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.
Biomed Pharmacother ; 159: 114228, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36623448

ABSTRACT

BACKGROUND: Canagliflozin (CANA) shows anti-inflammatory and anti-oxidative effects on endothelial cells (ECs). In diabetes mellitus (DM), excessive reactive oxygen species (ROS) generation, increased intracellular calcium (Ca2+) and enhanced extracellular signal regulated kinase (ERK) 1/2 phosphorylation are crucial precursors for inflammasome activation. We hypothesized that: (1) CANA prevents the TNF-α triggered ROS generation in ECs from diabetic donors and in turn suppresses the inflammasome activation; and (2) the anti-inflammatory effect of CANA is mediated via intracellular Ca2+ and ERK1/2. METHODS: Human coronary artery endothelial cells from donors with DM (D-HCAECs) were pre-incubated with either CANA or vehicle for 2 h before exposure to 50 ng/ml TNF-α for 2-48 h. NAC was applied to scavenge ROS, BAPTA-AM to chelate intracellular Ca2+, and PD 98059 to inhibit the activation of ERK1/2. Live cell imaging was performed at 6 h to measure ROS and intracellular Ca2+. At 48 h, ELISA and infra-red western blot were applied to detect IL-1ß, NLRP3, pro-caspase-1 and ASC. RESULTS: 10 µM CANA significantly reduced TNF-α related ROS generation, IL-1ß production and NLRP3 expression (P all <0.05), but NAC did not alter the inflammasome activation (P > 0.05). CANA and BAPTA both prevented intracellular Ca2+ increase in cells exposed to TNF-α (P both <0.05). Moreover, BAPTA and PD 98059 significantly reduced the TNF-α triggered IL-1ß production as well as NLRP3 and pro-caspase-1 expression (P all <0.05). CONCLUSION: CANA suppresses inflammasome activation by inhibition of (1) intracellular Ca2+ and (2) ERK1/2 phosphorylation, but not by ROS reduction.


Subject(s)
Diabetes Mellitus , Inflammasomes , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Canagliflozin/pharmacology , Calcium , Endothelial Cells/metabolism , Caspase 1/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha , Signal Transduction , Interleukin-1beta/metabolism
2.
Int J Mol Sci ; 22(11)2021 Jun 03.
Article in English | MEDLINE | ID: mdl-34205045

ABSTRACT

SGLT-2i's exert direct anti-inflammatory and anti-oxidative effects on resting endothelial cells. However, endothelial cells are constantly exposed to mechanical forces such as cyclic stretch. Enhanced stretch increases the production of reactive oxygen species (ROS) and thereby impairs endothelial barrier function. We hypothesized that the SGLT-2i's empagliflozin (EMPA), dapagliflozin (DAPA) and canagliflozin (CANA) exert an anti-oxidative effect and alleviate cyclic stretch-induced endothelial permeability in human coronary artery endothelial cells (HCAECs). HCAECs were pre-incubated with one of the SGLT-2i's (1 µM EMPA, 1 µM DAPA and 3 µM CANA) for 2 h, followed by 10% stretch for 24 h. HCAECs exposed to 5% stretch were considered as control. Involvement of ROS was measured using N-acetyl-l-cysteine (NAC). The sodium-hydrogen exchanger 1 (NHE1) and NADPH oxidases (NOXs) were inhibited by cariporide, or GKT136901, respectively. Cell permeability and ROS were investigated by fluorescence intensity imaging. Cell permeability and ROS production were increased by 10% stretch; EMPA, DAPA and CANA decreased this effect significantly. Cariporide and GKT136901 inhibited stretch-induced ROS production but neither of them further reduced ROS production when combined with EMPA. SGLT-2i's improve the barrier dysfunction of HCAECs under enhanced stretch and this effect might be mediated through scavenging of ROS. Anti-oxidative effect of SGLT-2i's might be partially mediated by inhibition of NHE1 and NOXs.


Subject(s)
Endothelial Cells/drug effects , Inflammation/drug therapy , Oxidative Stress/drug effects , Sodium-Glucose Transport Proteins/antagonists & inhibitors , Sodium-Hydrogen Exchanger 1/antagonists & inhibitors , Benzhydryl Compounds/pharmacology , Canagliflozin/pharmacology , Cell Membrane Permeability/drug effects , Endothelial Cells/metabolism , Glucosides/pharmacology , Guanidines/pharmacology , Humans , Inflammation/genetics , Inflammation/pathology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Oxidative Stress/genetics , Pyrazoles/pharmacology , Pyridones/pharmacology , Reactive Oxygen Species/metabolism , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Hydrogen Exchanger 1/genetics , Stress, Mechanical , Sulfones/pharmacology
3.
Eur J Pharmacol ; 791: 718-725, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27742593

ABSTRACT

Caveolae, lipid enriched invaginations of the plasma membrane, are epicentres of cellular signal transduction. The structural proteins of caveolae, caveolins, regulate effector pathways in anaesthetic-induced cardioprotection, including the RISK pathway. Helium (He) postconditioning (HePoc) is known to mimic anaesthetic conditioning and to prevent damage from myocardial infarction. We hypothesize that HePoc regulates caveolin-1 and caveolin-3 (Cav-1 and Cav-3) expression in the rat heart and activates the RISK pathway. Male Wistar rats (n=8, each group) were subjected to 25min of cardiac ischaemia followed by reperfusion (I/R) for 5, 15 or 30min (I/R 5/15/30). The HePoc groups underwent I/R with 70% helium ventilation during reperfusion (IR+He 5/15/30min). Sham animals received surgical treatment without I/R. After each protocol blood and hearts were retrieved. Tissue was obtained from the area-at-risk (AAR) and non-area-at-risk (NAAR) and processed for western blot analyses and reverse-transcription-real-time-polymerase-chain-reaction (RT-qPCR). Protein analyses revealed increased amounts of Cav-1 and Cav-3 in the membrane of I/R+He15 (AAR: Cav-1, P<0.05; Cav-3, P<0.05; both vs. I/R15). In serum, Cav-3 was found to be elevated in I/R+He15 (P<0.05 vs. I/R15). RT-qPCR showed increased expression of Cav-1 in IR+He15 in AAR tissue (P<0.05 vs. I/R15). Phosphorylation of RISK pathway proteins pERK1/2 (AAR: P<0.05 vs. I/R15) and pAKT (AAR: P<0.05; NAAR P<0.05; both vs. I/R15) was elevated in the cytosolic fraction of I/R+He15. These results suggest that 15min of HePoc regulates Cav-1 and Cav-3 and activates RISK pathway kinases ERK1/2 and AKT. These processes might be crucially involved in HePoc mediated cardioprotection.


Subject(s)
Caveolins/metabolism , Gene Expression Regulation/drug effects , Helium/pharmacology , Ischemic Postconditioning , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Protein Kinases/metabolism , Animals , Caveolin 1/genetics , Caveolin 1/metabolism , Caveolin 3/genetics , Caveolin 3/metabolism , Caveolins/genetics , Male , Myocardial Reperfusion Injury/metabolism , Myocardium/pathology , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects
4.
J Transl Med ; 14(1): 294, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27737678

ABSTRACT

BACKGROUND: The noble gas helium induces pre- and postconditioning in animals and humans. Volatile anesthetics induce cardioprotection in humans undergoing coronary artery bypass graft (CABG) surgery. We hypothesized that helium induces pre- and postconditioning in CABG-patients, affecting signaling molecules protein kinase C-epsilon (PKC-ε), p38 mitogen activated protein kinase (p38 MAPK), extracellular signal-regulated kinase 1/2 (ERK-1/2) and heat shock protein 27 (HSP-27) within cardiac tissue, and reducing postoperative troponin levels. METHODS: After ethical approval and informed consent, 125 elective patients undergoing CABG surgery were randomised into this prospective, placebo controlled, investigator blinded, parallel arm single-centre study. Helium preconditioning (3 × 5 min of 70 % helium and 30 % oxygen) was applied before aortic cross clamping; postconditioning (15 min of helium) was applied before release of the aortic cross clamp. Signaling molecules were measured in right atrial appendix specimens. Troponin-T was measured at 4, 12, 24 and 48 h postoperatively. RESULTS: Baseline characteristics of all groups were similar. Helium preconditioning did not significantly alter the primary outcome (molecular levels of kinases PKC-ε and HSP-27, ratio of activated p38 MAPK or ERK ½). Postoperative troponin T was 11 arbitrary units [5, 31; area-under-the-curve (interquartile range)] for controls, and no statistically significant changes were observed after helium preconditioning [He-pre: 11 (6, 18)], helium postconditioning [He-post: 11 (8, 15)], helium pre- and postconditioning [He-PP: 14 (6, 20)] and after sevoflurane preconditioning [APC: 12 (8, 24), p = 0.13]. No adverse effects related to study treatment were observed in this study. CONCLUSIONS: No effect was observed of helium preconditioning, postconditioning or the combination thereof on activation of p38 MAPK, ERK 1/2 or levels of HSP27 and PKC-ε in the human heart. Helium pre- and postconditioning did not affect postoperative troponin release in patients undergoing CABG surgery. Clinical trial number Dutch trial register ( http://www.trialregister.nl/ ) number NTR1226.


Subject(s)
Coronary Artery Bypass , Helium/pharmacology , Ischemic Postconditioning , Protein Kinases/metabolism , Signal Transduction/drug effects , Aged , Cytosol/drug effects , Cytosol/enzymology , Demography , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Hemodynamics/drug effects , Humans , Ischemic Preconditioning, Myocardial , Male , Middle Aged , Molecular Chaperones , Phosphorylation/drug effects , Protein Kinase C-epsilon/metabolism , Troponin T/blood , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Exp Cell Res ; 337(1): 37-43, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26096659

ABSTRACT

Helium induces preconditioning in human endothelium protecting against postischemic endothelial dysfunction. Circulating endothelial microparticles are markers of endothelial dysfunction derived in response to injury. Another noble gas, xenon, protected human umbilical vein endothelial cells (HUVEC) against inflammatory stress in vitro. We hypothesised that helium protects the endothelium in vitro against inflammatory and oxidative stress. HUVEC were isolated from fresh umbilical cords and grown upon confluence. Cells were subjected to starving medium for 12h before the experiment and treated for either 3 × 5 min or 1 × 30 min with helium (5% CO2, 25% O2, 70% He) or control gas (5% CO2, 25% O2, 70% N2) in a specialised gas chamber. Subsequently, cells were stimulated with TNF-α (40 ng/ml for 24h or 10 ng/ml for 2h) or H2O2 (500 µM for 2h) or left untreated. Adhesion molecule expression was analysed using real-time quantitative polymerase chain reaction. Caspase-3 expression and viability of the cells was measured by flowcytometry. Microparticles were investigated by nanoparticle tracking analysis. Helium had no effect on adhesion molecule expression after TNF-α stimulation but in combination with oxidative stress decreased cell viability (68.9 ± 1.3% and 58 ± 1.9%) compared to control. Helium further increased TNF-α induced release of caspase-3 containing particles compared to TNF-α alone (6.4 × 10(6) ± 1.1 × 10(6) and 2.9 × 10(6) ± 0.7 × 10(6), respectively). Prolonged exposure of helium increased microparticle formation (2.4 × 10(9) ± 0.5 × 10(9)) compared to control (1.7 × 10(9) ± 0.2 × 10(9)). Summarized, helium increases inflammatory and oxidative stress-induced endothelial damage and is thus not biologically inert. A possible noxious effects on the cellular level causing alterations in microparticle formation both in number and content should be acknowledged.


Subject(s)
Cardiovascular Agents/pharmacology , Helium/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Oxidative Stress/drug effects , Apoptosis , Caspase 3/metabolism , Cell-Derived Microparticles/metabolism , Cells, Cultured , Drug Evaluation, Preclinical , E-Selectin/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intercellular Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
6.
PLoS One ; 8(5): e63497, 2013.
Article in English | MEDLINE | ID: mdl-23717435

ABSTRACT

Sepsis is characterized by a generalized inflammatory response and organ failure, associated with mitochondrial dysfunction. Hydrogen sulfide donor NaHS has anti-inflammatory properties, is able to reduce metabolism and can preserve mitochondrial morphology and function. Rats were challenged with live Streptococcus pneumonia or saline and infused with NaHS (36 µmol/kg/h) or vehicle. Lung and kidney injury markers were measured as well as mitochondrial function, viability and biogenesis. Infusion of NaHS reduced heart rate and body temperature, indicative of a hypo-metabolic state. NaHS infusion reduced sepsis-related lung and kidney injury, while host defense remained intact, as reflected by unchanged bacterial outgrowth. The reduction in organ injury was associated with a reversal of a fall in active oxidative phosphorylation with a concomitant decrease in ATP levels and ATP/ADP ratio. Preservation of mitochondrial respiration was associated with increased mitochondrial expression of α-tubulin and protein kinase C-ε, which acts as regulators of respiration. Mitochondrial damage was decreased by NaHS, as suggested by a reduction in mitochondrial DNA leakage in the lung. Also, NaHS treatment was associated with upregulation of peroxisome proliferator-activated receptor-γ coactivator 1α, with a subsequent increase in transcription of mitochondrial respiratory subunits. These findings indicate that NaHS reduces organ injury in pneumosepsis, possibly via preservation of oxidative phosphorylation and thereby ATP synthesis as well as by promoting mitochondrial biogenesis. Further studies on the involvement of mitochondria in sepsis are required.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Energy Metabolism , Lung Injury/prevention & control , Pneumonia, Pneumococcal/drug therapy , Sulfides/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Electron Transport Complex I/metabolism , Heart Rate/drug effects , Kidney/drug effects , Kidney/physiopathology , Liver/drug effects , Liver/metabolism , Lung Injury/metabolism , Lung Injury/microbiology , Mitochondria/metabolism , Mitochondrial Turnover/drug effects , Oxidation-Reduction , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/physiopathology , Protein Kinase C-epsilon/metabolism , Rats , Rats, Sprague-Dawley , Sepsis , Sulfides/therapeutic use , Tubulin/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...