Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Pharmacol Sin ; 43(6): 1349-1359, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34697419

ABSTRACT

Pericytes are present tight around the intervals of capillaries, play an essential role in stabilizing the blood-brain barrier, regulating blood flow and immunomodulation, and persistent contraction of pericytes eventually leads to impaired blood flow and poor clinical outcomes in ischemic stroke. We previously show that iptakalim, an ATP-sensitive potassium (K-ATP) channel opener, exerts protective effects in neurons, and glia against ischemia-induced injury. In this study we investigated the impacts of iptakalim on pericytes contraction in stroke. Mice were subjected to cerebral artery occlusion (MCAO), then administered iptakalim (10 mg/kg, ip). We showed that iptakalim administration significantly promoted recovery of cerebral blood flow after cerebral ischemia and reperfusion. Furthermore, we found that iptakalim significantly inhibited pericytes contraction, decreased the number of obstructed capillaries, and improved cerebral microcirculation. Using a collagen gel contraction assay, we demonstrated that cultured pericytes subjected to oxygen-glucose deprivation (OGD) consistently contracted from 3 h till 24 h during reoxygenation, whereas iptakalim treatment (10 µM) notably restrained pericyte contraction from 6 h during reoxygenation. We further showed that iptakalim treatment promoted K-ATP channel opening via suppressing SUR2/EPAC1 complex formation. Consequently, it reduced calcium influx and ET-1 release. Taken together, our results demonstrate that iptakalim, targeted K-ATP channels, can improve microvascular disturbance by inhibiting pericyte contraction after ischemic stroke. Our work reveals that iptakalim might be developed as a promising pericyte regulator for treatment of stroke.


Subject(s)
Ischemic Stroke , Stroke , Adenosine Triphosphate , Animals , Mice , Microcirculation , Pericytes , Propylamines , Stroke/drug therapy
2.
Cell Death Dis ; 12(1): 23, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33414461

ABSTRACT

Extracellular vesicles (EVs), as a novel intercellular communication carrier transferring cargo microRNAs (miRNAs), could play important roles in the brain remodeling process after ischemic stroke. However, the detailed mechanisms involved in EVs derived miRNAs-mediated cellular interactions in the brain remain unclear. Several studies indicated that microRNA-98 (miR-98) might participate in the pathogenesis of ischemic stroke. Here, we showed that expression of miR-98 in penumbra field kept up on the first day but dropped sharply on the 3rd day after ischemic stroke in rats, indicating that miR-98 could function as an endogenous protective factor post-ischemia. Overexpression of miR-98 targeted inhibiting platelet activating factor receptor-mediated microglial phagocytosis to attenuate neuronal death. Furthermore, we showed that neurons transferred miR-98 to microglia via EVs secretion after ischemic stroke, to prevent the stress-but-viable neurons from microglial phagocytosis. Therefore, we reveal that EVs derived miR-98 act as an intercellular signal mediating neurons and microglia communication during the brain remodeling after ischemic stroke. The present work provides a novel insight into the roles of EVs in the stroke pathogenesis and a new EVs-miRNAs-based therapeutic strategy for stroke.


Subject(s)
Brain Ischemia/genetics , Extracellular Vesicles/metabolism , MicroRNAs/metabolism , Microglia/metabolism , Neurons/metabolism , Acute Disease , Animals , Disease Models, Animal , Humans , Ischemic Stroke , Phagocytosis , Rats
3.
Front Immunol ; 11: 178, 2020.
Article in English | MEDLINE | ID: mdl-32194542

ABSTRACT

Background: Glioblastoma (GBM) is one of the most malignant and aggressive primary brain tumors. The incurability of glioblastoma is heavily influenced by the glioma microenvironment. FTY720, a potent immunosuppressant, has been reported to exert anti-tumor effects in glioblastoma. However, the impact of FTY720 on the glioma microenvironment remains unclear. Methods: We examined the effects of FTY720 on the distribution and polarization of glioma-associated microglia and macrophages (GAMs) in glioma-bearing rats using immunofluorescence staining. qRT-PCR and Western blotting were used to detect the expressions of CXCR4 and MAPK pathway-related signal molecules on microglia in the coculture system. The levels of inflammatory factors were tested via ELISA. Wound healing assay and Matrigel invasion assay were used to determine the migration and invasion of C6 glioma cells. Results: We discovered that FTY720 could inhibit the growth, migration, and invasion of glioma by targeting GAMs to impede their effect on glioma cells. Simultaneously, FTY720 could block the chemoattraction of GAMs by inhibiting MAPK-mediated secretion of IL-6 through increased internalization of CXCR4. Moreover, microglia and macrophages are polarized from pro-glioma to an anti-tumor phenotype. Conclusion: These results provide novel insights into the inhibitory effects of FTY720 on glioma by targeting GAMs-glioma interaction in the tumor microenvironment.


Subject(s)
Antineoplastic Agents/administration & dosage , Fingolimod Hydrochloride/administration & dosage , Glioblastoma/drug therapy , Glioblastoma/metabolism , Microglia/drug effects , Receptors, CXCR4/metabolism , Tumor Microenvironment/drug effects , Allografts , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Polarity/drug effects , Disease Models, Animal , Glioblastoma/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Microglia/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, CXCR4/genetics , Signal Transduction/drug effects
4.
Aging Cell ; 17(4): e12774, 2018 08.
Article in English | MEDLINE | ID: mdl-29740932

ABSTRACT

Microglia-mediated neuroinflammation plays a dual role in various brain diseases due to distinct microglial phenotypes, including deleterious M1 and neuroprotective M2. There is growing evidence that the peroxisome proliferator-activated receptor γ (PPARγ) agonist rosiglitazone prevents lipopolysaccharide (LPS)-induced microglial activation. Here, we observed that antagonizing PPARγ promoted LPS-stimulated changes in polarization from the M1 to the M2 phenotype in primary microglia. PPARγ antagonist T0070907 increased the expression of M2 markers, including CD206, IL-4, IGF-1, TGF-ß1, TGF-ß2, TGF-ß3, G-CSF, and GM-CSF, and reduced the expression of M1 markers, such as CD86, Cox-2, iNOS, IL-1ß, IL-6, TNF-α, IFN-γ, and CCL2, thereby inhibiting NFκB-IKKß activation. Moreover, antagonizing PPARγ promoted microglial autophagy, as indicated by the downregulation of P62 and the upregulation of Beclin1, Atg5, and LC3-II/LC3-I, thereby enhancing the formation of autophagosomes and their degradation by lysosomes in microglia. Furthermore, we found that an increase in LKB1-STRAD-MO25 complex formation enhances autophagy. The LKB1 inhibitor radicicol or knocking down LKB1 prevented autophagy improvement and the M1-to-M2 phenotype shift by T0070907. Simultaneously, we found that knocking down PPARγ in BV2 microglial cells also activated LKB1-AMPK signaling and inhibited NFκB-IKKß activation, which are similar to the effects of antagonizing PPARγ. Taken together, our findings demonstrate that antagonizing PPARγ promotes the M1-to-M2 phenotypic shift in LPS-induced microglia, which might be due to improved autophagy via the activation of the LKB1-AMPK signaling pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/drug effects , Microglia/drug effects , PPAR gamma/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinase Kinases , Animals , Benzamides/pharmacology , Cells, Cultured , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Microglia/metabolism , PPAR gamma/metabolism , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Rosiglitazone/pharmacology
5.
J Cell Mol Med ; 22(6): 3159-3166, 2018 06.
Article in English | MEDLINE | ID: mdl-29536648

ABSTRACT

Fingolimod (FTY720) is used as an immunosuppressant for multiple sclerosis. Numerous studies indicated its neuroprotective effects in stroke. However, the mechanism remains to be elucidated. This study was intended to investigate the mechanisms of phosphorylated FTY720 (pFTY720), which was the principle active molecule in regulating astrocyte-mediated inflammatory responses induced by oxygen-glucose deprivation (OGD). Results demonstrated that pFTY720 could protect astrocytes against OGD-induced injury and inflammatory responses. It significantly decreased pro-inflammatory cytokines, including high mobility group box 1 (HMGB1) and tumour necrosis factor-α (TNF-α). Further, studies displayed that pFTY720 could prevent up-regulation of Toll-like receptor 2 (TLR2), phosphorylation of phosphoinositide 3-kinase (PI3K) and nuclear translocation of nuclear factor kappa B (NFκB) p65 subunit caused by OGD. Sphingosine-1-phosphate receptor 3 (S1PR3) knockdown could reverse the above change. Moreover, administration of TLR2/4 blocker abolished the protective effects of pFTY720. Taken together, this study reveals that pFTY720 depends on S1PR3 to protect astrocytes against OGD-induced neuroinflammation, due to inhibiting TLR2/4-PI3K-NFκB signalling pathway.


Subject(s)
Fingolimod Hydrochloride/pharmacology , Inflammation/drug therapy , Receptors, Lysosphingolipid/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Animals , Astrocytes/drug effects , Cultural Deprivation , Cytokines/genetics , Disease Models, Animal , Fingolimod Hydrochloride/chemistry , HMGB1 Protein/genetics , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Inflammation/genetics , Inflammation/pathology , NF-kappa B/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Primary Cell Culture , Rats , Receptors, Lysosphingolipid/chemistry , Signal Transduction/drug effects , Sphingosine-1-Phosphate Receptors , Tumor Necrosis Factor-alpha/genetics
6.
Exp Neurol ; 247: 209-17, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23639832

ABSTRACT

Asthma is the most common chronic childhood illness today. However, little attention is paid for the impacts of chronic asthma-induced hypoxia on cognitive function in children. The present study used immature mice to establish ovalbumin-induced chronic asthma model, and found that chronic asthma impaired learning and memory ability in Morris Water Maze test. Further study revealed that chronic asthma destroyed synaptic structure, impaired long-term potentiation (LTP) maintaining in the CA1 region of mouse hippocampal slices. We found that intermittent hypoxia during chronic asthma resulted in down-regulation of c-fos, Arc and neurogenesis, which was responsible for the impairment of learning and memory in immature mice. Moreover, our results showed that budesonide treatment alone was inadequate for attenuating chronic asthma-induced cognitive impairment. Therefore, our findings indicate that chronic asthma might result in cognitive dysfunction in children, and more attention should be paid for chronic asthma-induced brain damage in the clinical therapy.


Subject(s)
Asthma/complications , Cognition Disorders/etiology , Developmental Disabilities/etiology , Animals , Animals, Newborn , Asthma/blood , Asthma/chemically induced , Asthma/drug therapy , Bronchodilator Agents/therapeutic use , Budesonide/therapeutic use , Chronic Disease , Cognition Disorders/prevention & control , Cytoskeletal Proteins/metabolism , Developmental Disabilities/prevention & control , Disease Models, Animal , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Hippocampus/drug effects , Hippocampus/physiopathology , Hippocampus/ultrastructure , In Vitro Techniques , Ki-67 Antigen/metabolism , Lung/pathology , Lung/ultrastructure , Maze Learning/drug effects , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/metabolism , Ovalbumin/adverse effects , Pneumonia/drug therapy , Pneumonia/etiology , Time Factors , Vascular Endothelial Growth Factor A/blood
7.
PLoS One ; 7(11): e49701, 2012.
Article in English | MEDLINE | ID: mdl-23166749

ABSTRACT

Paeoniflorin (PF), the principal component of Paeoniae Radix prescribed in traditional Chinese medicine, has been reported to exhibit many pharmacological effects including protection against ischemic injury. However, the mechanisms underlying the protective effects of PF on cerebral ischemia are still under investigation. The present study showed that PF treatment for 14 days could significantly inhibit transient middle cerebral artery occlusion (MCAO)-induced over-activation of astrocytes and microglia, and prevented up-regulations of pro-inflamamtory mediators (TNFα, IL-1ß, iNOS, COX(2) and 5-LOX) in plasma and brain. Further study demonstrated that chronic treatment with PF suppressed the activations of JNK and p38 MAPK, but enhanced ERK activation. And PF could reverse ischemia-induced activation of NF-κB signaling pathway. Moreover, our in vitro study revealed that PF treatment protected against TNFα-induced cell apoptosis and neuronal loss. Taken together, the present study demonstrates that PF produces a delayed protection in the ischemia-injured rats via inhibiting MAPKs/NF-κB mediated peripheral and cerebral inflammatory response. Our study reveals that PF might be a potential neuroprotective agent for stroke.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzoates/pharmacology , Brain Ischemia/metabolism , Bridged-Ring Compounds/pharmacology , Glucosides/pharmacology , Inflammation/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Astrocytes/drug effects , Astrocytes/metabolism , Benzoates/administration & dosage , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Ischemia/drug therapy , Bridged-Ring Compounds/administration & dosage , Cerebral Infarction/drug therapy , Cerebral Infarction/metabolism , Cerebral Infarction/pathology , Cyclooxygenase 2/metabolism , Cytochromes c/genetics , Cytochromes c/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Glucosides/administration & dosage , Hippocampus/drug effects , Inflammation/drug therapy , Interleukin-1beta/blood , Interleukin-1beta/genetics , Lipoxygenase/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Monoterpenes , NF-kappa B/antagonists & inhibitors , Neurons/drug effects , Neurons/metabolism , Nitric Oxide Synthase Type II/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
8.
CNS Neurosci Ther ; 18(7): 536-46, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22530672

ABSTRACT

BACKGROUND AND PURPOSE: Resveratrol has been regarded as a promising candidate for cancer prevention and treatment. The present study was to investigate the impact of resveratrol on the antitumor effects of temozolomide (TMZ), a standard treatment regiment of glioblastoma (GBM), in vitro and in vivo. METHODS AND RESULTS: We found that the combination of resveratrol and TMZ significantly resulted in G(2)/M cell cycle arrest by flow cytometry, triggered a robust increase in expression of astrocyte differentiation marker glial fibrillary acid protein (GFAP), downregulated the expression of matrix metalloproteinase-9 (MMP-9) by immunohistochemistry and western blot analysis as well as inhibited cell migration by scratch wound assay. Further study revealed that TMZ in combination with resveratrol remarkably increased reactive oxygen species (ROS) production, which serves as an upstream signal for AMP-activated protein kinase (AMPK) activation. Subsequently, activated AMPK inhibited mTOR signaling and downregulated antiapoptosis protein Bcl-2, which was contributed to the additive antiproliferation effects of combination treatment. In an orthotopic xenograft model of GBM, TMZ plus resveratrol treatment significantly reduced the volume of tumor, which was confirmed by decreased expression of Ki-67, a marker of proliferation index. CONCLUSIONS: Our findings demonstrate for the first time that resveratrol can enhance TMZ-mediated antitumor effects in GBM in vitro and in vivo, via ROS-dependent AMPK-TSC-mTOR signaling pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Stilbenes/administration & dosage , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Dacarbazine/administration & dosage , Drug Synergism , Drug Therapy, Combination , Female , Glioblastoma/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Resveratrol , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Temozolomide , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...