Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cardiovasc Toxicol ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38916845

ABSTRACT

Oxidative stress results from the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in quantities exceeding the potential activity of the body's antioxidant system and is one of the risk factors for the development of vascular dysfunction in diabetes and exposure to ionizing radiation. Being the secondary products of normal aerobic metabolism in living organisms, ROS and RNS act as signaling molecules that play an important role in the regulation of vital organism functions. Meanwhile, in high concentrations, these compounds are toxic and disrupt various metabolic pathways. The various stress factors (hyperglycemia, gamma-irradiation, etc.) trigger free oxygen and nitrogen radicals accumulation in cells that are capable to damage almost all cellular components including ion channels and transporters such as Na+/K+-ATPase, BKCa, and TRP channels. Vascular dysfunctions are governed by interaction of ROS and RNS. For example, the reaction of ROS with NO produces peroxynitrite (ONOO-), which not only oxidizes DNA, cellular proteins, and lipids, but also disrupts important signaling pathways that regulate the cation channel functions in the vascular endothelium. Further increasing in ROS levels and formation of ONOO- leads to reduced NO bioavailability and causes endothelial dysfunction. Thus, imbalance of ROS and RNS and their affect on membrane ion channels plays an important role in the pathogenesis of vascular dysfunction associated with various disorders.

2.
Acta Physiol (Oxf) ; 237(3): e13922, 2023 03.
Article in English | MEDLINE | ID: mdl-36599422

ABSTRACT

AIM: Gold nanoparticles are widely used for biomedical applications, but the precise molecular mechanism of their interaction with cellular structures is still unclear. Assuming that intracellular calcium fluctuations associated with surface plasmon-induced calcium entry could modulate the activity of potassium channels, we studied the effect of 5 nm gold nanoparticles on calcium-dependent potassium channels and associated calcium signaling in freshly isolated rat pulmonary artery smooth muscle cells and cultured hippocampal neurons. METHODS: Outward potassium currents were recorded using patch-clamp techniques. Changes in intracellular calcium concentration were measured using the high affinity Ca2+ fluorescent indicator fluo-3 and laser confocal microscope. RESULTS: In pulmonary artery smooth muscle cells, plasmonic gold nanoparticles increased the amplitude of currents via large-conductance Ca2+ -activated potassium channels, which was potentiated by green laser irradiation near plasmon resonance wavelength (532 nm). Buffering of intracellular free calcium with ethylene glycol-bis-N,N,N',N'-tetraacetic acid (EGTA) abolished these effects. Furthermore, using confocal laser microscopy it was found that application of gold nanoparticles caused oscillations of intracellular calcium concentration that were decreasing in amplitude with time. In cultured hippocampal neurons gold nanoparticles inhibited the effect of EGTA slowing down the decline of the BKCa current while partially restoring the amplitude of the slow after hyperpolarizing currents. CONCLUSION: We conclude that fluctuations in intracellular calcium can modulate plasmonic gold nanoparticles-induced gating of BKCa channels in smooth muscle cells and neurons through an indirect mechanism, probably involving the interaction of plasmon resonance with calcium-permeable ion channels, which leads to a change in intracellular calcium level.


Subject(s)
Hippocampus , Metal Nanoparticles , Myocytes, Smooth Muscle , Potassium Channels , Animals , Rats , Calcium/metabolism , Egtazic Acid , Gold/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Metal Nanoparticles/therapeutic use , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Neurons/metabolism , Potassium Channels/metabolism , Pulmonary Artery/metabolism
3.
J Liposome Res ; 31(4): 399-408, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33319630

ABSTRACT

The aim of this study was to establish the mechanisms of action of a novel liposomal nitric oxide (NO) carrier on large-conductance Ca2+-activated channels (BKCa or Maxi-K) expressed in vascular smooth muscle cells (VSMCs) isolated from the rat main pulmonary artery (MPA). Experimental design comprised of both whole-cell and cell-attached single-channel recordings using the patch-clamp techniques. The liposomal form of NO, Lip(NO), increased whole-cell outward K+ currents in a dose dependent manner while shifting the activation curve negatively by about 50 mV with respect to unstimulated cells with the EC50 value of 0.55 ± 0.17 µM. At the single channel level, Lip(NO) increased the probability of the open state (Po) of Maxi-K channels from 0.0020 ± 0.0008 to 0.74 ± 0.02 with half-maximal activation occurring at 4.91 ± 0.01 µM, while sub-maximal activation was achieved at 10-5 M Lip(NO). Channel activation was mainly due to significant decrease in the mean closed dwell time (about 500-fold), rather than an increase in the mean open dwell time, which was comparatively modest (about twofold). There was also a slight decrease in the amplitude of the elementary Maxi-K currents (approximately 15%) accompanied by an increase in current noise, which might indicate some non-specific effects of Lip(NO) on the plasma membrane itself and/or on the phospholipids environment of the channels. In conclusion, the activating action of Lip(NO) on the Maxi-K channel is due to the destabilization of the closed conformation of the channel protein, which causes its more frequent openings and, accordingly, increases the probability of channel transition to its open state.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels , Nitric Oxide , Animals , Calcium/metabolism , Liposomes , Myocytes, Smooth Muscle/metabolism , Nitric Oxide/metabolism , Pulmonary Artery/metabolism , Rats
4.
Clin Exp Pharmacol Physiol ; 46(11): 1022-1029, 2019 11.
Article in English | MEDLINE | ID: mdl-31314914

ABSTRACT

Hypoxic pulmonary vasoconstriction (HPV) is the most important feature of intact lung circulation that matches local blood perfusion to ventilation. The main goal of this work was to study the effects of diabetes on the development of HPV in rats. The experimental design comprised diabetes mellitus induction by streptozotocin, video-morphometric measurements of the lumen area of intrapulmonary arteries (iPAs) using perfused lung tissue slices and patch-clamp techniques. It was shown that iPA lumen size was significantly reduced under physical and chemical hypoxia (7-10 mm Hg) in normal iPA, but, on the contrary, it clearly increased in diabetic lung slices. The amplitude of the outward K+ current in diabetic iPAs smooth muscle cells (SMCs) was two-fold greater than that seen in healthy cells. Chemical hypoxia led to significant decrease in the amplitude of the K+ outward current in healthy iPA SMCs while it was without effect in diabetic cells. The data obtained clearly indicate a significant dysregulation of vascular tone in pulmonary circulation under diabetes, ie diabetes damages the adaptive mechanism for regulating blood flow from poorly ventilated to better ventilated regions of the lung under hypoxia. This effect could be clinically important for patients with diabetes who have acute or chronic lung diseases associated with the lack of blood oxygenation.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/physiopathology , Hypoxia/complications , Pulmonary Artery/physiopathology , Vasoconstriction , Animals , Electrophysiological Phenomena , Male , Potassium/metabolism , Pulmonary Artery/metabolism , Rats , Rats, Wistar
5.
Nanomedicine ; 19: 1-11, 2019 07.
Article in English | MEDLINE | ID: mdl-30981819

ABSTRACT

Possessing unique physical and chemical properties, C60 fullerenes are arising as a potential nanotechnological tool that can strongly affect various biological processes. Recent molecular modeling studies have shown that C60 fullerenes can interact with ion channels, but there is lack of data about possible effects of C60 molecule on ion channels expressed in smooth muscle cells (SMC). Here we show both computationally and experimentally that water-soluble pristine C60 fullerene strongly inhibits the large conductance Ca2+-dependent K+ (BKCa), but not voltage-gated K+ (Kv) channels in pulmonary artery SMC. Both molecular docking simulations and analysis of single channel activity indicate that C60 fullerene blocks BKCa channel pore in its open state. In functional tests, C60 fullerene enhanced phenylephrine-induced contraction of pulmonary artery rings by about 25% and reduced endothelium-dependent acetylcholine-induced relaxation by up to 40%. These findings suggest a novel strategy for biomedical application of water-soluble pristine C60 fullerene in vascular dysfunction.


Subject(s)
Fullerenes/pharmacology , Kv Channel-Interacting Proteins/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Animals , Dynamic Light Scattering , Humans , Ion Channel Gating/drug effects , Male , Mice, Inbred BALB C , Molecular Docking Simulation , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Rats, Wistar
6.
J Liposome Res ; 29(1): 94-101, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29671361

ABSTRACT

The effects of quercetin-loaded liposomes (PCL-Q) and their constituents, that is, free quercetin (Q) and 'empty' phosphatidylcholine vesicles (PCL), on maxi-K channel activity were studied in single mouse ileal myocytes before and after H2O2-induced oxidative stress. Macroscopic Maxi-K channel currents were recorded using whole-cell patch clamp techniques, while single BKCa channel currents were recorded in the cell-attached configuration. Bath application of PCL-Q (100 µg/ml of lipid and 3 µg/ml of quercetin) increased single Maxi-K channel activity more than threefold, from 0.010 ± 0.003 to 0.034 ± 0.004 (n = 5; p < 0.05), whereas single-channel conductance increased non-significantly from 138 to 146 pS. In the presence of PCL-Q multiple simultaneous channel openings were observed, with up to eight active channels in the membrane patch. Surprisingly, 'empty' PCL (100 µg/ml) also produced some channel activation, although it was less potent compared to PCL-Q, that is, these increased NPo from 0.010 ± 0.003 to 0.019 ± 0.003 (n = 5; p < 0.05) and did not affect single-channel conductance (139 pS). Application of PCL-Q restored macroscopic Maxi-K currents suppressed by H2O2-induced oxidative stress in ileal smooth muscle cells. We conclude that PCL-Q can activate Maxi-K channels in ileal myocytes mainly by increasing channel open probability, as well as maintain Maxi-K-mediated whole-cell current under the conditions of oxidative stress. While fusion of the 'pure' liposomes with the plasma membrane may indirectly activate Maxi-K channels by altering channel's phospholipids environment, the additional potentiating action of quercetin may be due to its better bioavailability.


Subject(s)
Antioxidants/administration & dosage , Liposomes , Muscle, Smooth/metabolism , Oxidative Stress/drug effects , Quercetin/administration & dosage , Animals , Cell Line , Ileum/cytology , Ileum/metabolism , Large-Conductance Calcium-Activated Potassium Channels/drug effects , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Liposomes/chemistry , Male , Mice , Patch-Clamp Techniques
7.
Biochem Pharmacol ; 159: 121-139, 2019 01.
Article in English | MEDLINE | ID: mdl-30508525

ABSTRACT

Ionizing radiation (IR) leads to a variety of the cardiovascular diseases, including the arterial hypertension. A number of studies have demonstrated that blood vessels represent important target for IR, and the endothelium is one of the most vulnerable components of the vascular wall. IR causes an inhibition of nitric oxide (NO)-mediated endothelium-dependent vasodilatation and generation of reactive oxygen (ROS) and nitrogen (RNS) species trigger this process. Inhibition of NO-mediated vasodilatation could be due to endothelial NO synthase (eNOS) down-regulation, inactivation of endothelium-derived NO, and abnormalities in diffusion of NO from the endothelial cells (ECs) leading to a decrease in NO bioavailability. Beside this, IR suppresses endothelial large conductance Ca2+-activated K+ channels (BKCa) activity, which control NO synthesis. IR also leads to inhibition of the BKCa current in vascular smooth muscle cells (SMCs) which is mediated by protein kinase C (PKC). On the other hand, IR-evoked enhanced vascular contractility may result from PKC-mediated increase in SMCs myofilament Ca2+ sensitivity. Also, IR evokes vascular wall inflammation and atherosclerosis development. Vascular function damaged by IR can be effectively restored by quercetin-filled phosphatidylcholine liposomes and mesenchymal stem cells injection. Using RNA-interference technique targeted to different PKC isoforms can also be a perspective approach for pharmacological treatment of IR-induced vascular dysfunction.


Subject(s)
Atherosclerosis/etiology , Radiation, Ionizing , Vasodilation/radiation effects , Animals , Atherosclerosis/physiopathology , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , Endothelium, Vascular/physiopathology , Humans , Ion Channels/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/radiation effects , Nitric Oxide/metabolism , Prostaglandins/metabolism , Reactive Oxygen Species/metabolism , Vascular Diseases/drug therapy , Vascular Diseases/etiology , Vasodilation/drug effects , Vasodilation/physiology
8.
Vascul Pharmacol ; 72: 190-6, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26044181

ABSTRACT

Colloidal gold nanoparticles (AuNPs) of ~5nm core size and Zeta-potential of -35mV, having absorption maximum and plasmon resonance in the range of 510-570nm, were studied as a potential K(+)-channel opener in vascular smooth muscle (SM) cells. Experimental design of the study comprised SM contractile recordings. When externally applied to the organ bath, AuNPs (10(-6)-3×10(-4)M) led to decrease in amplitude of norepinephrine-induced contractions in a concentration-dependent and endothelium-independent manner in SM thoracic aorta, with mean value of pD2 (-log EC50) 4.2±0.03, Emax=55±4%. Being added to the bath solution in concentration of 10(-4)M, AuNPs significantly increased whole cell peak outward current at +70mV from 32±2pA/pF to 59±5pA/pF (n=14, P<0.05). External irradiation using a 5mW/532nm green laser, to facilitate plasmon resonance, led to an increment in the AuNPs-induced macroscopic outward potassium current (IK) from 59±5pA/pF to 74±1pA/pF (n=10, P<0.05). Paxilline (500nM), when added to the external bathing solution, significantly decreased AuNPs-induced increment of IK in SM cells. Single channel recordings provided a direct confirmation of BKCa activation by AuNPs at the single-channel level. Application of AuNPs to the bath potentiated BKCa activity with a delay of 1-2min, as was seen initially by more frequent channel openings followed by the progressive appearance of additional open levels corresponding to multiple openings of channels with identical single-channel amplitudes. Eventually, after 10-15min in the presence of AuNPs and especially when combined with the green laser illumination, there was a massive increase in channel activity with >10 channels evident. When irradiated by laser, AuNPs significantly increased the amplitude of maximal AuNPs-induced relaxation from 55±5% to 85±5% (n=10, P<0.05) while the sensitivity of SM to AuNPs was without changes. In summary, plasmonic AuNPs possess the ability to activate BKCa channel opening in vascular SM. Laser irradiation facilitates this effect due to local plasmon resonance that, in turn, further increases BKCa channel activity causing SM relaxation.


Subject(s)
Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Gold/pharmacology , Metal Nanoparticles/administration & dosage , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Potassium Channels/metabolism , Animals , Indoles/pharmacology , Male , Membrane Potentials/drug effects , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar
9.
Int J Cardiol ; 174(2): 230-42, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24794552

ABSTRACT

Diabetes mellitus (DM) is a complex syndrome which leads to multiple dysfunctions including vascular disorders. Hyperglycemia is considered to be a key factor responsible for the development of diabetic vascular complications and can mediate their adverse effects through multiple pathways. One of those mechanisms is the activation of protein kinase C (PKC). This important regulatory enzyme is involved in a signal transduction of several vascular functions including vascular smooth muscle contractility. Many studies have shown that hyperglycemia in DM results in oxidative stress. Overproduction of reactive oxygen species (ROS) by different oxidases and the mitochondrial electron transport chain (ETC), advanced glycation end products, polyol pathway flux, and hyperglicemia-induced rising in diacylglycerol (DAG) contribute to the activation of PKC. Activation of endothelial PKC in DM leads to endothelium-dependent vasodilator dysfunction. The main manifestations of this are inhibition of vasodilatation mediated by nitric oxide (NO), endothelium-derived hyperpolarizing factor (EDHF) and prostacyclin, and activation of vasoconstriction mediated by endothelin-1 (ET-1), prostaglandin E2 (PGE2) and thromboxane A2 (TXA2). Activated PKC in DM also increases vascular endothelial growth factor (VEGF) expression and activates NADPH oxidases leading to raised ROS production. On the other hand, PKC in DM is involved in enhancement of vascular contractility in an endothelium-independent manner by inactivation of K(+) channels and Ca(2+) sensitization of myofilaments in vascular smooth muscle cells. This shows that PKC is a potential therapeutic target for treating vascular diabetic complications.


Subject(s)
Diabetes Mellitus/enzymology , Diabetes Mellitus/physiopathology , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiopathology , Protein Kinase C/physiology , Animals , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Humans , Muscle Contraction
10.
J Basic Clin Physiol Pharmacol ; : 1-9, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24468620

ABSTRACT

Abstract Background: Endothelium and K+ channel functionality in smooth muscle cells (SMCs) regulates vascular function and is exposed to damage in diabetes. The regulatory enzyme protein kinase C (PKC) is known to play a key role in vascular tone regulation in health and disease. In this study, we evaluated the effect of PKC-δ gene silencing using small interfering RNAs (siRNAs) on endothelial dysfunction and acquired potassium channelopathy in vascular SMCs in diabetes. Methods: The experimental design comprised diabetes induction by streptozotocin (65 mg/kg) in rats, RNA interference, isolated aortic ring contractile recordings, whole-cell patch-clamp technique, measurements of reactive oxygen species (ROS), and real-time polymerase chain reaction technique. Animals were killed by cervical dislocation following ketamine (45 mg/kg, i.p.) and xylazine (10 mg/kg, i.p.) anesthesia administration on the third month of diabetes and on the seventh day after intravenous injection of siRNAs. Results: The aortas of diabetic rats demonstrated depressed endothelium-dependent relaxation and integral SMCs outward K+ currents as compared with those of controls. On the seventh day, PKC-δ gene silencing effectively restored K+ currents and increased the amplitude of vascular relaxation up to control levels. An increased level of PKC-δ mRNA in diabetic aortas appeared to be reduced after targeted PKC-δ gene silencing. Similarly, the level of ROS production that was increased in diabetes came back to control values after siRNAs administration. Conclusions: The silencing of PKC-δ gene expression using siRNAs led to restoration of vasodilator potential in rats with diabetes mellitus. It is likely that the siRNA technique can be a good therapeutic tool to normalize vascular function in diabetes.

11.
Eur J Pharmacol ; 718(1-3): 401-7, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23973649

ABSTRACT

Potassium conductance in vascular smooth muscle (VSM) is known to be altered in arterial hypertension. High level of protein kinase C (PKC) activity is a common feature for hypertension of different genesis. The main goal of this study was to investigate the efficacy of the RNA interference (RNAi) technique targeting PKC delta-isoform gene as a possible pharmacological tool to restore vasodilator potential in spontaneously hypertensive rats (SHR). Experimental design of the study comprised RNAi and patch-clamp techniques, RT-PCR analysis and standard acetylcholine test. Total outward currents and acetylcholine-induced endothelium-dependent relaxant responses were blunted in SHR. BKCa alpha subunit mRNA expression in SHR was unchanged whereas KV and KATP mRNA expression appeared significantly increased. PKC inhibitor, chelerythrine (100 nM), restored potassium channels activity in SHR. PKC-delta-isoform protein expression and PKC-delta-isoform mRNA expression are 2.5-4 fold increased in VSM from SHR. PKC gene silencing with the short hairpin RNAs (shRNAs)-plasmid delivery system administered intravenously led to an increment in maximal amplitude of acetylcholine-relaxation, restored outward K(+) currents and PKC-delta-isoform mRNA and protein expression. Arterial blood pressure in SHR was normalized following shRNAs administration. We conclude that BKCa channels are likely to be the most PKC-dependent member of K(+) channels family responsible for vascular hypercontractility in SHR while Kv and KATP channels may constitute a reserve mechanism for the maintenance of vasodilator potential under BKCa channelopathy. It is likely that RNAi technique is a good therapeutic approach to inactivate PKC gene and to normalize vascular functions and high arterial blood pressure in SHR.


Subject(s)
Genetic Techniques , Protein Kinase C-delta/deficiency , Protein Kinase C-delta/genetics , RNA Interference , RNA, Small Interfering/genetics , Vasoconstriction/genetics , Acetylcholine/pharmacology , Animals , In Vitro Techniques , Male , Rats , Rats, Inbred SHR , Vasoconstriction/drug effects
12.
Cardiovasc Res ; 99(3): 404-11, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23708740

ABSTRACT

AIMS: To determine the role of gap junctions (GJs) in hypoxic pulmonary vasoconstriction (HPV). METHODS AND RESULTS: Studies were performed in rat isolated intrapulmonary arteries (IPAs) mounted on a myograph and in anaesthetized rats. Hypoxia induced a biphasic HPV response in IPAs preconstricted with prostaglandin F2α (PGF2α, 3 µM) or 20 mM K⁺. The GJ inhibitors 18ß-glycyrrhetinic acid (18ß-GA, 30 µM), heptanol (3.5 mM), or 2-aminoethoxydiphenyl borate (2-APB) (75 µM) had little effect on the transient Phase 1 of HPV, but abolished the sustained Phase 2 which is associated with Ca²âº sensitization. The voltage-dependent Ca²âº channel blocker diltiazem (10 µM) had no effect on HPV, and did not alter the inhibitory action of 18ß-GA. Sustained HPV is enhanced by high glucose (15 mM) via potentiation of Ca²âº sensitization, in the presence of high glucose 18ß-GA still abolished sustained HPV. Simultaneous measurement of tension and intracellular Ca²âº using Fura PE-3 demonstrated that whilst 18ß-GA abolished tension development during sustained HPV, it did not affect the elevation of intracellular Ca²âº. Consistent with this, 18ß-GA abolished hypoxia-induced phosphorylation of the Rho kinase target MYPT-1. In anaesthetized rats hypoxia caused a biphasic increase in systolic right ventricular pressure. Treatment with oral 18ß-GA (25 mg/kg) abolished the sustained component of the hypoxic pressor response. CONCLUSION: These results imply that GJs are critically involved in the signalling pathways leading to Rho kinase-dependent Ca²âº sensitization during sustained HPV, but not elevation of intracellular Ca²âº, and may explain the dependence of the former on an intact endothelium.


Subject(s)
Calcium/metabolism , Gap Junctions/metabolism , Hypoxia/physiopathology , Pulmonary Artery/physiopathology , Vasoconstriction/physiology , Animals , Blood Pressure/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Diltiazem/pharmacology , Gap Junctions/drug effects , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , Hypoxia/metabolism , Male , Phosphorylation , Protein Phosphatase 1/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Vasoconstriction/drug effects , rho-Associated Kinases/metabolism
13.
Vascul Pharmacol ; 57(2-4): 119-23, 2012.
Article in English | MEDLINE | ID: mdl-22706072

ABSTRACT

The goal of this study was to clarify the mechanisms of hypoxic pulmonary vasoconstriction (HPV) reversal following selective glycolysis blockade and to assess possible contribution of endothelial electrogenesis to this phenomenon as a trigger mechanism. We compared smooth muscle (SM) contractility and endothelial cell (EC) membrane potential (MP) during acute hypoxia before and after glycolysis blockade. MPs were recorded from the endothelium of guinea pig pulmonary artery (GPPA) and thoracic aorta (GPTA) using the patch-clamp technique. Acute hypoxia caused hyperpolarization in GPTA EC, while EC from GPPA were depolarized. Also, acute hypoxia elicited constriction in isolated GPPA and dilatation in GPTA. Selective glycolysis inhibition always reversed both electrical and contractile responses in GPPA to hypoxia, but in GPTA this only occurred in 30% of experiments. It is likely that an unknown glycolysis-driven mechanism in EC mediates vascular tone regulation under hypoxia and underlies the paradoxical difference in the response of pulmonary and systemic arterial SM to hypoxia. Our data suggest that HPV development in GPPA might, at least partially, be driven by EC depolarization spreading to the underlying SM cells.


Subject(s)
Aorta, Thoracic/metabolism , Glycolysis/physiology , Pulmonary Artery/metabolism , Vasoconstriction/physiology , Animals , Cell Hypoxia , Endothelial Cells/metabolism , Guinea Pigs , Membrane Potentials/physiology , Muscle Contraction , Muscle, Smooth, Vascular/metabolism , Patch-Clamp Techniques , Pulmonary Artery/cytology
14.
Vascul Pharmacol ; 56(3-4): 142-9, 2012.
Article in English | MEDLINE | ID: mdl-22226998

ABSTRACT

It is likely that large-conductance Ca²âº-activated K⁺ (BK(Ca)) channels channelopathy tightly involved in vascular malfunctions and arterial hypertension development. In the present study, we compared the results of siRNAs-induced α-BK(Ca) gene silencing and vascular abnormalities produced by whole-body ionized irradiation in rats. The experimental design comprised RT-PCR and patch clamp technique, thoracic aorta smooth muscle (SM) contractile recordings and arterial blood pressure (BP) measurements on the 30th day after whole body irradiation (6Gy) and following siRNAs KCNMA1 gene silencing in vivo. The expression profile of BK(Ca) mRNA transcripts in SM was significantly decreased in siRNAs-treated rats in a manner similar to irradiated SM. In contrast, the mRNA levels of K(v) and K(ATP) were significantly increased while L-type calcium channels mRNA transcripts demonstrated tendency to increment. The SMCs obtained from irradiated animals and after KCNMA1 gene silencing showed a significant decrease in total K⁺ current density amplitude. Paxilline (500 nM)-sensitive components of outward current were significantly decreased in both irradiated and gene silencing SMCs. KCNMA1 gene silencing increased SM sensitivity to norepinephrine while Ach-induced relaxation had decreased. The silencing of KCNMA1 had no significant effect on BP while radiation produced sustained arterial hypertension. Therefore, radiation alters the form and function of the BK(Ca) channel and this type of channelopathy may contribute to related vascular abnormalities. Nevertheless, it is unlikely that BK(Ca) can operate as a crucial factor for radiation-induced arterial hypertension.


Subject(s)
Aorta, Thoracic/metabolism , Hypertension/etiology , Hypertension/metabolism , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Muscle, Smooth, Vascular/metabolism , Radiation Injuries/physiopathology , Vasoconstriction , Animals , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Aorta, Thoracic/radiation effects , Blood Pressure/radiation effects , Cells, Cultured , Gamma Rays/adverse effects , Gene Expression Regulation/radiation effects , Gene Silencing , Hypertension/physiopathology , In Vitro Techniques , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channels/genetics , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Muscle, Smooth, Vascular/radiation effects , Norepinephrine/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels/chemistry , Potassium Channels/genetics , Potassium Channels/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Vasoconstriction/drug effects
15.
Int J Radiat Biol ; 86(4): 291-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20353339

ABSTRACT

PURPOSE: The aim of this study was to estimate the effects of non-fatal whole-body gamma-irradiation on outward potassium plasma membrane conductivity in rat vascular smooth muscle cells (VSMC), and to identify underlying mechanisms. MATERIALS AND METHODS: Rats were exposed to a 6 Gy dose irradiation from a cobalt(60) source. Whole-cell potassium current was measured in freshly isolated rat aorta smooth muscle cells using standard patch-clamp technique. RESULTS: We have determined that whole-body ionising irradiation significantly inhibits whole-cell outward K(+) current in rat aortic VSMC obtained from irradiated rats 9 and 30 days after irradiation, and this inhibition appears to be increased throughout post-irradiation period. Using selective inhibitors of small conductance Ca(2+)-activated K(+) channels (SK(Ca)), apamin (1 microM), intermediate conductance Ca(2+)-activated K(+) channels (IK(Ca,)), charybdotoxin (1 microM) and a large conductance Ca(2+)-activated K(+) channels (BK(Ca)), paxilline (500 nM), we established that the main component of whole-cell outward K(+) current in rat aortic VSMC is due to BK(Ca). It is clear that on the 9th day after irradiation paxilline had only a small effect on whole-cell outward K(+) current in VSMC, and was without effect on the 30th day post-irradiation, suggesting complete suppression of the BK(Ca) current. The PKC inhibitor, chelerythrine (100 nM), effectively reversed the suppression of whole-cell outward K(+) current induced by ionising irradiation in the post-irradiation period of 9 and 30 days. CONCLUSIONS: The results suggest that irradiation-evoked inhibition of the BK(Ca) current in aortic VSMC is mediated by PKC. Taken together, our data indicate that one of the mechanisms leading to elevation of vascular tone and related arterial hypertension development under ionising irradiation impact is a PKC-mediated inhibition of BK(Ca) channels in VSMC.


Subject(s)
Aorta, Thoracic/cytology , Gamma Rays , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Myocytes, Smooth Muscle/radiation effects , Protein Kinase C/metabolism , Animals , Electric Conductivity , Electrophysiology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism , Patch-Clamp Techniques , Rats , Time Factors , Whole-Body Irradiation
16.
J Physiol Sci ; 60(2): 161-72, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20052569

ABSTRACT

The effect of intravenous administration of human mesenchymal stromal stem cells (hMSC) has been evaluated by means of large-conductance calcium-dependent potassium channel (BK(Ca)) activity measurements in thoracic aorta smooth muscle cells (SMC) obtained from non-fatal whole-body irradiated rats, using the patch clamp technique in whole-cell modification, and the standard acetylcholine (ACh) test to evaluate functional endothelium integrity using SM contractile recordings. Myofilament calcium sensitivity was estimated using simultaneous contractile recordings versus [Ca(2+)](i). Arterial blood was measured in intact and irradiated rats before and after hMSC administration. Stimulation of isolated SMC from the control group of animals with depolarizing voltage steps showed that outward K(+) currents sensitive to the BK(Ca) inhibitor paxilline were expressed. Outward currents in SMC obtained from irradiated animals were significantly reduced on the 30th day of post-irradiation. Irradiation led to a significant elevation in arterial blood pressure and reduced ACh-induced relaxation responses in irradiated rats as compared with the control group. Simultaneous measurements of contractile force and [Ca(2+)](i) showed that myofilament Ca(2+) sensitivity had increased following irradiation. Intravenously injected hMSC effectively restored BK(Ca) current and the amplitude of ACh-induced endothelium-dependent vasodilatation in vascular tissues obtained from post-irradiated rats. SMC obtained from irradiated rats treated with hMSC demonstrated a significantly increased paxilline-sensitive component of outward potassium currents, indicating that BK(Ca) activity had been restored. hMSC administration normalized increased blood pressure and myofilament Ca(2+) sensitivity in irradiated animals. When administered to healthy rats, hMSC were without effects on either of these. This study does not provide any immunohistochemical proof of hMSC engraftment in the host rats. PCR analysis showed that hMSCs were negative for hematopoietic cell markers and positive for hMSC markers. There were no clinical signs of graft-versus-host disease throughout the experimental period of 30 days. The data obtained suggest that hMSC demonstrate a clearly expressed ability to normalize vascular function damaged following irradiation, i.e. to reduce an elevated arterial blood pressure and myofilament Ca(2+) sensitivity, and to repair BK(Ca) function and endothelium-dependent relaxation in vascular tissues obtained from irradiated animals. Thus, hMSC seem to be worthwhile therapeutic approach in cases of ionizing irradiation accident or radiation beam therapy.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Radiation Injuries/physiopathology , Radiation Injuries/surgery , Vascular Diseases/physiopathology , Vascular Diseases/surgery , Whole-Body Irradiation/adverse effects , Adult , Animals , Cells, Cultured , Humans , Male , Middle Aged , Radiation Injuries/etiology , Rats , Rats, Wistar , Treatment Outcome , Vascular Diseases/etiology
17.
Sheng Li Xue Bao ; 61(3): 201-10, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19536431

ABSTRACT

The goal of the present study was to investigate the effects of quercetin-filled phosphatidylcholine liposomes (PCL-Q) on the currents carried by large conductance Ca(2+)-dependent K(+) channels (BK(Ca)) in rat thoracic aorta following non-fatal whole-body ionizing irradiation. Using patch-clamp technique, it is found that the outward K(+) currents of isolated smooth muscle cells (SMCs) stimulated by depolarizing voltage steps were sensitive to BK(Ca) inhibitor, paxilline, and this kind of outward K(+) currents in SMCs from irradiated animals demonstrated a significant decrease in amplitude. Radiation-induced BK(Ca) suppression was evident 9 days post-irradiation and progressively increased over 30 days of experimental period. Thus, the vasorelaxing force of these SMCs may be diminished following irradiation. PCL-Q effectively restored BK(Ca) function in post-irradiated SMCs. It is noteworthy that the constituents of PCL-Q, i.e., free quercetin (Q) and "empty" liposomes (PCL), being taken separately, showed a decreased ability to recover BK(Ca) function as compared with combined composition. These results suggest that PCL-Q is able to regain normal function of BK(Ca) following irradiation. The protective effects of PCL-Q can be explained by its antioxidant and membrane repairing properties as well as its ability to inhibit protein kinase C activity. Thus, the lipid encapsulation of flavonoid, PCL-Q, appears to be a potential medication in the case of ionizing irradiation accident, and for the patients with neoplasm who have to receive external radiotherapy as well.


Subject(s)
Aorta, Thoracic/radiation effects , Large-Conductance Calcium-Activated Potassium Channels/physiology , Liposomes/chemistry , Phosphatidylcholines/chemistry , Quercetin/pharmacology , Animals , Aorta, Thoracic/drug effects , Myocytes, Smooth Muscle/physiology , Patch-Clamp Techniques , Radiation, Ionizing , Rats
18.
Life Sci ; 84(5-6): 164-71, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19070626

ABSTRACT

AIMS: The goal of this study was to evaluate the influence of gamma-irradiation on Ca(2+)-activated K(+) channel (BK(Ca)) function and expression in rat thoracic aorta. MAIN METHODS: Aortic cells or tissues were studied by the measurement of force versus [Ca(2+)](i), patch-clamp technique, and RT-PCR. KEY FINDINGS: Stimulation of smooth muscle cells with depolarizing voltage steps showed expression of outward K(+) currents. Paxilline, an inhibitor of BK(Ca) channels, decreased outward K(+) current density. Outward currents in smooth muscle cells obtained from irradiated animals 9 and 30 days following radiation exposure demonstrated a significant decrease in K(+) current density. Paxilline decreased K(+) current in cells obtained 9 days, but was without effect 30 days after irradiation suggesting the absence of BK(Ca) channels. Aortic tissue from irradiated animals showed progressively enhanced contractile responses to phenylephrine in the post-irradiation period of 9 and 30 days. The concomitant Ca(2+) transients were significantly smaller, as compared to tissues from control animals, 9 days following irradiation but were increased above control levels 30 days following irradiation. Irradiation produced a decrease in BK(Ca) alpha- and beta(1)-subunit mRNA levels in aortic smooth muscle cells suggesting that the vasorelaxant effect of these channels may be diminished. SIGNIFICANCE: These results suggest that the enhanced contractility of vascular tissue from animals exposed to radiation may result from an increase in myofilament Ca(2+) sensitivity in the early post-irradiation period and a decrease in BK(Ca) channel expression in the late post-irradiation period.


Subject(s)
Aorta, Thoracic/radiation effects , Gamma Rays/adverse effects , Ion Channel Gating/radiation effects , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Muscle, Smooth, Vascular/radiation effects , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/metabolism , Cells, Cultured , Large-Conductance Calcium-Activated Potassium Channels/biosynthesis , Male , Muscle Contraction/radiation effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Patch-Clamp Techniques , Paxillin/pharmacology , RNA/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Whole-Body Irradiation
19.
Int J Radiat Biol ; 83(3): 161-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17378524

ABSTRACT

PURPOSE: The goal of this study was to evaluate the influence of ionizing irradiation on large conductance Ca2+-dependent potassium (BKCa) channels in rat coronary endothelial cells. MATERIALS AND METHODS: Rats were exposed to a 6 Gy dose from a cobalt60 source. Experimental design of this study comprised recording of contractile force using isolated rat aortic rings and whole-cell patch clamp techniques to study whole-cell potassium currents in isolated rat coronary artery endothelial cells. RESULTS: It has been shown that outward potassium currents in endothelial cells 9 days after irradiation appear to be suppressed or even totally abolished. The reversal potential for these currents in irradiated cells was shifted to more positive values. Paxilline (500 nM), an inhibitor of BKCa channels, had no or only a negligible effect on irradiated cells. The experiments using isolated aortic rings demonstrated that both paxilline and irradiation significantly shifted the acetylcholine dependent concentration-relaxation response curve to the right. Irradiated tissues were insensitive to paxilline. CONCLUSION: The results suggest that non-fatal, whole-body gamma-irradiation suppresses large conductance, calcium-activated potassium channels, which control the driving force for Ca2+ entry and therefore Ca2+ dependent nitric oxide (NO) synthesis in endothelial cells. This may contribute, in part, to radiation-induced endothelium dysfunction and an increase in arterial blood pressure.


Subject(s)
Coronary Vessels/radiation effects , Endothelial Cells/radiation effects , Potassium Channels, Calcium-Activated/physiology , Acetylcholine/pharmacology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Aorta, Thoracic/radiation effects , Coronary Vessels/cytology , Coronary Vessels/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/physiology , Gamma Rays , Male , Membrane Potentials/drug effects , Membrane Potentials/radiation effects , Patch-Clamp Techniques , Paxillin/pharmacology , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Rats , Rats, Wistar , Vasodilation/drug effects , Vasodilation/radiation effects , Vasodilator Agents/pharmacology , Whole-Body Irradiation
20.
Am J Physiol Regul Integr Comp Physiol ; 289(3): R755-62, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15890787

ABSTRACT

Radiation exposure increases vascular responsiveness, and this change involves endothelial damage, as well as direct effects on vascular smooth muscle. In this study, we tested the hypothesis that myofilament Ca(2+) sensitivity in vascular smooth muscle is increased from single whole body gamma irradiation (6 Gy). We measured contractile responses from intact and permeabilized rat thoracic aortic rings combined with cytosolic Ca(2+) ([Ca(2+)](i)) measurements. The sensitivity to KCl and phenylephrine increased significantly in tissues from animals on the 9th and 30th days postirradiation compared with control. Irradiation also significantly increased Ca(2+) sensitivity in beta-escin permeabilized smooth muscle on the 9th and 30th days postirradiation. Inhibitors of protein kinase C, chelerythrine, and staurosporine, had no effect on the pCa-tension curves in control permeabilized tissues but significantly decreased Ca(2+) sensitivity in permeabilized tissues on the 9th and 30th days postirradiation. Phorbol dibutyrate (PDBu, 10(-7) M) increased Ca(2+) sensitivity in control skinned smooth muscle but was without effect in irradiated vascular rings. Simultaneous measurement of contractile force and [Ca(2+)](i) showed that myofilament Ca(2+) sensitivity defined as the ratio of force change to [Ca(2+)](i) significantly increased following gamma-irradiation. PDBu (10(-6) M) stimulation of intact aorta produced a sustained contraction, while the increase in [Ca(2+)](i) was transient. In irradiated tissues, PDBu-induced contractions were greater than those seen in control tissues but there was no elevation in [Ca(2+)](i). Taken together, these data strongly support the hypothesis that irradiation increases the sensitivity of vascular smooth muscle myofilaments to Ca(2+) and this effect is dependent on activation of protein kinase C.


Subject(s)
Actin Cytoskeleton/physiology , Actin Cytoskeleton/radiation effects , Calcium/physiology , Muscle, Smooth, Vascular/physiology , Muscle, Smooth, Vascular/radiation effects , Protein Kinase C/physiology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Aorta, Thoracic/radiation effects , Calcium/metabolism , Capillary Permeability , In Vitro Techniques , Intracellular Membranes/metabolism , Osmolar Concentration , Potassium Chloride/pharmacology , Rats , Rats, Inbred WKY , Vasoconstriction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...