Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int Immunopharmacol ; 63: 43-57, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30075428

RESUMEN

BACKGROUND: Intracerebral hemorrhage (ICH) is a common neurological emergency with higher mortality and disability rate than cerebral ischemia. Although diverse therapeutic interventions have been explored for potential neuroprotection from ICH, no effective drugs until now are available for improvement of survival rate or the life quality of survivors after ICH. Just like cerebral ischemia, inflammatory mechanism is highly thought to play a vital role in hemorrhagic brain injury. Ligustilide (LIG) has potent anti-inflammatory effects, which were shown to be closely related to its neuroprotective effects against ischemic brain injury. Senkyunolide H (SH) and senkyunolide I (SI) are natural degradation products of LIG, which contain the mother nucleus structure of LIG as that of phthalide. However, no reports have been retrieved about the neuroprotective effects of the three phthalide compounds on ICH, especially from the perspectives of inflammatory pathways. Accordingly, this study investigated the neuroprotective potentials and mechanisms of LIG, SH and SI on experimental ICH in mice. METHODS: ICH was induced in adult male CD-1 mice by intracerebral injection of autologous blood. LIG, SH and SI, respectively, was administrated after ICH induction. Neurological deficits, brain edema, injury volume, the number of surviving/dying neurons and inflammatory gene expression were evaluated at 3 days after ICH. RESULTS: Neurological deficits, brain edema, neuronal injury, microglia and astrocytes activation as well as peripheral immune cells infiltration were all significantly improved by LIG and SH, yet SI not. Moreover, the expression of TLR4, p-NF-kB p65, TNF-α and IL-6, was significantly downregulated by LIG and SH treatment. So was Prx1 expression and release. CONCLUSIONS: LIG and SH provide the potent neuroprotective effects against hemorrhagic stroke by inhibiting Prx1/TLR4/NF-kB signaling and the subsequent immune and neuroinflammation lesions.


Asunto(s)
4-Butirolactona/análogos & derivados , Antiinflamatorios/farmacología , Hemorragia Cerebral/metabolismo , Fármacos Neuroprotectores/farmacología , 4-Butirolactona/farmacología , 4-Butirolactona/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/fisiopatología , Proteínas de Homeodominio/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , FN-kappa B/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
2.
Oncotarget ; 8(53): 90979-90995, 2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-29207618

RESUMEN

The inhibition of extracellular inflammatory peroxiredoxin (Prx) signaling appears to be a potential therapeutic strategy for neuroinflammatory injury after acute ischemic stroke. Gastrodin (Gas) is a phenolic glycoside that is used for the treatment of cerebral ischemia, accompanied by regulation of the autoimmune inflammatory response. The present study investigated the neuroprotective effects of Gas and its derivative, Gas-D, with a focus on the potential mechanism associated with inflammatory Prx-Toll-like receptor 4 (TLR4) signaling. Gas-D significantly inhibited Prx1-, Prx2-, and Prx4-induced inflammatory responses in RAW264.7 macrophages and H2O2-mediated oxidative injury in SH-SY5Y nerve cells. In rats, intraperitoneal Gas-D administration 10 h after reperfusion following 2-h middle cerebral artery occlusion (MCAO) ameliorated neurological deficits, brain infarction, and neuropathological alterations, including neuron loss, astrocyte and microglia/macrophage activation, T-lymphocyte invasion, and lipid peroxidation. Delayed Gas-D treatment significantly inhibited postischemic Prx1/2/4 expression and spillage, TLR4 signaling activation, and inflammatory mediator production. In contrast, Gas had no significant effects in either cell model or in MCAO rats under the same conditions. These results indicate that Gas-D may be a drug candidate with an extended therapeutic time window that blocks inflammatory responses and attenuates the expression and secretome of inflammatory Prxs in acute ischemic stroke.

3.
Int Immunopharmacol ; 41: 82-89, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27821296

RESUMEN

The proinflammatory properties of extracellular peroxiredoxins (Prxs) via induction of Toll-like receptor 4 (TLR4) activation have been gradually revealed under diverse stress conditions, including cerebral ischemia but not hemorrhage. Prx1 is proposed to be a major hemorrhagic stress-inducible isoform of Prxs during acute and subacute phases of intracerebral hemorrhage (ICH). However, the potential of Prx1 in the neuroinflammatory injury after ICH remains unclear. This study investigated the proinflammatory effect and underlying mechanism of extracellular Prx1 in cultured murine macrophages and a collagenase-induced mouse ICH model. The current results show that incubation of exogenous Prx1 (0-50nM) with murine RAW264.7 macrophages resulted in increased expression of TLR4, nuclear translocation of nuclear factor κB (NF-κB) p65 and production of proinflammatory mediators (NO, TNF-a and IL-6) in a concentration-dependent manner. In addition, ICH induced murine neurological deficits, cerebral edema and neuropathological alterations, such as neuron injury, astrocyte and microglia/macrophage activation, and neutrophil and T lymphocyte invasion up to 72h after ICH. Moreover, ICH stimulated Prx1 expression and extracellular release, TLR4/NF-κB signaling activation, reflected by increases in TLR4 expression, extracellular signal-regulated kinase (ERK) 1/2 and NF-κB activation, and production of cytokines (TNF-α, IL-6 and IL-17). Taken together, these findings suggest that extracellular Prx1-mediated TLR4/NF-κB pathway activation probably contributes to neuroinflammatory injury after ICH, and thus blocking Prx1-TLR4 signaling might provide a novel anti-neuroinflammatory strategy with extended therapeutic window for hemorrhagic stroke.


Asunto(s)
Hemorragia Cerebral/metabolismo , FN-kappa B/metabolismo , Peroxirredoxinas/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Hemorragia Cerebral/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Transducción de Señal
4.
PLoS One ; 11(10): e0164586, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27716839

RESUMEN

BACKGROUND: Peroxiredoxins (Prxs) are proposed to function as damage-associated molecular patterns (DAMPs) and contribute to post-ischemic neuroinflammation and brain injury by activating Toll-like receptor (TLR) 4 at the acute and subacute phases after ischemic stroke. However, there are few studies concerning the inflammatory profiles of six distinct subtypes of Prxs (Prx1-Prx6). Our previous study demonstrated that the protective effect of ligustilide (LIG) against cerebral ischemia was associated with inhibition of neuroinflammatory response and Prx/TLR4 signaling in rats. Herein, the present study explored the inflammatory members of Prxs and the effect of LIG on their inflammatory responses in macrophages. METHODOLOGY/PRINCIPAL FINDINGS: The murine RAW264.7 macrophages were treated with each of exogenous recombinant Prxs at a range of 1 to 50 nM for 24 h. The WST-1 test showed that Prx3 exhibited a significant cytotoxicity, whereas the rest five Prxs did not affect cellular viability. The quantitative measurements with spectrometry or ELISA indicated that three subtypes, Prx1, Prx2 and Prx4, increased production of proinflammatory mediators, including nitric oxide (NO) metabolites, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) in a concentration-dependent manner. Immunostaining demonstrated that 20 nM Prx1, Prx2 or Prx4 significantly increased expression of TLR4 and iNOS and nuclear translocation of NF-κB p65. However, Prx5 and Prx6 showed no poinflammatory effect in macrophages. Remarkably, LIG treatment effectively inhibited the inflammatory response induced by Prx1, Prx2 and Prx4. CONCLUSION: Three members of Prxs, Prx1, Prx2 and Prx4, are inflammatory DAMPs that induce TLR4 activation and inflammatory response in macrophages, which is effectively inhibited by LIG. These results suggest that inflammatory Prxs-activated macrophages may provide a novel cellular model for screening the potential inhibitors of DAMPs-associated inflammatory diseases such as stroke. Moreover, selective blocking strategies targeting the inflammatory subtypes of Prxs probably provide promising therapeutic approaches with a prolonged time window for stroke.


Asunto(s)
4-Butirolactona/análogos & derivados , Antiinflamatorios/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Peroxirredoxinas/metabolismo , 4-Butirolactona/metabolismo , Animales , Línea Celular , Interleucina-6/metabolismo , Ratones , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
5.
Fitoterapia ; 109: 52-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26704993

RESUMEN

Migraine is a highly prevalent neurovascular disorder in the brain. An optimal therapy for migraine has not yet been developed. Gastrodin (Gas), the main effective constitute from Gastrodiae Rhizoma (Tianma in Chinese), has been indicated for migraine treatment and prophylaxis more than 30 years, with demonstrated safety. However, Gas is a phenolic glycoside, with relatively low concentrations and weak efficacy in the central nervous system. To develop more effective anti-migraine agents, we synthesized a novel Gas derivative (Gas-D). In the present study, comparative pharmacodynamic evaluations of Gas and Gas-D were performed in a model of nitroglycerin (NTG)-induced migraine in rats and the hot-plate test in mice. Following behavioral testing in this migraine model, external jugular vein blood and the trigeminal nucleus caudalis (TNC) were collected to analyze plasma nitric oxide (NO) and calcitonin gene-related peptide (CGRP) concentrations and c-Fos expression in the TNC. The acute oral toxicity of Gas and Gas-D was also examined. We found that Gas-D had potent anti-migraine effects, likely attributable to inhibition of both trigeminal nerve activation at central sites and the peripheral release of CGRP following NO scavenging. Additionally, Gas-D exerted significant anti-nociceptive effect in response to thermal pain compared with Gas. Furthermore, a single dose of 2.048 g/kg Gas or Gas-D presented no acute oral toxicity in mice. Altogether, the potent anti-migraine and anti-hyperalgesic effects of Gas-D suggest that it might be a potentially novel drug candidate for migraine treatment or prophylaxis.


Asunto(s)
Alcoholes Bencílicos/farmacología , Glucósidos/farmacología , Trastornos Migrañosos/tratamiento farmacológico , Dolor/tratamiento farmacológico , Núcleos del Trigémino/efectos de los fármacos , Analgésicos/farmacología , Animales , Alcoholes Bencílicos/síntesis química , Péptido Relacionado con Gen de Calcitonina/sangre , Femenino , Glucósidos/síntesis química , Masculino , Ratones , Ratones Endogámicos ICR , Trastornos Migrañosos/inducido químicamente , Estructura Molecular , Óxido Nítrico/sangre , Nitroglicerina/efectos adversos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Pruebas de Toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...