Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Neurobiol ; 54(1): 115-124, 2017 01.
Article in English | MEDLINE | ID: mdl-26732594

ABSTRACT

Previous study demonstrates that intracerebral hemorrhage (ICH) promotes microglia activation and inflammation. However, the exact mechanism of microglia activation induced by ICH is not clear. In this experiment, microglia autophagy was examined using electron microscopy, conversion of light chain 3(LC3), and monodansylcadaverine (MDC) staining to detect autophagic vacuoles. We found that ICH induced microglia autophagy and activation. The suppression of autophagy using either pharmacologic inhibitors (3-methyladenine, bafilomycin A1) or RNA interference in essential autophagy genes (BECN1 and ATG5) decreased the microglia activation and inflammation in ICH. Moreover, autophagy inhibitors reduced brain damage in ICH. In conclusion, these data indicate that ICH contributes to microglia autophagic activation through BECN1 and ATG5 and provide the therapeutical strategy for ICH.


Subject(s)
Autophagy-Related Protein 5/metabolism , Autophagy/physiology , Beclin-1/metabolism , Cerebral Hemorrhage/metabolism , Microglia/metabolism , Signal Transduction/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine/therapeutic use , Animals , Autophagy/drug effects , Autophagy-Related Protein 5/antagonists & inhibitors , Beclin-1/antagonists & inhibitors , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/pathology , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythrocytes/pathology , Macrolides/pharmacology , Macrolides/therapeutic use , Mice , Microglia/drug effects , Microglia/pathology , Signal Transduction/drug effects
2.
Immunol Lett ; 179: 114-121, 2016 11.
Article in English | MEDLINE | ID: mdl-27717876

ABSTRACT

Neuroinflammation plays an important role in the recovery of brain injury in ICH. Macrophage is the major executor in the neuroinflammation and initiates neurological defects. Programmed death 1 (PD-1) delivers inhibitory signals that regulate the balance between T cell activation, tolerance, and immunopathology. PD-1 expression by macrophages plays a pathologic role in the innate inflammatory response. However, the exact role of PD-1 on inflammatory responses following ICH has not been well identified. In this experiment, PD-1 KO (PD-1 -/-) ICH mice and Wild-type (WT) ICH mice were caused by intracranial injection of type IV collagenase. The level of macrophage activation, inflammatory cytokines and fibrinogen-like protein 2 (Fgl-2) were detected using immunofluorescence staining and ELISA assays. In addition, brain edema and neurological scores of ICH mice were also measured. Our data demonstrated that ICH promoted PD-1 expression of macrophage and enhanced inflammatory cytokines and Fgl-2 concentrations. PD-1 -/- mice exhibited significantly higher expression of the inflammatory cytokines which initiate Fgl-2, than did their wild-type (WT) littermates. As a result, macrophage activation, cerebral edema and neurological deficit scores of PD-1 -/- mice were higher. In conclusion, our data demonstrate that PD-1 plays a vital role in brain inflammation via regulation of Fgl-2 after ICH, and that manipulation of PD-1 might be a promising therapeutical target in ICH.


Subject(s)
Cerebral Hemorrhage/complications , Encephalitis/etiology , Encephalitis/metabolism , Fibrinogen/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Programmed Cell Death 1 Receptor/metabolism , Animals , Cerebral Hemorrhage/etiology , Cytokines/metabolism , Disease Models, Animal , Encephalitis/pathology , Fibrinogen/genetics , Gene Expression , Immunohistochemistry , Inflammation Mediators/metabolism , Mice , Mice, Knockout , Programmed Cell Death 1 Receptor/genetics
3.
J Neuroimmunol ; 289: 87-92, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26616876

ABSTRACT

Scavenger receptor A (SRA) has been shown to participate in the pattern recognition of pathogen infection. However, its role in intracerebral hemorrhage has not been well defined. In this study, we detected SRA and TLR4 expression and inflammatory response of microglia treated with erythrocyte lysate in vitro, and observed the cerebral water content and neurological deficit of ICH mice in vivo. We found that SRA deficiency leads to greater sensitivity to erythrocyte lysate-induced inflammatory response. SRA down-regulated inflammatory response expression in microglia by suppressing TLR4-induced activation. Collectively, we have identified the molecular linkage between SRA and the TLR4 signaling pathways in ICH. And our results reveal that SRA has important clinical implications for TLR-targeted immunotherapeutical strategy in ICH.


Subject(s)
Cerebral Hemorrhage/complications , Cerebral Hemorrhage/pathology , Microglia/metabolism , Scavenger Receptors, Class A/metabolism , Toll-Like Receptor 4/metabolism , Analysis of Variance , Animals , Brain Edema/etiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Erythrocytes/cytology , Erythrocytes/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System Diseases/etiology , RNA Interference/physiology , Scavenger Receptors, Class A/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Time Factors , Toll-Like Receptor 4/genetics
4.
J Neuroinflammation ; 12: 206, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26552593

ABSTRACT

BACKGROUND: Intracerebral hemorrhage (ICH) induces microglial activation and the release of inflammatory cytokines, leading to inflammation in the brain. IRAK4, an essential component of the MyD88-dependent pathway, activates subsets of divergent signaling pathways in inflammation. METHODS: In the experiment, microglia were stimulated with erythrocyte lysates, and then miR-367, IRAK4, NF-ĸB activation and downstream proinflammatory mediator production were analyzed. In addition, inflammation, brain edema, and neurological functions in ICH mice were also assessed. RESULTS: Here, we report that ICH downregulated miR-367 expression but upregulated IRAK4 expression in primary microglia. We also demonstrate that miR-367 suppressed IRAK4 expression by directly binding its 3'-untranslated region. MiR-367 inhibited NF-ĸB activation and downstream proinflammatory mediator production. Knocking down IRAK4 in microglia significantly decreased the IRAK4 expression and inhibited the NF-ĸB activation and the downstream production of proinflammatory mediators. In addition, our results indicate that miR-367 could inhibit expression of proinflammatory cytokines, reduce brain edema, and improve neurological functions in ICH mice. CONCLUSIONS: In conclusion, our study demonstrates that miR-367/IRAK4 pathway plays an important role in microglial activation and neuroinflammation in ICH. Our finding also suggests that miR-367 might represent a potential therapeutic target for ICH.


Subject(s)
Inflammation/drug therapy , MicroRNAs/pharmacology , Microglia/drug effects , Protein Serine-Threonine Kinases/drug effects , Animals , Body Water/metabolism , Brain Chemistry , Brain Edema/pathology , Erythrocytes/drug effects , Gene Knockdown Techniques , Inflammation Mediators/metabolism , Injections, Intraventricular , NF-kappa B/drug effects , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
5.
J Neuroimmunol ; 287: 71-5, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26439964

ABSTRACT

Numerous evidence have shown that microglia mediated inflammation plays a pivotal role in the development of brain injury after intracerebral hemorrhage (ICH). Therefore anti-inflammation therapy represents a potentially promising approach to ICH. Recently, NLRP3 inflammasome was discovered to facilitate the inflammatory response. However, the effect of NLRP3 inflammasome after ICH has not been fully studied. To explore the potential of NLRP3 inflammasome, we detected NLRP3 expression, inflammation, brain edema and neurological functions in vitro and in vivo. We found that ICH activated the NLRP3 inflammasome and inflammation. However, NLRP3 RNAi could attenuate inflammation and brain injury after ICH. Therefore, the findings suggested that recombinant adenovirus encoding NLRP3 RNAi might be valuable as a potential strategy for anti-inflammation therapy in ICH.


Subject(s)
Brain Injuries/drug therapy , Carrier Proteins/metabolism , Cerebral Hemorrhage/complications , Inflammation/drug therapy , RNA Interference/physiology , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Brain Edema/etiology , Carrier Proteins/genetics , Caspases/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Embryo, Mammalian , Inflammation/etiology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein , Neurons/drug effects , Transduction, Genetic
6.
J Neurooncol ; 125(1): 23-32, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26290143

ABSTRACT

Malignant glioma is among the most challenging of all cancers to treat successfully. Despite recent advances in surgery, radiotherapy and chemotherapy, current treatment regimens have only a marginal impact on patient survival. In this study, we constructed a novel nanoparticle containing neuritin peptide with grp170. The nanoparticle could elicit a neuritin-specific cytotoxic T lymphocyte response to lyse glioma cells in vitro. In addition, the nanoparticle could inhibit tumor growth and improve the lifespan of tumor-bearing mice in vivo. Taken together, the results demonstrated that the nanoparticle can inhibit tumor growth and represents a promising therapy for glioma.


Subject(s)
Brain Neoplasms/drug therapy , Glioma/drug therapy , Glycoproteins/therapeutic use , HSP70 Heat-Shock Proteins/therapeutic use , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Animals , Brain Neoplasms/immunology , Cell Line, Tumor , Chromium/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glioma/immunology , Humans , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Microscopy, Electron, Transmission , Nanoparticles/administration & dosage , Nanoparticles/ultrastructure , Peptides/therapeutic use , T-Lymphocytes, Cytotoxic/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...