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.
Mol Med Rep ; 19(1): 327-337, 2019 01.
Article in English | MEDLINE | ID: mdl-30431087

ABSTRACT

Early brain injury (EBI)­induced neuronal apoptosis is primarily responsible for the subsequent complications of aneurysmal subarachnoid hemorrhage (aSAH), which may increase the risk of mortality in patients with aSAH. c­Jun N­terminal kinase (JNK) has been demonstrated to be a promoter of EBI­induced cell apoptosis, although the mechanism has yet to be fully elucidated. The present study aimed to explore whether the role of JNK1 is associated with tumor protein p53 (p53), which is one of the most important factor that triggers cell apoptosis. JNK1 expression was downregulated via in vivo small interfering RNA transfection in an aSAH rat model in order to assess differences in the behavior, survival times, morphology and genetics of the experimental animals. The results revealed that JNK1 inhibition improved the neurological scores and survival times of SAH rats by interrupting cascaded neuronal apoptosis. The interruption of EBI­induced neuronal apoptosis may originate from a decrease in the level of p53 phosphorylation and deactivation of the downstream mitochondrial apoptotic pathway. Taken together, these results suggest that JNK1 may be a promising target for improving the prognosis of patients with aSAH.


Subject(s)
Apoptosis , Brain Injuries/pathology , Mitochondria/pathology , Mitogen-Activated Protein Kinase 8/metabolism , Neurons/pathology , Subarachnoid Hemorrhage/pathology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Brain Injuries/metabolism , Cells, Cultured , Male , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/genetics , Neurons/metabolism , Phosphorylation , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/metabolism , Tumor Suppressor Protein p53/genetics
2.
CNS Neurosci Ther ; 19(12): 926-36, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24165291

ABSTRACT

AIMS: To study the role of curcumin on glioma cells via the SHH/GLI1 pathway in vitro and vivo. METHODS: The effects of curcumin on proliferation, migration, apoptosis, SHH/GLI1 signaling, and GLI1 target genes expression were evaluated in multiple glioma cell lines in vitro. A U87-implanted nude mice model was used to study the role of curcumin on tumor volume and the suppression efficacy of GLI1. RESULTS: Curcumin showed cytotoxic effects on glioma cell lines in vitro. Both mRNA and protein levels of SHH/GLI1 signaling (Shh, Smo, GLI1) were downregulated in a dose- and time-dependent manner. Several GLI1-dependent target genes (CyclinD1, Bcl-2, Foxm1) were also downregulated. Curcumin treatment prevented GLI1 translocating into the cell nucleus and reduced the concentration of its reporter. Curcumin suppressed cell proliferation, colony formation, migration, and induced apoptosis which was mediated partly through the mitochondrial pathway after an increase in the ratio of Bax to Bcl2. Intraperitoneal injection of curcumin in vivo reduced tumor volume, GLI1 expression, the number of positively stained cells, and prolonged the survival period compared with the control group. CONCLUSION: This study shows that curcumin holds a great promise for SHH/GLI1 targeted therapy against gliomas.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Curcumin/therapeutic use , Glioma/drug therapy , Signal Transduction/drug effects , Animals , Brain Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colony-Forming Units Assay , Disease Models, Animal , Glioma/metabolism , Hedgehog Proteins/metabolism , Humans , Kaplan-Meier Estimate , Mice , Transcription Factors/metabolism , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1
3.
Oncol Rep ; 30(6): 2852-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24100685

ABSTRACT

Recent studies have implicated the acid-sensing ion channel 1 (ASIC1), a proton-gated cation channel that belongs to the epithelial sodium channel (ENaC)/Degenerin family, plays an important role in glioma cell migration. Among the ASIC subunits, only ASIC1a has been found be calcium permeable. However, it has not been determined whether Ca2+/calmodulin-dependent protein kinase II (CaMKII) regulates ASIC1 in glioblastoma multiforme (GBM). Herein, we report that ASIC1 and CaMKII assemble to form a functional complex at the plasma membrane of GBM cells. We found that migration ability was significantly attenuated in GBM cells that were pre-treated with autocamtide-2-related inhibitory peptide (AIP), a CaMKII-specific inhibitor, or psalmotoxin 1 (PcTX-1), a selective ASIC1 blocker. Furthermore, the inhibitory effect of AIP or PcTX-1 on migration was diminished when ASIC1 was knocked down in GBM cells; when ASIC1 knockdown GBM cells were concurrently treated with these two inhibitors, cell migration was slightly but significantly decreased. Using whole-cell patch-clamp recordings, we detected an amiloride-sensitive current in GBM cells, and this current was significantly inhibited by both PcTX-1 and AIP. Moreover, the magnitude of this current was dramatically decreased when ASIC1 was knocked down in GBM cells. The addition of AIP failed to further decrease the amplitude of this current. Taken together, these data suggest that ASIC1 and CaMKII form a functional complex in GBM cells. Furthermore, it can be concluded that CaMKII regulates the activity of ASIC1, which is associated with the ability of GBM cells to migrate.


Subject(s)
Acid Sensing Ion Channels/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium/metabolism , Glioblastoma/genetics , Acid Sensing Ion Channels/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Patch-Clamp Techniques
4.
Eur J Pharm Biopharm ; 84(3): 505-16, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23376242

ABSTRACT

Targeting of intracerebral functional regions has been limited by the inability to transport drugs across the blood-brain barrier (BBB) and by poor accumulation in these regions. To overcome these hurdles, liposomes modified with P-aminophenyl-α-d-mannopyranoside (MAN) were used as a fluorescent dye carrier through the BBB and used the specific distribution of liposomes (LIP) modified with MAN (MAN-LIP) to target various functional regions of the brain. An in vitro BBB model was established to evaluate the transendothelial ability of MAN-LIP, and liposomes uptake by C6 glioma cells was analyzed by flow cytometry and live cell imaging. Liposome targeting was evaluated using in vivo and ex vivo imaging. After MAN-LIP administration, the transendothelial ability and the delivery of fluorescent dye to the brain significantly increased. MAN-LIP concentrated in the cortex at 4 h, shifting distribution to the cerebellum and brainstem at 12 h. The fluorescence intensity in the hippocampus and pontine nuclei remained high and stable over a period of 12 h. The results demonstrate that MAN-LIP is able to enhance cellular uptake in vitro and also promotes penetration through the BBB and accumulation in the brain with a distinct spatio-temporal pattern.


Subject(s)
Aniline Compounds/chemistry , Brain/physiology , Drug Carriers , Liposomes/chemistry , Mannosides/chemistry , Animals , Blood-Brain Barrier , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Culture Media/chemistry , Endocytosis , Flow Cytometry , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mice, Inbred BALB C , Rats , Time Factors
5.
CNS Neurosci Ther ; 19(2): 109-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23253187

ABSTRACT

AIMS: To examine a novel strategy to enhance the survival of grafted neural stem cells (NSCs) in stroke model. METHODS: Using a cell counting kit-8 (CCK-8) and TUNEL assay to test the protective effects of p53 inhibitor, pifithrin-α (PFT-α), on oxygen glucose deprivation (OGD) in NSCs. We compared the effects of vehicle + NSCs and FFT-α + NSCs on the efficacy of transplantation in stroke rat model using behavioral analysis, immunohistochemistry, etc. RESULTS: Pifithrin-α increased viability and decreased apoptosis in NSCs after OGD in vitro. By in vivo studies, we showed that the best recovery of neurological function in the stroke rats and the maximum survival of grafted NSCs were found in the PFT-α + NSCs group. Twelve hours after cell transplantation, p53 was localized to the nuclei of grafted NSCs in the vehicle + NSCs group but was primarily localized to the cytoplasm in the PFT-α + NSCs group. The p53-upregulated modulator of apoptosis (PUMA) was highly expressed among the grafted cells in the vehicle + NSCs group compared with that in the PFT-α + NSCs group. CONCLUSION: Our results indicate that PFT-α enhances the survival of grafted NSCs through the inhibition of p53 translocation into the nucleus.


Subject(s)
Benzothiazoles/administration & dosage , Neural Stem Cells/transplantation , Stem Cell Transplantation/methods , Stroke/therapy , Toluene/analogs & derivatives , Tumor Suppressor Protein p53/antagonists & inhibitors , Up-Regulation/physiology , Animals , Cell Nucleus/drug effects , Cell Nucleus/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Combined Modality Therapy/methods , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology , Stroke/genetics , Stroke/metabolism , Toluene/administration & dosage , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL