Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Oxid Med Cell Longev ; 2019: 2853534, 2019.
Article in English | MEDLINE | ID: mdl-31011401

ABSTRACT

Salvianolic acid A (SAA) is a bioactive polyphenol extracted from Salviae miltiorrhizae Bunge, which possesses a variety of pharmacological activities. In our previous study, we have demonstrated that SAA effectively attenuates kidney injury and inflammation in an established animal model of 5/6 nephrectomized (5/6Nx) rats. However, there has been limited research regarding the antioxidative effects of SAA on chronic kidney disease (CKD). Here, we examined the antioxidative effects and underlying mechanisms of SAA in 5/6Nx rats. The rats were injected with SAA (2.5, 5, and 10 mg·kg-1·d-1, ip) for 28 days. Biochemical, flow cytometry, and Western blot analyses showed that SAA significantly increased the activities of total superoxide dismutase (T-SOD), glutathione peroxidase (GPx), and catalase (CAT) and lowered the levels of malondialdehyde (MDA), reactive oxygen species (ROS), and NADPH oxidase 4 (NOX-4) in a dose-dependent manner in 5/6Nx rats and in H2O2-induced HK-2 cells in vitro. Moreover, SAA enhanced the activation of the protein kinase B/glycogen synthase kinase-3ß/nuclear factor-erythroid-2-related factor 2 (Akt/GSK-3ß/Nrf2) signaling pathway in a dose-dependent manner and subsequently increased the expression of heme oxygenase-1 (HO-1) in the kidney of 5/6Nx rats, which were consistent with those obtained in H2O2-induced HK-2 cells in vitro shown by Western blot analysis. Furthermore, SAA significantly increased the expression of intranuclear Nrf2 and HO-1 proteins compared to HK-2 cells stimulated by LPS on the one hand, which can be enhanced by QNZ to some extent; on the other hand, SAA significantly lowered the expression of p-NF-κB p65 and ICAM-1 proteins compared to HK-2 cells stimulated by H2O2, which can be abrogated by ML385 to some extent. In conclusion, our results demonstrated that SAA effectively protects the kidney against oxidative stress in 5/6Nx rats. One of the pivotal mechanisms for the protective effects of SAA on kidney injury was mainly related with its antioxidative roles by activating the Akt/GSK-3ß/Nrf2 signaling pathway and inhibiting the NF-κB signaling pathway.


Subject(s)
Caffeic Acids/pharmacology , Kidney/pathology , Lactates/pharmacology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nephrectomy , Oxidative Stress/drug effects , Protective Agents/pharmacology , Signal Transduction/drug effects , Animals , Cell Line , Cell Survival/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Hydrogen Peroxide/toxicity , Lipopolysaccharides , Male , Models, Biological , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley
2.
Acta Pharmacol Sin ; 39(12): 1855-1864, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29795135

ABSTRACT

Salvianolic acid A (SAA) is a minor phenolic carboxylic acid extracted from Salviae miltiorrhizae Bunge (Danshen). SAA exhibits a variety of pharmacological activities, such as antioxidative, anti-thrombotic, neuroprotective, and anti-fibrotic effects, as well as protection from myocardial ischemia and prevention of diabetes and other diseases. Furthermore, SAA has shown renal-protective effects in doxorubicin-induced nephropathy. However, there has been limited research regarding the effects of SAA and underlying mechanisms in chronic kidney disease (CKD). Here, we examined the effects and molecular mechanisms of SAA in an established animal model of 5/6 nephrectomized (5/6Nx) rats. The rats were injected with SAA (2.5, 5, and 10 mg/kg per day, intraperitoneally (ip)) for 28 days. SAA dose-dependently lowered the levels of urine protein, blood urea nitrogen, serum creatinine, plasma total cholesterol, and plasma triglycerides in 5/6Nx rats. Histological examination revealed that SAA dose-dependently attenuated renal pathological lesions, evidenced by reduced renal tubulointerstitial fibrosis by decreasing the expression levels of tumor growth factor-ß1 and α-smooth muscle actin in 5/6Nx rats. Moreover, SAA dose-dependently inhibited the activation of nuclear factor-κB (NF-κB) and p38 mitogen-activated protein kinase (MAPK) signaling pathways, subsequently attenuating the secretion of tumor necrosis factor-α and interleukin-1ß and inhibiting the expression of monocyte chemotactic protein-1, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1 in kidneys of 5/6Nx rats. The above results were consistent with those obtained in lipopolysaccharide-induced HK-2 cells in vitro (a recognized in vitro inflammatory model). In conclusion, our results demonstrated that SAA effectively attenuates kidney injury in 5/6Nx rats. The therapeutic effects of SAA on kidney injury can be attributed to its anti-inflammatory activities through inhibition of the activation of the NF-κB and p38 MAPK signaling pathways.


Subject(s)
Caffeic Acids/therapeutic use , Lactates/therapeutic use , MAP Kinase Signaling System/drug effects , Renal Insufficiency, Chronic/prevention & control , Transcription Factor RelA/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , I-kappa B Proteins/metabolism , Kidney/pathology , Male , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/pathology
3.
J Neurol Sci ; 369: 109-118, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27653874

ABSTRACT

NS1619, a calcium-activated potassium channel (Kca channel) activator, can selectively and time-dependently accelerate the formation of transport vesicles in both the brain tumor capillary endothelium and tumor cells within 15min of treatment and then increase the permeability of the blood-brain tumor barrier (BTB). However, the mechanism involved is still under investigation. Using a rat brain glioma (C6) model, the expression of caveolin-1, FoxO1 and p-FoxO1 protein were examined at different time points after intracarotid infusion of NS1619 at a dose of 30µg/kg/min. Internalization of Cholera toxin subunit (CTB) labeled fluorescently was monitored by flow cytometry. The expression of caveolin-1 and FoxO1 protein at tumor microvessels was enhanced and caveolae-mediated CTB endocytosis was increased by NS1619 infusion for 15min. Compared with the 15min group, the expression of caveolin-1 protein was significantly decreased and the level of phosphorylation of FoxO1 was significantly increased in the NS1619 2h group. In addition, inhibitors of reactive oxygen species (ROS) or PI3K or PKB significantly attenuated the level of FoxO1 phosphorylation and also increased the expression of caveolin-1 protein in Human Brain Microvascular Endothelial Cells (HBMECs) cocultured with human glioma cells (U87) 2h after NS1619 treatment. This led to the conclusion that NS1619-mediated transport vesicle increase is, at least partly, related to the ROS/PI3K/PKB/FoxO1 signaling pathway.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Caveolin 1/metabolism , Endothelial Cells/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Brain Neoplasms/pathology , Carotid Arteries/cytology , Caveolin 1/genetics , Cell Line, Tumor , Cholera Toxin/metabolism , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Glioma/pathology , Humans , Male , Morpholines/pharmacology , Rats , Rats, Wistar , Time Factors , Xenograft Model Antitumor Assays
4.
Neuropharmacology ; 75: 407-15, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23973310

ABSTRACT

Adenosine 5'-triphosphate-sensitive potassium channel (KATP channel) activator, minoxidil sulfate (MS), can selectively increase the permeability of the blood-tumor barrier (BTB); however, the mechanism by which this occurs is still under investigation. Using a rat brain glioma (C6) model, we first examined the expression levels of occludin and claudin-5 at different time points after intracarotid infusion of MS (30 µg/kg/min) by western blotting. Compared to MS treatment for 0 min group, the protein expression levels of occludin and claudin-5 in brain tumor tissue of rats showed no changes within 1 h and began to decrease significantly after 2 h of MS infusion. Based on these findings, we then used an in vitro BTB model and selective inhibitors of diverse signaling pathways to investigate whether reactive oxygen species (ROS)/RhoA/PI3K/PKB pathway play a key role in the process of the increase of BTB permeability induced by MS. The inhibitor of ROS or RhoA or PI3K or PKB significantly attenuated the expression of tight junction (TJ) protein and the increase of the BTB permeability after 2 h of MS treatment. In addition, the significant increases in RhoA activity and PKB phosphorylation after MS administration were observed, which were partly inhibited by N-2-mercaptopropionyl glycine (MPG) or C3 exoenzyme or LY294002 pretreatment. The present study indicates that the activation of signaling cascades involving ROS/RhoA/PI3K/PKB in BTB was required for the increase of BTB permeability induced by MS. Taken together, all of these results suggested that MS might increase BTB permeability in a time-dependent manner by down-regulating TJ protein expression and this effect could be related to ROS/RhoA/PI3K/PKB signal pathway.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Neoplasms/pathology , Glioma/pathology , Minoxidil/analogs & derivatives , Signal Transduction/drug effects , Vasodilator Agents/pharmacology , Animals , Blood-Brain Barrier/physiopathology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Claudins/metabolism , Dose-Response Relationship, Drug , Electric Impedance , Enzyme Inhibitors/pharmacology , Horseradish Peroxidase/pharmacokinetics , In Vitro Techniques , Male , Minoxidil/pharmacology , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , rhoA GTP-Binding Protein/metabolism
5.
J Mol Neurosci ; 51(2): 344-51, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23494637

ABSTRACT

Curcumin (Cur) is a major active component of the food flavor turmeric isolated from the powdered dry rhizome of Curcuma longa Linn., which has been used in traditional Chinese medicine to ameliorate intracerebral ischemic damage and reduce brain edema. However, the effects of Cur on the disruption of the blood-brain barrier (BBB) induced by brain ischemia are still unclear. The effects of Cur on the disruption of BBB and changes of tight junction (TJ) proteins induced by oxygen glucose deprivation (OGD) were studied in BBB in vitro. The transendothelial electrical resistance and the flux of horseradish peroxidase in BBB in vitro were measured. The expression and localization of the TJ proteins occludin and zonula occludens-1 (ZO-1) were evaluated by Western blots and immunofluorescence microscopy. The protein levels of heme oxygenase-1 (HO-1) were also analyzed via Western blots. Cur attenuated OGD-induced disruption of paracellular permeability and increased the expression of HO-1 protein in rat brain microvascular endothelial cells (RBMECs). After administration of OGD, the expression of occludin and ZO-1 proteins was restored by Cur, and this effect was blocked by a HO-1 inhibitor, zinc protoporphyrin (ZnPP). Cur protects RBMECs against OGD-induced disruption of TJ and barrier dysfunction via the HO-1 pathway. We propose that Cur is capable of improving the barrier function of BBB under ischemic conditions and this beneficial effect might be reversed by a HO-1 inhibitor, ZnPP.


Subject(s)
Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Cell Hypoxia , Curcumin/pharmacology , Endothelial Cells/drug effects , Heme Oxygenase-1/metabolism , Up-Regulation , Animals , Brain/blood supply , Cells, Cultured , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Glucose/metabolism , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Microvessels/cytology , Oxygen/metabolism , Protoporphyrins/pharmacology , Rats , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
6.
J Mol Neurosci ; 48(1): 302-12, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22581438

ABSTRACT

The calcium-activated potassium channel (K (Ca) channel) activator, NS1619, has been shown to selectively and time-dependently increase the permeability of the blood-tumor barrier (BTB) by downregulating the expression of tight junction (TJ) protein. However, the role of signaling cascades in this process has not been precisely elucidated. This study was performed to determine the role of signaling cascades involving reactive oxygen species (ROS)/RhoA/PI3K/PKB in increasing the permeability of the BTB induced by NS1619. Using an in vitro BTB model and selective inhibitors of signaling pathways, we investigated whether ROS/RhoA/PI3K/PKB pathway plays a key role in the process of the increase in BTB permeability induced by NS1619. The results revealed that the BTB permeability was increased and the expression of TJ proteins were significantly decreased by NS1619, and selective inhibitors of identified signaling pathways reversed the observed alterations. Moreover, the significant increases in ROS, RhoA activity, and PKB phosphorylation after NS1619 administration were observed, which were partly inhibited by N-2-mercaptopropionyl glycine or C3 exoenzyme or LY294002 pretreatment. The present study demonstrates that the activation of signaling cascades involving ROS/RhoA/PI3K/PKB in rat brain microvascular endothelial cells was required for the increase in BTB permeability induced by NS1619.


Subject(s)
Benzimidazoles/pharmacology , Blood-Brain Barrier/drug effects , Brain Neoplasms/drug therapy , Endothelial Cells/drug effects , Glioma/drug therapy , Signal Transduction/drug effects , Animals , Blood-Brain Barrier/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Capillary Permeability/drug effects , Capillary Permeability/physiology , Claudin-5/metabolism , Endothelial Cells/metabolism , Glioma/blood supply , Glioma/metabolism , Occludin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Potassium Channels/agonists , Potassium Channels/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Tight Junctions/drug effects , Tight Junctions/metabolism , rhoA GTP-Binding Protein/metabolism
7.
Cell Mol Neurobiol ; 31(8): 1213-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21688124

ABSTRACT

This study was performed to investigate the mechanism of blood-brain barrier (BBB) permeability change, which was induced by aminoguanidine (AG) after surgical brain injury (SBI) in rats. Compared to control group, AG (150 mg/kg, i.p.) significantly reduced Evans blue extravasation into brain tissue at 24 h after surgical resection, it also induced a 32% decrease of malondialdehyde (MDA) values and a 1.1-fold increase of the glutathione (GSH) levels at 12 h after injury. The expression of inducible nitric oxide synthase (iNOS) reached the peak value at 24 h after SBI, which was significantly attenuated after AG treatment. In addition, ZO-1 protein was up-regulated by AG (150 mg/kg) treatment at 24 h after SBI. Our results indicated that AG could protect the BBB after SBI, which could be correlated with antioxidative property, the down-regulation of iNOS and up-regulation of tight junction protein expression.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Injuries/pathology , Guanidines/pharmacology , Animals , Blood-Brain Barrier/physiology , Brain Injuries/metabolism , Glutathione/metabolism , Humans , Male , Malondialdehyde/metabolism , Membrane Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Permeability , Phosphoproteins/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Zonula Occludens-1 Protein
8.
Cell Mol Neurobiol ; 31(4): 629-34, 2011 May.
Article in English | MEDLINE | ID: mdl-21331626

ABSTRACT

This study was performed to determine whether minoxidil sulfate (MS), a selective Adenosine 5'-triphosphate-sensitive potassium channel (K (ATP) channel) activator, has an effect on the expression of caveolin-1 in the rat's brain tumor tissue. Using a rat brain glioma (C6) model, we found that the expression of caveolin-1 protein at tumor sites was greatly increased after intracarotid infusion of MS at a dose of 30 µg/kg/min for 15, 30, and 60 min via Western blot analysis. And the peak value of the caveolin-1 expression was observed in rats with glioma after 15 min of MS perfusion, which was significantly attenuated by reactive oxygen species (ROS) scavenger (N-2-mercaptopropionyl glycine, MPG). In addition, MPG also significantly inhibited the increase of blood-brain tumor barrier (BTB) permeability which was induced by MS. This led to the conclusion that the MS-induced BTB permeability increase may be related to the accelerated formation of caveolin-1 protein, and could be mediated by ROS.


Subject(s)
Brain Neoplasms/metabolism , Caveolin 1/metabolism , Glioma/metabolism , KATP Channels/metabolism , Minoxidil/analogs & derivatives , Up-Regulation/drug effects , Animals , Blood-Brain Barrier/drug effects , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Capillaries/drug effects , Capillaries/metabolism , Disease Models, Animal , Evans Blue/metabolism , Glioma/blood supply , Glioma/pathology , Minoxidil/pharmacology , Permeability/drug effects , Rats , Rats, Wistar , Tiopronin/pharmacology
9.
Neurosci Lett ; 493(3): 140-4, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21334421

ABSTRACT

This study was performed to investigate the mechanism of the blood-brain tumor-barrier (BTB) permeability increase, which was induced by NS1619, a selective K(Ca) channel activator. Using a rat brain glioma (C6) model, we exam the expression of ZO-1 and occludin in mRNA and protein level at different time point after intracarotid infusion of NS1619 (30 µg/kg/min) to tumor sites via RT-PCR and Western blot analysis. The mRNA and protein expression of ZO-1 and occludin had no great change before infusion and began to decrease significantly after 2 h NS1619 infusion, which was significantly attenuated by reactive oxygen species (ROS) scavenger (N-2-mercaptopropionyl glycine, MPG). In addition, MPG also significantly inhibited the increase of BTB permeability and malonaldehyde (MDA) level induced by NS1619. This led to the conclusion that NS1619 could time-dependently increase the BTB permeability by down-regulating the expression of tight junction protein, and this effect could be reversed by ROS.


Subject(s)
Benzimidazoles/pharmacokinetics , Brain Neoplasms/metabolism , Down-Regulation/physiology , Glioma/metabolism , Membrane Proteins/antagonists & inhibitors , Potassium Channels, Calcium-Activated/metabolism , Tight Junctions/metabolism , Animals , Benzimidazoles/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/genetics , Disease Models, Animal , Down-Regulation/drug effects , Glioma/drug therapy , Glioma/pathology , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Occludin , Phosphoproteins/biosynthesis , Potassium Channels, Calcium-Activated/agonists , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Tight Junctions/drug effects , Tight Junctions/pathology , Zonula Occludens-1 Protein
10.
Brain Res ; 1259: 1-6, 2009 Mar 09.
Article in English | MEDLINE | ID: mdl-19401176

ABSTRACT

This study was performed to determine whether dexamethasone (DEX) had an effect on calcium-activated potassium channels (KCa channels) in blood-brain tumor barrier (BTB).Using a rat brain glioma model, we found that the expression of KCa channels protein was significantly increased in brain tumor tissue. And bradykinin-induced increase of KCa channels protein was further enhanced after DEX pretreatment for 3 days. In addition, DEX pretreatment enhanced bradykinin-mediated up-regulation of the density of IKCa in the rat brain C6 cells in vitro BTB. Bradykinin markedly increased BTB permeability independent of DEX pretreatment. All of these results strongly suggest that DEX could regulate the target in the transcellular pathway of BTB-KCa channels.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Blood-Brain Barrier/drug effects , Brain Neoplasms/metabolism , Brain/drug effects , Dexamethasone/pharmacology , Glioma/metabolism , Potassium Channels, Calcium-Activated/metabolism , Animals , Blood-Brain Barrier/metabolism , Bradykinin/pharmacology , Brain/metabolism , Brain Neoplasms/drug therapy , Capillary Permeability , Cell Line, Tumor , Central Nervous System Agents/pharmacology , Disease Models, Animal , Female , Glioma/drug therapy , Membrane Potentials/drug effects , Neoplasm Transplantation , Random Allocation , Rats , Rats, Wistar , Up-Regulation/drug effects
11.
Neurosci Lett ; 452(2): 114-8, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19135131

ABSTRACT

This study was performed to determine whether dexamethasone (DEX) had an effect on calcium-activated potassium channels (K(Ca) channels) and Occludin protein in blood-brain tumor barrier (BTB). Using a rat brain glioma model, we found that the expression of K(Ca) channels protein and Occludin protein was significantly increased in brain tumor tissue after DEX treatment for 3 days. Compared with non-DEX-treated animals, Evans Blue levels were greatly attenuated in DEX-treated animals. These effects were significantly reversed by the glucocorticoid receptor antagonist RU38486. In addition, DEX treatment enhanced the density of I(KCa) in the rat brain microvascular endothelial cells (RBMECs) in vitro BTB. All of these results strongly suggest that DEX could be involved in the regulation of both transcellular and paracellular pathway.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Edema/drug therapy , Brain Neoplasms/complications , Cerebral Arteries/drug effects , Dexamethasone/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Blood-Brain Barrier/physiology , Brain Edema/etiology , Brain Edema/physiopathology , Cerebral Arteries/metabolism , Cerebral Arteries/physiopathology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Evans Blue , Female , Indicators and Reagents , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mifepristone/pharmacology , Occludin , Rats , Rats, Wistar , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/metabolism
12.
Neuroreport ; 20(2): 145-9, 2009 Jan 28.
Article in English | MEDLINE | ID: mdl-19151597

ABSTRACT

The study was performed to investigate whether the ischemic gray matter and white matter show distinct patterns of aquaporin-4 (AQP4) expression in the reperfusion phase using an in-vivo transient spinal cord ischemia model in rats. We investigated to the time course of AQP4 expression at the blood-spinal cord interface by the quantitative immunogold and western blots methods. The results showed that disruption of AQP4 anchoring at the perivascular membrane did not lead to a net loss of protein. This is the first systematic and extensive study fully showing AQP4 expression dynamics after transient spinal cord ischemia and the findings are of major clinical and experimental interest.


Subject(s)
Aquaporin 4/metabolism , Spinal Cord Ischemia/metabolism , Spinal Cord/metabolism , Analysis of Variance , Animals , Blotting, Western , Immunohistochemistry , Male , Neuroglia/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/blood supply , Spinal Cord Injuries/metabolism
13.
Neurosci Lett ; 450(2): 172-5, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-18983897

ABSTRACT

Bradykinin has been shown to increase the permeability of blood-tumor barrier (BTB) selectively. This study was performed to determine whether tumor necrosis factor-alpha (TNF-alpha) was involved in the regulation of this biological process. We found that the levels of TNF-alpha mRNA and heat shock factor-1 (HSF1) protein in C6 cells were markedly up-regulated by bradykinin via real-time RT-PCR and Western blot methods. And the most obvious increase of HSF1 protein and TNF-alpha mRNA in C6 cells were observed at 5 min and 10 min of bradykinin perfusion, respectively. In addition, the radioactivity of TNF-alpha in C6 cells' culture fluid also mostly increased at 15 min of bradykinin perfusion. And the Evans blue content of brain tumor tissues in rats and the concentration of TNF-alpha reached the maximum at 15 min of bradykinin perfusion. Our results suggested that the bradykinin-mediated BTB permeability increase is due to accelerated release of TNF-alpha, which could cause the increase of BTB permeability by promoting to the release HSF1 from neurospongioma cells.


Subject(s)
Blood-Brain Barrier/drug effects , Bradykinin/therapeutic use , Brain Neoplasms/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Animals , Blood-Brain Barrier/physiology , Bradykinin/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Capillary Permeability/drug effects , Capillary Permeability/genetics , Cell Line, Tumor , Chaperonin 10/genetics , Chaperonin 10/metabolism , Disease Models, Animal , Male , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Suppressor Factors, Immunologic/genetics , Suppressor Factors, Immunologic/metabolism , Time Factors , Tumor Necrosis Factor-alpha/genetics , Up-Regulation/drug effects
14.
Brain Res ; 1246: 11-8, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-18951883

ABSTRACT

The study investigated whether bradykinin (BK) preconditioning could regulate the expression of aquaporin-4 (AQP4) using an in vivo transient spinal cord ischemia model in rats. BK was infused continuously via the left femoral artery with infusion pump for 15 min (10 microg/kg/min) then we induced ischemia for 20 min and reperfusion for 24 and 72 h respectively. The results demonstrated that the central part of the white matter exhibited loss of perivascular AQP4 and showed a partial recovery toward 72 h of reperfusion. The border zone of white matter was different from the central part of the white matter by showing no loss of perivascular AQP4 at 24 h of reperfusion but rather a slight increase. BK significantly reduced the expression level of AQP4 protein in the white matter, but it had none of this effect in the gray matter region at 72 h post-reperfusion. There was no difference in AQP4 protein levels between BK group and control group at the two above-mentioned spinal cord regions at 24 h after reperfusion. In addition, the changes in AQP4 protein induced by BK preconditioning were obvious at 72 h after reperfusion, which were accompanied by a reduction of spinal cord edema. Our results demonstrated that the expression of AQP4 protein after spinal cord ischemia/reperfusion was region-specific, time-dependent and also indicated that the attenuation of AQP4 expression induced by BK could be one of the important molecular mechanisms in physiopathology of spinal cord ischemic edema.


Subject(s)
Aquaporin 4/metabolism , Bradykinin/pharmacology , Gene Expression , Ischemia/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord/blood supply , Spinal Cord/metabolism , Analysis of Variance , Animals , Blotting, Western , Disease Models, Animal , Immunohistochemistry , Microscopy, Electron , Random Allocation , Rats , Rats, Sprague-Dawley
15.
Brain Res ; 1162: 1-8, 2007 Aug 08.
Article in English | MEDLINE | ID: mdl-17618607

ABSTRACT

This study was performed to determine whether dexamethasone (DEX) had an effect on ATP-sensitive potassium channels (K(ATP) channels) in blood-brain tumor barrier (BTB). Using a rat brain glioma model, we found that DEX could significantly increase the expression of K(ATP) channels protein at tumor sites. And bradykinin-induced increase of K(ATP) channels protein was further enhanced after DEX pretreatment for 3 consecutive days via Western blots and immunohistochemistry methods. In addition, DEX pretreatment enhanced bradykinin-mediated increase of the density of I(KATP) in the cultured rat C6 glioma cells using the patch-clamp technique in a whole-cell configuration. DEX significantly decreased the BTB permeability, but it did not reduce bradykinin-mediated BTB permeability increase, which were significantly attenuated by the K(ATP) channel antagonist glibenclamide. This led to the conclusion that DEX-mediated change in BTB permeability is, at least partly, due to accelerated formation of K(ATP) channel, an important target in the biochemical regulation of this process.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Antineoplastic Agents, Hormonal/pharmacology , Blood-Brain Barrier/drug effects , Brain Neoplasms , Dexamethasone/pharmacology , Glioma , Potassium Channels, Inwardly Rectifying/metabolism , Thionucleotides/pharmacology , Adenosine Monophosphate/pharmacology , Analysis of Variance , Animals , Bradykinin/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Cell Line, Tumor , Disease Models, Animal , Drug Interactions , Female , Glioma/metabolism , Glioma/pathology , Glioma/secondary , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Patch-Clamp Techniques , Permeability/drug effects , Potassium Channels, Inwardly Rectifying/agonists , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Rats , Rats, Wistar , Xenograft Model Antitumor Assays
16.
Brain Res ; 1144: 33-41, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17331483

ABSTRACT

Bradykinin has been shown to selectively transiently increase the permeability of the blood-brain barrier (BBB). This study was performed to determine whether ATP-sensitive potassium (K(ATP)) channels mediate the increase in permeability of brain tumor microvessels induced by BK. Using a rat brain glioma (C6) model, we found increased expression of K(ATP) channels at tumor sites via Western blot analysis, after intracarotid infusion of bradykinin at a dose of 10 microg/kg/min for 15 min. A significant increase (73.58%) of the integrated density value (IDV) of the K(ATP) channel Kir6.2 subunit was observed in rats with glioma after 10 min of bradykinin perfusion. The over-expression of K(ATP) channels with bradykinin was significantly attenuated by the K(ATP) channel antagonist glibenclamide. Immunohistochemistry and immunolocalization experiments showed that the over-expression of K(ATP) channels was more obvious near tumor capillaries of 10 microm in diameter. I(KATP) modulation by bradykinin in cultured C6 cells was also studied using the patch-clamp technique in a whole-cell configuration. Administration of bradykinin led to a significant opening of K(ATP) channels in a time-dependent manner. This led to the conclusion that the bradykinin-mediated BBB permeability increase is due to accelerated formation of K(ATP) channels, which are thus as an important target in the biochemical regulation of this process.


Subject(s)
Adenosine Triphosphate/pharmacology , Blood-Brain Barrier/drug effects , Bradykinin/pharmacology , Glioma/blood supply , Glioma/metabolism , Potassium Channels, Inwardly Rectifying/physiology , Animals , Cell Line, Tumor , Female , Gene Expression Regulation/drug effects , Glioma/physiopathology , Glyburide/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Neoplasm Metastasis/drug therapy , Patch-Clamp Techniques/methods , Permeability/drug effects , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar , Time Factors , Xenograft Model Antitumor Assays
17.
Neurosci Lett ; 413(2): 126-31, 2007 Feb 14.
Article in English | MEDLINE | ID: mdl-17240062

ABSTRACT

This study investigated whether dexamethasone (DEX) treatment could regulate the expression of aquaporin-4 (AQP4) in rats with intracerebral hemorrhage (ICH). The results demonstrated that DEX significantly reduced AQP4 mRNA level in the perihematomal area compared with control group, but it increased the level in the brain area surrounding the third ventricle at day 1 post-ICH. There was no difference in AQP4 protein levels between DEX group and control group at the two above-mentioned brain regions at day 1 after ICH. The changes in AQP4 protein induced by DEX were marked at day 3 following surgery and still lasted at day 5 post-ICH, which were accompanied by a reduction of brain edema. Our results demonstrated that the expression of AQP4 protein after ICH was region-specific, time-dependent, and also indicated that DEX-induced cerebral edema clearance was correlated with the regulation of AQP4 expression in different brain regions.


Subject(s)
Aquaporin 4/drug effects , Blood-Brain Barrier/drug effects , Brain Edema/drug therapy , Cerebral Hemorrhage/drug therapy , Dexamethasone/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Aquaporin 4/genetics , Aquaporin 4/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Brain Edema/etiology , Brain Edema/physiopathology , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/physiopathology , Dexamethasone/therapeutic use , Down-Regulation/drug effects , Down-Regulation/physiology , Male , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Telencephalon/blood supply , Telencephalon/drug effects , Telencephalon/physiopathology , Third Ventricle/anatomy & histology , Third Ventricle/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...