery occlusion. Neurobehavioral scores, infarction volumes, neuronal apoptosis, and HMGB1 levels were evaluated after reperfusion. The α7nAChR agonist PHA-543613 and the antagonist α-bungarotoxin (α-BGT) were used to investigate the role of the α7nAChR in mediating neuroprotective effects. The roles of the α7nAChR and HMGB1 release in neuroprotection were further tested in neuronal cultures exposed to oxygen and glucose deprivation (OGD). RESULTS: Our results showed that the expression of α7nAChR was significantly decreased after reperfusion. EA pretreatment prevented the reduction in neuronal expression of α7nAChR after reperfusion in the ischemic penumbra. Pretreatment with PHA-543613 afforded neuroprotective effects against ischemic damage. Moreover, EA pretreatment reduced infarct volume, improved neurological outcome, inhibited neuronal apoptosis and HMGB1 release following reperfusion, and the beneficial effects were attenuated by α-BGT. The HMGB1 levels in plasma and the penumbral brain tissue were correlated with the number of apoptotic neurons in the ischemic penumbra. Furthermore, OGD in cultured neurons triggered HMGB1 release into the culture medium, and this effect was efficiently suppressed by PHA-543,613. Pretreatment with α-BGT reversed the inhibitory effect of PHA-543,613 on HMGB1 release. CONCLUSION: These data demonstrate that EA pretreatment strongly protects the brain against transient cerebral ischemic injury, and inhibits HMGB1 release through α7nAChR activation in rats. These findings suggest the novel potential for stroke interventions harnessing the anti-inflammatory effects of α7nAChR activation, through acupuncture or pharmacological strategies.


Asunto(s)
Lesiones Encefálicas/prevención & control , Electroacupuntura/métodos , Proteína HMGB1/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Bungarotoxinas/farmacología , Células Cultivadas , Corteza Cerebral/citología , Infarto Cerebral/etiología , Infarto Cerebral/prevención & control , Modelos Animales de Enfermedad , Embrión de Mamíferos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Glucosa/deficiencia , Hipoxia/terapia , Etiquetado Corte-Fin in Situ , Infarto de la Arteria Cerebral Media/complicaciones , Inyecciones Intraventriculares , L-Lactato Deshidrogenasa/metabolismo , Masculino , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Fosfopiruvato Hidratasa/metabolismo , Unión Proteica/efectos de los fármacos , Quinuclidinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Estadísticas no Paramétricas , Sales de Tetrazolio , Tiazoles , Receptor Nicotínico de Acetilcolina alfa 7
8.
Neuropharmacology ; 61(4): 815-24, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21664366

RESUMEN

We previously found that ginsenoside Rd (Rd), one of the main active ingredients in Panax ginseng, protects against ischemic brain damage induced by oxygen-glucose deprivation in vitro and middle cerebral artery occlusion (MCAO) in vivo. Considering stroke happens frequently in aged individuals, we herein sought to further define the protective effects of Rd in the aged mice. 16-18-month-old mice administered with Rd (0.1-200 mg/kg) or vehicle were subjected to transient MCAO. Rd at the doses of 10-50 mg/kg significantly reduced both cortical and striatal infarct volume. This protection was associated with an improvement in neurological function and was sustained for at least 2 weeks after the insult. Importantly, Rd was effective even when administered up to 4 h after recirculation. To evaluate the underlying mechanisms, oxidative DNA damage was identified by 8-hydroxy-deoxyguanosine immunostaining, oxidative protein damage was identified by the assessment of protein carbonyl, and lipid peroxidation was estimated by determining the malondialdehyde formation. Rd significantly suppressed the accumulations of DNA, protein and lipid peroxidation products at 24 h post-ischemia. Rd also protected mitochondria at 4 and 24 h after reperfusion as indicated by preserved respiratory chain complex activities and aconitase activity, lowered mitochondrial hydrogen peroxide production, and hyperpolarized mitochondrial membrane potential. Furthermore, Rd partly enhanced endogenous antioxidant activities following MCAO. Collectively, these findings demonstrated that Rd exerts neuroprotection against transient focal ischemia in the aged brain, which may be associated with the attenuation of redox imbalance.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Ginsenósidos/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Factores de Edad , Animales , Relación Dosis-Respuesta a Droga , Ginsenósidos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción/efectos de los fármacos , Resultado del Tratamiento
9.
Neurotherapeutics ; 8(3): 515-25, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21647765

RESUMEN

Ginsenoside Rd (Rd), one of the main active ingredients in Panax ginseng, has been demonstrated to protect against ischemic cerebral damage in vitro and in vivo. In this study, we aimed to further define the preclinical characteristics of Rd. We show that Rd passes the intact blood-brain barrier and exerts protection in both transient and permanent middle cerebral artery occlusion (MCAO) in rats. In the dose-response study, Rd (10-50 mg/Kg) significantly reduced the infarct volume on postoperative days (PODs) 1, 3, and 7. This protection was associated with an improved neurological outcome for as many as 6 weeks after transient MCAO, as assessed by modified neurological severity score, modified sticky-tape test, and corner test. For comparison, Rd was significantly more effective than edaravone and slightly more effective than N-tert-butyl-alpha-phenylnitrone (PBN). In the therapeutic window study, Rd exhibited remarkable neuroprotection, even when administered for as many as 4 h after the recirculation of transient MCAO or after the onset of permanent MCAO. Furthermore, in female rats or 16-month-old male rats, the salutary effects of Rd were also observed. These findings suggest Rd is a promising neuroprotectant and provide support for future clinical studies to confirm whether Rd is beneficial in ischemic stroke.


Asunto(s)
Infarto Encefálico/prevención & control , Ginsenósidos/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Animales , Conducta Animal/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Infarto Encefálico/etiología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Masculino , Actividad Motora/efectos de los fármacos , Examen Neurológico , Equilibrio Postural/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
10.
Neurochem Int ; 58(3): 391-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21185898

RESUMEN

We previously found that ginsenoside Rd (Rd), one of the main active ingredients in Panax ginseng, attenuates neuronal oxidative damage in vitro induced by hydrogen peroxide and oxygen-glucose deprivation. In this study, we sought to investigate the potential protective effects and associated mechanisms of Rd in a rat model of focal cerebral ischemia. Rats administered with Rd (0.1-200mg/kg) or vehicle was subjected to transient middle cerebral artery occlusion. Rd at the dose of 10-50mg/kg significantly reduced the infarct volume and improved the long-term neurological outcome up to 6 weeks after ischemia. To evaluate the underlying mechanisms, in vivo free radical generation was monitored using microdialysis, oxidative DNA damage was identified by 8-hydroxy-deoxyguanosine immunostaining, oxidative protein damage was identified by the assessment of protein carbonyl and advanced glycosylation end products, and lipid peroxidation was estimated by determining the malondialdehyde and 4-hydroxynonenal formations. Microdialysis results displayed a prominent inhibitory effect of Rd on the hydroxy radical formation trapped as 2,3- and 2,5-DHBA. Early accumulations of DNA, protein and lipid peroxidation products were also suppressed by Rd treatment. Although Rd partly preserved endogenous antioxidant activities in the ischemic penumbra, in sham rats without stroke, endogenous antioxidant activities were not affected by Rd. Furthermore, we assayed sequential inflammatory response in a later phase after ischemia. Rd significantly eliminated inflammatory injury as indicated by the suppression of microglial activation, inducible nitric oxide synthase and cyclooxygenase-2 expression. Collectively, these findings demonstrated that Rd exerts neuroprotection in transient focal ischemia, which may involve early free radicals scavenging pathway and a late anti-inflammatory effect.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Ginsenósidos/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/patología , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Ginsenósidos/uso terapéutico , Masculino , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/fisiología , Panax/química , Ratas , Ratas Sprague-Dawley
11.
Stroke ; 42(2): 389-96, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21183751

RESUMEN

BACKGROUND AND PURPOSE: Our previous study has demonstrated that the rapid tolerance to cerebral ischemia by electroacupuncture (EA) pretreatment was possibly mediated through an endocannabinoid system-related mechanism. The purpose of this study was to investigate whether activation of epsilon protein kinase C (εPKC) was involved in EA pretreatment-induced neuroprotection via cannabinoid receptor type 1 in a rat model of transient focal cerebral ischemia. METHODS: The activation of εPKC in the ipsilateral brain tissues after EA pretreatment was investigated in the presence or absence of cannabinoid receptor antagonists. At 2 hours after the end of EA pretreatment, focal cerebral ischemia was induced by middle cerebral artery occlusion for 120 minutes in rats. The neurobehavioral scores, infarction volumes, neuronal apoptosis, and the expression of Bcl-2 and Bax were evaluated after reperfusion in the presence or absence of εPKC-selective peptide inhibitor (TAT-εV1-2) or activator (TAT-ψεRACK). RESULTS: EA pretreatment enhanced εPKC activation. Systemic delivery of TAT-ψεRACK conferred neuroprotection against a subsequent cerebral ischemic event when delivered 2 hours before ischemia. Pretreatment with EA reduced infarct volumes, improved neurological outcome, inhibited neuronal apoptosis, and increased the Bcl-2-to-Bax ratio after reperfusion, and the beneficial effects were attenuated by TAT-εV1-2. In addition, the blockade of cannabinoid receptor type 1, but not cannabinoid receptor type 2 receptor, reversed the increase in εPKC activation and neuroprotection induced by EA pretreatment. CONCLUSIONS: EA pretreatment may activate endogenous εPKC-mediated anti-apoptosis to protect against ischemic damage after focal cerebral ischemia via cannabinoid receptor type 1, which represents a new mechanism of EA pretreatment-induced rapid tolerance to focal cerebral ischemia in rats.


Asunto(s)
Apoptosis/fisiología , Isquemia Encefálica/metabolismo , Electroacupuntura/métodos , Proteína Quinasa C-epsilon/fisiología , Receptor Cannabinoide CB1/fisiología , Animales , Isquemia Encefálica/prevención & control , Activación Enzimática/fisiología , Masculino , Proteína Quinasa C-epsilon/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Resultado del Tratamiento
12.
Neurol Res ; 31(8): 853-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19278575

RESUMEN

OBJECTIVE: This study investigates the effects of electroacupuncture (EA) preconditioning on blood-brain barrier (BBB) integrity and matrix metalloproteinase-9 (MMP-9) expression in subsequent ischemic hemisphere. METHODS: Focal cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) in rats. Animals were randomly divided into four groups: normal, sham-operated, MCAO and EA groups. In EA group, rats received electroacupuncture stimuli at the Baihui acupoint (GV 20) 30 minutes/day for 5 days. Twenty-four hours after last treatment, the MCAO was performed. The brain water content and BBB permeability were measured 24 hours after MCAO. MMP-9 expression and activity were measured at 6, 12 and 24 hours after MCAO. RESULTS: The results showed that the brain water content of ischemic hemisphere was lower in EA group (81.45 +/- 1.09%) compared with MCAO group (83.98 +/- 1.30%; p<0.05). Similarly, the Evans blue content in EA group (4.90 +/- 1.77 microg/g) was lower compared with MCAO group (9.41 +/- 2.87 microg/g; p<0.05). The protein expression and enzyme activity of MMP-9 increased and reached maximum at 24 hours after reperfusion. However, the protein expression was lower in EA group at 12 and 24 hours after reperfusion (p<0.01, versus MCAO group), and enzyme activity was lower in EA group only at 24 hours (p<0.01, versus MCAO group). DISCUSSION: EA preconditioning could attenuate brain edema and BBB disruption caused by subsequent cerebral ischemia. EA preconditioning could decrease MMP-9 expression and activity, which may be an important mechanism of cerebral ischemic tolerance.


Asunto(s)
Isquemia Encefálica/enzimología , Isquemia Encefálica/terapia , Electroacupuntura , Infarto de la Arteria Cerebral Media/enzimología , Infarto de la Arteria Cerebral Media/terapia , Metaloproteinasa 9 de la Matriz/metabolismo , Animales , Barrera Hematoencefálica/fisiopatología , Western Blotting , Agua Corporal/metabolismo , Encéfalo/enzimología , Encéfalo/patología , Encéfalo/fisiopatología , Edema Encefálico/fisiopatología , Isquemia Encefálica/fisiopatología , Permeabilidad Capilar/fisiología , Electroforesis en Gel de Poliacrilamida , Azul de Evans , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA