Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurochem ; 136(4): 871-883, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26617114

ABSTRACT

Interferon regulatory factor 3 (IRF3) is a transcription factor that plays a central role in the innate immune response, apoptosis, and oncogenesis. Previous studies have shown that endogenous IRF3 does not affect stroke in mice; however, paradoxically, elevated IRF3 expression was observed in the rat brains following cerebral ischemia/reperfusion (I/R) injury, indicating that IRF3 may have different functions during stroke in rats than in mice. A clear and comprehensive study of the effect of IRF3 on stroke in rats has been hampered by the lack of an IRF3-knockout rat strain. In this study, a novel IRF3 knockout rat strain and a transgenic rat strain with neuronal-specific IRF3 over-expression (IRF3-TG) were created. Subsequently, the generated IRF3-knockout rats, the neuronal-specific IRF3 over-expressing rats and their corresponding controls were subjected to transient middle cerebral artery occlusion and followed by reperfusion, to investigate the exact role of IRF3 in cerebral I/R in rats. In contrast to the results in mice, IRF3 deficiency in rats provided significant protection against cerebral I/R injury and inhibited neuronal apoptosis, inflammation, and oxidative stress after cerebral I/R injury; the opposite patterns were observed in neuronal-specific IRF3 over-expressing rats. Taken together, these data demonstrate that IRF3 plays a negative regulatory role in cerebral I/R in rats, and IRF3 may be an attractive therapeutic target for preventing stroke. In the present study, we discovered that the transcription factor IRF3, which plays a central role in the innate immune response, apoptosis, and oncogenesis, could exacerbate cerebral ischemia/reperfusion (I/R) injury via activating caspase-dependent neuronal apoptosis, inducing inflammation and oxidative stress. These findings suggest that IRF3 may be an attractive therapeutic target for the prevention of stroke.

2.
Hypertension ; 66(3): 604-16, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26269654

ABSTRACT

Neuronal death after ischemic stroke involves multiple pathophysiological events, as well as a complex molecular mechanism. Inhibiting a single therapeutic target that is involved in several ischemic signaling cascades may be a promising strategy for stroke management. Here, we report the versatile biological roles of tumor necrosis factor receptor-associated factor 3 (TRAF3) in ischemic stroke. Using several genetically manipulated mouse strains, we also demonstrated that TRAF3 inhibition can be neuroprotective. TRAF3 expression, which is robustly induced in response to ischemia/reperfusion (I/R) injury, was detected in neurons. Overexpression of TRAF3 in neurons led to aggravated neuronal loss and enlarged infarcts; these effects were reversed in TRAF3-knockout mice. Neuronal TRAF3 also contributed to c-Jun kinase-, nuclear factor κB- and Rac-1-induced neuronal death, inflammation, and oxidative stress. Mechanistically, we showed that TRAF3 interacts with transforming growth factor-ß-activated kinase 1 (TAK1) and potentiates phosphorylation and activation of TAK1. Phosphorylated TAK1 sequentially initiated activation of nuclear factor κB, Rac-1/NADPH oxidase, and c-Jun kinase/c-Jun signaling cascades. Using a combination of adenoviruses encoding dominant-negative TAK1 and the TAK1 inhibitor 5Z-7-oxozeaenol, we demonstrated that the TRAF3-mediated activation of ischemic cascades was TAK1-dependent. More importantly, the adverse phenotypes observed in TRAF3-overexpressing mice were completely reversed when the TRAF3-TAK1 interaction was prevented. Therefore, we have shown that TRAF3 is a central regulator of ischemic pathways, including nuclear factor κB, Rac-1, and c-Jun kinase signaling, via its interaction with and activation of TAK1. Furthermore, certain components of the TRAF3-TAK1 signaling pathway are potentially promising therapeutic targets in ischemic stroke.


Subject(s)
Brain Ischemia/metabolism , Cell Death/physiology , Neurons/metabolism , Stroke/metabolism , TNF Receptor-Associated Factor 3/metabolism , Animals , Brain Ischemia/pathology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 8/metabolism , NF-kappa B/metabolism , Neurons/pathology , Signal Transduction/physiology , Stroke/pathology , TNF Receptor-Associated Factor 3/genetics , Up-Regulation
3.
J Neurosci ; 35(34): 12047-62, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26311783

ABSTRACT

Cell-surface receptors provide potential targets for the translation of bench-side findings into therapeutic strategies; however, this approach for the treatment of stroke is disappointing, at least partially due to an incomplete understanding of the targeted factors. Previous studies of oncostatin M (OSM), a member of the gp130 cytokine family, have been limited, as mouse models alone may not strongly resemble the human condition enough. In addition, the precise function of OSM in the CNS remains unclear. Here, we report that human OSM is neuroprotective in vivo and in vitro by recruiting OSMRß in the setting of ischemic stroke. Using gain- and loss-of-function approaches, we demonstrated that decreased neuronal OSMRß expression results in deteriorated stroke outcomes but that OSMRß overexpression in neurons is cerebroprotective. Moreover, administering recombinant human OSM to mice before the onset of I/R showed that human OSM can be protective in rodent models of ischemic stroke. Mechanistically, OSM/OSMRß activate the JAK2/STAT3 prosurvival signaling pathway. Collectively, these data support that human OSM may represent a promising drug candidate for stroke treatment. SIGNIFICANCE STATEMENT: OSM, a member of the gp130 cytokine family, regulates neuronal function and survival. OSM engages a second receptor, either LIFRα or OSMRß, before recruiting gp130. However, it is not clear whether OSM/OSMRß signaling is involved in neuroprotection in the setting of ischemic stroke. Recent studies show that, compared with mouse disease models, the OSM receptor system in rats more closely resembles that in humans. In the present study, we use genetic manipulations of OSMRß in both mouse and rat stroke models to demonstrate that OSMRß in neurons is critical for neuronal survival during cerebral ischemic/reperfusion. Interestingly, administration of human OSM also leads to improved stroke outcomes. Therefore, OSM may represent a promising drug candidate for stroke treatment.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/prevention & control , Oncostatin M Receptor beta Subunit/biosynthesis , Oncostatin M/biosynthesis , Stroke/metabolism , Stroke/prevention & control , Animals , Brain Ischemia/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pregnancy , Rats , Rats, Sprague-Dawley , Stroke/pathology
4.
J Pathol ; 237(2): 249-62, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26011492

ABSTRACT

Toll-like receptor (TLR) signalling plays an important role in regulating cerebral ischaemia-reperfusion (I/R) injury. Toll-interacting protein (Tollip) is an endogenous negative modulator of TLR signalling that is involved in several inflammatory diseases. Our previous study showed that Tollip inhibits overload-induced cardiac remodelling. However, the role of Tollip in neurological disease remains unknown. In the present study, we proposed that Tollip might contribute to the progression of stroke and confirmed this hypothesis. We found that Tollip expression was significantly increased in I/R-challenged brain tissue of humans, mice and rats in vivo and in primary neurons subjected to oxygen and glucose deprivation in vitro, indicating the involvement of Tollip in I/R injury. Next, using genetic approaches, we revealed that Tollip deficiency protects mice against I/R injury by attenuating neuronal apoptosis and inflammation, as demonstrated by the decreased expression of pro-apoptotic and pro-inflammatory genes and the increased expression of anti-apoptotic genes. By contrast, neuron-specific Tollip over-expression exerted the opposite effect. Mechanistically, the detrimental effects of Tollip on neuronal apoptosis and inflammation following I/R injury were largely mediated by the suppression of Akt signalling. Additionally, to further support our findings, a Tollip knockout rat strain was generated via CRISPR-Cas9-mediated gene inactivation. The Tollip-deficient rats were also protected from I/R injury, based on dramatic decreases in neuronal apoptosis and ischaemic inflammation through Akt activation. Taken together, our findings demonstrate that Tollip acts as a novel modulator of I/R injury by promoting neuronal apoptosis and ischaemic inflammation, which are largely mediated by suppression of Akt signalling.


Subject(s)
Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/metabolism , Reperfusion Injury/metabolism , Signal Transduction , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Brain/pathology , CRISPR-Cas Systems , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Glucose/deficiency , Humans , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/prevention & control , Inflammation Mediators/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Mice, Knockout , Neurons/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Time Factors , Transfection
5.
J Neurochem ; 134(2): 211-21, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25824575

ABSTRACT

Vinexin-ß is an adaptor protein that regulates cell adhesion, cytoskeletal organization and signal transduction. Our previous work showed that Vinexin-ß protects against cardiac hypertrophy. However, its function in stroke is largely unknown. In the present study, we observed a significant increase in Vinexin-ß expression in both human intracerebral haemorrhage and mouse cerebral ischaemia/reperfusion (I/R) injury model, indicating that Vinexin-ß is involved in stroke. Next, using Vinexin-ß knockout mice, we further demonstrated that Vinexin-ß deficiency significantly protected against cerebral I/R injury, as demonstrated by a dramatic decrease in the infarct volume and an improvement in neurological function. Additionally, immunofluorescence and western blotting showed that the deletion of Vinexin-ß attenuated neuronal apoptosis. Mechanically, we found that Akt signalling was up-regulated in the brains of the Vinexin-ß knockout mice compared with those of the WT control mice after ischaemic injury. Taken together, our results demonstrate that the deletion of Vinexin-ß potently protects against ischaemic injury by inhibiting neuronal apoptosis, and this effect may occur via the up-regulation of Akt signalling. Our findings revealed that Vinexin-ß acts as a novel modulator of ischaemic injury, suggesting that Vinexin-ß may represent an attractive therapeutic target for the prevention of stroke.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Brain Ischemia/metabolism , Muscle Proteins/metabolism , Neurons/pathology , Reperfusion Injury/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Brain Ischemia/pathology , Fluorescent Antibody Technique , Humans , In Situ Nick-End Labeling , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reperfusion Injury/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...