Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 108
1.
Eur J Clin Invest ; 52(4): e13690, 2022 Apr.
Article En | MEDLINE | ID: mdl-34662431

BACKGROUND: Ceramide is involved in regulating metabolism and energy expenditure, and its abnormal myocardial accumulation may contribute to heart injury or lipotoxic cardiomyopathy. Whether ceramide can modulate the electrophysiology of pulmonary veins (PVs) remains unknown. MATERIALS AND METHODS: We used conventional microelectrodes to measure the electrical activity of isolated rabbit PV tissue preparations before and after treatment with various concentrations of ceramide with or without H2 O2 (2 mM), MitoQ, wortmannin or 740 YP. A whole-cell patch clamp and fluorescence imaging were used to record the ionic currents, calcium (Ca2+ ) transients, and intracellular reactive oxygen species (ROS) and sodium (Na+ ) in isolated single PV cardiomyocytes before and after ceramide (1 µM) treatment. RESULTS: Ceramide (0.1, 0.3, 1 and 3 µM) reduced the beating rate of PV tissues. Furthermore, ceramide (1 µM) suppressed the 2 mM H2 O2 -induced faster PV beating rate, triggered activities and burst firings, which were further reduced by MitoQ. In the presence of wortmannin, ceramide did not change the PV beating rate. The H2 O2 -induced faster PV beating rate could be counteracted by MitoQ or wortmannin with no additive effect from the ceramide. Ceramide inhibited pPI3K. Ceramide reduced Ca2+ transients, sarcoplasmic reticulum Ca2+ contents, L-type Ca2+ currents, Na+ currents, late Na+ currents, Na+ -hydrogen exchange currents, and intracellular ROS and Na+ in PV cardiomyocytes, but did not change Na+ -Ca2+ exchange currents. CONCLUSION: C2 ceramide may exert the distinctive electrophysiological effect of modulating PV activities, which may be affected by PI3K pathway-mediated oxidative stress, and might play a role in the pathogenesis of PV arrhythmogenesis.


Ceramides/physiology , Myocytes, Cardiac/metabolism , Oxidative Stress/physiology , Pulmonary Veins/cytology , Animals , Electrophysiological Phenomena , Male , Rabbits
2.
J Mol Cell Cardiol ; 161: 86-97, 2021 12.
Article En | MEDLINE | ID: mdl-34375616

Delayed rectifier K+ current (IKs) is a key contributor to repolarization of action potentials. This study investigated the mechanisms underlying the adrenoceptor-induced potentiation of IKs in pulmonary vein cardiomyocytes (PVC). PVC were isolated from guinea pig pulmonary vein. The action potentials and IKs current were recorded using perforated and conventional whole-cell patch-clamp techniques. The expression of IKs was examined using immunocytochemistry and Western blotting. KCNQ1, a IKs pore-forming protein was detected as a signal band approximately 100 kDa in size, and its immunofluorescence signal was found to be mainly localized on the cell membrane. The IKs current in PVC was markedly enhanced by both ß1- and ß2-adrenoceptor stimulation with a negative voltage shift in the current activation, although the potentiation was more effectively induced by ß2-adrenoceptor stimulation than ß1-adrenoceptor stimulation. Both ß-adrenoceptor-mediated increases in IKs were attenuated by treatment with the adenylyl cyclase (AC) inhibitor or protein kinase A (PKA) inhibitor. Furthermore, the IKs current was increased by α1-adrenoceptor agonist but attenuated by the protein kinase C (PKC) inhibitor. PVC exhibited action potentials in normal Tyrode solution which was slightly reduced by HMR-1556 a selective IKs blocker. However, HMR-1556 markedly reduced the ß-adrenoceptor-potentiated firing rate. The stimulatory effects of ß- and α1-adrenoceptor on IKs in PVC are mediated via the PKA and PKC signal pathways. HMR-1556 effectively reduced the firing rate under ß-adrenoceptor activation, suggesting that the functional role of IKs might increase during sympathetic excitation under in vivo conditions.


Delayed Rectifier Potassium Channels/metabolism , Myocytes, Cardiac/metabolism , Pulmonary Veins/metabolism , Receptors, Adrenergic/metabolism , Action Potentials/drug effects , Adrenergic alpha-Agonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Guinea Pigs , Heart Atria/metabolism , Isoproterenol/pharmacology , KCNQ1 Potassium Channel/metabolism , Myocytes, Cardiac/drug effects , Norepinephrine/pharmacology , Patch-Clamp Techniques , Protein Kinase C/metabolism , Pulmonary Veins/cytology , Signal Transduction
3.
Eur J Clin Invest ; 51(9): e13585, 2021 Sep.
Article En | MEDLINE | ID: mdl-34002387

INTRODUCTION: Phosphodiesterase (PDE) isoform inhibitors have mechanical and electrical effects on the heart. Inhibition of PDE-1 enzymes is a novel strategy for treating heart failure. However, the electrophysiological effects of PDE-1 inhibition on the heart remain unclear. This study explored the effects of PDE-1 inhibition using ITI-214 on electrical activity in the pulmonary vein (PV), the most common trigger of atrial fibrillation, and investigated the underlying ionic mechanisms. METHODS: Conventional microelectrodes or whole-cell patch clamps were employed to study the effects of ITI-214 (0.1-10 µM) on PV electrical activity, mechanical responses and ionic currents in isolated rabbit PV tissue specimens and isolated single PV cardiomyocytes. RESULTS: ITI-214 at 1 µM and 10 µM (but not 0.1 µM) significantly reduced PV spontaneous beating rate (10 ± 2% and 10 ± 3%, respectively) and PV diastolic tension (11 ± 3% and 17 ± 3%, respectively). ITI-24 (1 µM) significantly reduced late sodium current (INa-Late ), L-type calcium current (ICa-L ) and the reverse mode of the sodium-calcium exchanger (NCX), but it did not affect peak sodium currents. CONCLUSIONS: ITI-214 reduces PV spontaneous activity and PV diastolic tension by reducing INa-Late , ICa-L and NCX current. Considering its therapeutic potential in heart failure, targeting PDE-1 inhibition may provide a novel strategy for managing atrial arrhythmogenesis.


Action Potentials/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Myocytes, Cardiac/drug effects , Phosphodiesterase Inhibitors/pharmacology , Pulmonary Veins/drug effects , Animals , Calcium/metabolism , Patch-Clamp Techniques , Pulmonary Veins/cytology , Rabbits
4.
PLoS Comput Biol ; 17(4): e1008930, 2021 04.
Article En | MEDLINE | ID: mdl-33878108

In this work, non-invasive high-spatial resolution three-dimensional (3D) X-ray micro-computed tomography (µCT) of healthy mouse lung vasculature is performed. Methodologies are presented for filtering, segmenting, and skeletonizing the collected 3D images. Novel methods for the removal of spurious branch artefacts from the skeletonized 3D image are introduced, and these novel methods involve a combination of distance transform gradients, diameter-length ratios, and the fast marching method (FMM). These new techniques of spurious branch removal result in the consistent removal of spurious branches without compromising the connectivity of the pulmonary circuit. Analysis of the filtered, skeletonized, and segmented 3D images is performed using a newly developed Vessel Network Extraction algorithm to fully characterize the morphology of the mouse pulmonary circuit. The removal of spurious branches from the skeletonized image results in an accurate representation of the pulmonary circuit with significantly less variability in vessel diameter and vessel length in each generation. The branching morphology of a full pulmonary circuit is characterized by the mean diameter per generation and number of vessels per generation. The methods presented in this paper lead to a significant improvement in the characterization of 3D vasculature imaging, allow for automatic separation of arteries and veins, and for the characterization of generations containing capillaries and intrapulmonary arteriovenous anastomoses (IPAVA).


Algorithms , Image Processing, Computer-Assisted/methods , Lung/blood supply , Tomography, X-Ray Computed/methods , Animals , Mice, Inbred C57BL , Pulmonary Artery/cytology , Pulmonary Veins/cytology
5.
J Physiol Sci ; 70(1): 6, 2020 Feb 11.
Article En | MEDLINE | ID: mdl-32046630

Pulmonary vein (PV) cardiomyocytes have the potential to generate spontaneous activity, in contrast to working myocytes of atria. Different electrophysiological properties underlie the potential automaticity of PV cardiomyocytes, one being the hyperpolarization-activated inward current (Ih), which facilitates the slow diastolic depolarization. In the present study, we examined pharmacological characteristics of the Ih of PV cardiomyocytes in rat, guinea pig and rabbit. The results showed that guinea pig and rat PV cardiomyocytes possessed sizeable amplitudes of the Ih, and the Ih of guinea pig was suppressed by Cs+, a blocker of the hyperpolarization-activated cation current. However, the Ih of rat was not suppressed by Cs+, but by Cd2+, a blocker of the Cl- current. The current density of the Ih of rabbit PV cardiomyocytes was significantly smaller than those of other species. This suggests that the ion channels that carry the Ih of PV cardiomyocytes differ among the animal species.


Action Potentials/physiology , Myocytes, Cardiac/physiology , Pulmonary Veins/cytology , Action Potentials/drug effects , Animals , Barium , Cadmium , Cesium , Guinea Pigs , Myocytes, Cardiac/drug effects , Rabbits , Rats , Species Specificity
6.
Am J Physiol Cell Physiol ; 318(3): C555-C569, 2020 03 01.
Article En | MEDLINE | ID: mdl-31940248

Unlike the pulmonary artery (PA), the pathophysiological changes of the pulmonary vein (PV) in the development of pulmonary hypertension (PH) remain largely unknown. In this study, we comprehensively investigated the structural and functional changes in the PV isolated from the chronic hypoxia (CH; 10% O2, 21 days)-induced PH rat model (CHPH). Results showed that CH caused an increase in right ventricular pressure but did not affect the mean pulmonary venous pressure and the left atrial pressure. Similar to the PA, vascular lumen stenosis and medial thickening were also observed in the intrapulmonary veins isolated from the CHPH rats. Notably, CH induced more severe loss in the endothelium of intrapulmonary veins than the arteries. Then, the contractile response to 5-HT and U46619 was significantly greater in the intrapulmonary small veins (ISPV) and arteries (ISPA) isolated from CHPH rats than those from normoxic rats but not in the extrapulmonary and intrapulmonary large veins. Treatment with nifedipine (Nif), SKF96365 (SKF), or ryanodine and caffeine either partially attenuated (Nif) or dramatically abolished (SKF or ryanodine and caffeine) 5-HT-induced maximal contraction in ISPV from both normoxic and CHPH rats. Because of the severe loss of endothelium in the PV of CHPH rats, the decrease in acetylcholine (ACh)-induced endothelium-dependent relaxation was significantly larger in ISPV than ISPA, whereas the sodium nitroprusside-induced endothelium-independent relaxation was not altered in both ISPA and ISPV. In conclusion, our results provide fundamental data to comprehensively define the PV system in CHPH rat model.


Disease Models, Animal , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Pulmonary Veins/cytology , Pulmonary Veins/physiology , Animals , Cells, Cultured , Chronic Disease , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/pathology , Hypoxia/pathology , Male , Organ Culture Techniques , Pulmonary Veins/drug effects , Rats , Rats, Sprague-Dawley , Vasoconstrictor Agents/toxicity , Vasodilator Agents/pharmacology
7.
J Biol Chem ; 294(44): 16049-16061, 2019 11 01.
Article En | MEDLINE | ID: mdl-31506297

Pulmonary veins (PVs) are the major origin of atrial fibrillation. Recently, we recorded hyperpolarization-activated Cl- current (ICl, h) in rat PV cardiomyocytes. Unlike the well-known chloride channel protein 2 (CLCN2) current, the activation curve of ICl, h was hyperpolarized as the Cl- ion concentration ([Cl-] i ) increased. This current could account for spontaneous activity in PV cardiomyocytes linked to atrial fibrillation. In this study, we aimed to identify the channel underlying ICl, h Using RT-PCR amplification specific for Clcn2 or its homologs, a chloride channel was cloned from rat PV and detected in rat PV cardiomyocytes using immunocytochemistry. The gene sequence and electrophysiological functions of the protein were identical to those previously reported for Clcn2, with protein activity observed as a hyperpolarization-activated current by the patch-clamp method. However, the [Cl-] i dependence of activation was entirely different from the observed ICl, h of PV cardiomyocytes; the activation curve of the Clcn2-transfected cells shifted toward positive potential with increased [Cl-] i , whereas the ICl, h of PV and left ventricular cardiomyocytes showed a leftward shift. Therefore, we used MS to explore the possibility of additional proteins interacting with CLCN2 and identified an individual 71-kDa protein, HSPA8, that was strongly expressed in rat PV cardiomyocytes. With co-expression of HSPA8 in HEK293 and PC12 cells, the CLCN2 current showed voltage-dependent activation and shifted to negative potential with increasing [Cl-] i Molecular docking simulations further support an interaction between CLCN2 and HSPA8. These findings suggest that CLCN2 in rat heart contains HSPA8 as a unique accessory protein.


Action Potentials , Chloride Channels/metabolism , HSC70 Heat-Shock Proteins/metabolism , Myocytes, Cardiac/metabolism , Pulmonary Veins/cytology , Animals , Binding Sites , CLC-2 Chloride Channels , Cells, Cultured , Chloride Channels/chemistry , HEK293 Cells , HSC70 Heat-Shock Proteins/chemistry , HSC70 Heat-Shock Proteins/genetics , Heart Ventricles/cytology , Humans , Male , Molecular Docking Simulation , Myocytes, Cardiac/physiology , PC12 Cells , Protein Binding , Pulmonary Veins/metabolism , Rats , Rats, Wistar
8.
J Pharmacol Sci ; 141(1): 9-16, 2019 Sep.
Article En | MEDLINE | ID: mdl-31521490

The role of the Na+ current in the automaticity of the pulmonary vein myocardium was examined in isolated guinea pig pulmonary vein cardiomyocytes and tissue preparations. Tetrodotoxin inhibited the automaticity of pulmonary vein tissue preparations by suppressing the diastolic depolarization of the action potential. ATX-II, which increased the density of persistent component of the Na+ current (late INa), induced a depolarization of the resting membrane potential followed by spontaneous firing of action potentials. GS-458967, which inhibited the late INa, suppressed the diastolic depolarization and the firing of action potentials. Pilsicainide, which inhibited only the transient component of Na+ current (peak INa), had no effect on the firing frequency. GS-458967 had no effect on the contractile force of the working myocardium. In conclusion, late INa is involved in the diastolic depolarization and automaticity of the pulmonary vein myocardium. Late INa inhibitors appear to be effective therapeutic agents for atrial fibrillation with minimum adverse effects on the working myocardium.


Myocytes, Cardiac/metabolism , Pulmonary Veins/cytology , Pyridines/pharmacology , Sodium/metabolism , Triazoles/pharmacology , Action Potentials/drug effects , Animals , Atrial Fibrillation/drug therapy , Cells, Cultured , Diastole/drug effects , Guinea Pigs , Lidocaine/analogs & derivatives , Lidocaine/pharmacology , Male , Membrane Potentials/drug effects , Pulmonary Veins/metabolism , Pyridines/therapeutic use , Tetrodotoxin/pharmacology , Triazoles/therapeutic use
9.
Int J Mol Sci ; 20(12)2019 Jun 14.
Article En | MEDLINE | ID: mdl-31207916

Cardiomyocytes and myocardial sleeves dissociated from pulmonary veins (PVs) potentially generate ectopic automaticity in response to noradrenaline (NA), and thereby trigger atrial fibrillation. We developed a mathematical model of rat PV cardiomyocytes (PVC) based on experimental data that incorporates the microscopic framework of the local control theory of Ca2+ release from the sarcoplasmic reticulum (SR), which can generate rhythmic Ca2+ release (limit cycle revealed by the bifurcation analysis) when total Ca2+ within the cell increased. Ca2+ overload in SR increased resting Ca2+ efflux through the type II inositol 1,4,5-trisphosphate (IP3) receptors (InsP3R) as well as ryanodine receptors (RyRs), which finally triggered massive Ca2+ release through activation of RyRs via local Ca2+ accumulation in the vicinity of RyRs. The new PVC model exhibited a resting potential of -68 mV. Under NA effects, repetitive Ca2+ release from SR triggered spontaneous action potentials (APs) by evoking transient depolarizations (TDs) through Na+/Ca2+ exchanger (APTDs). Marked and variable latencies initiating APTDs could be explained by the time courses of the α1- and ß1-adrenergic influence on the regulation of intracellular Ca2+ content and random occurrences of spontaneous TD activating the first APTD. Positive and negative feedback relations were clarified under APTD generation.


Action Potentials , Catecholamines/pharmacology , Models, Theoretical , Myocytes, Cardiac/metabolism , Pulmonary Veins/metabolism , Animals , Calcium Signaling , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Pulmonary Veins/cytology , Pulmonary Veins/drug effects , Pulmonary Veins/physiology , Rats , Receptors, Adrenergic/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sodium-Calcium Exchanger/metabolism
10.
Kardiologiia ; 58(6): 37-43, 2018 06.
Article Ru | MEDLINE | ID: mdl-30362435

Recently, the notion that in 60-80 % of cases the origin of the pulmonary veins (PV) is the place of origin of atrial fibrillation (AF) has become widespread. It has been shown that in this area, under the action of norepinephrine (HA), in the absence of stimulation, an intrinsic rhythm appears. Using two-channel microelectrode leads (from the mouth and distal part of the PV) in rats weighing 350-450 grams, it was found that: 1) in the distal part of PV there are cells with depolarized resting potential (RP) up to -50 mV, which under normal conditions are not excitable; 2) in 17 experiments out of 23, various blocks of excitation conduction along PV were revealed; 3) in 8 experiments out of 23, a reflected excitation wave - echo from PV - was recorded. Myocardium of PV is an extremely heterogeneous medium with a strong variance in the duration of the action potential and variable rate of conduction, which contributes to the occurrence of different types of conduction blocks and causes echoes and other rhythm disturbances.


Atrial Fibrillation/physiopathology , Pulmonary Veins/physiology , Action Potentials , Animals , Atrial Fibrillation/pathology , Electrophysiology , Muscle, Smooth, Vascular/physiology , Pulmonary Veins/cytology , Rats , Rats, Wistar
11.
J Physiol ; 596(13): 2491-2506, 2018 07.
Article En | MEDLINE | ID: mdl-29790164

KEY POINTS: We investigated the excitation-contraction coupling mechanisms in small pulmonary veins (SPVs) in rat precision-cut lung slices. We found that SPVs contract strongly and reversibly in response to extracellular ATP and other vasoconstrictors, including angiotensin-II and endothelin-1. ATP-induced vasoconstriction in SPVs was associated with the stimulation of purinergic P2Y2 receptors in vascular smooth muscle cell, activation of phospholipase C-ß and the generation of intracellular Ca2+ oscillations mediated by cyclic Ca2+ release events via the inositol 1,4,5-trisphosphate receptor. Active constriction of SPVs may play an important role in the development of pulmonary hypertension and pulmonary oedema. ABSTRACT: The small pulmonary veins (SPVs) may play a role in the development of pulmonary hypertension and pulmonary oedema via active changes in SPV diameter, mediated by vascular smooth muscle cell (VSMC) contraction. However, the excitation-contraction coupling mechanisms during vasoconstrictor stimulation remain poorly understood in these veins. We used rat precision-cut lung slices and phase-contrast and confocal microscopy to investigate dynamic changes in SPV cross-sectional luminal area and intracellular Ca2+ signalling in their VSMCs. We found that the SPV (∼150 µm in diameter) contract strongly in response to extracellular ATP and other vasoconstrictors, including angiotensin-II and endothelin-1. ATP-induced SPV contraction was fast, concentration-dependent, completely reversible upon ATP washout, and inhibited by purinergic receptor antagonists suramin and AR-C118925 but not by MRS2179. Immunofluorescence showed purinergic P2Y2 receptors expressed in SPV VSMCs. ATP-induced SPV contraction was inhibited by phospholipase Cß inhibitor U73122 and accompanied by intracellular Ca2+ oscillations in the VSMCs. These Ca2+ oscillations and SPV contraction were inhibited by the inositol 1,4,5-trisphosphate receptor inhibitor 2-APB but not by ryanodine. The results of the present study suggest that ATP-induced vasoconstriction in SPVs is associated with the activation of purinergic P2Y2 receptors in VSMCs and the generation of Ca2+ oscillations.


Calcium/physiology , Muscle Contraction , Myocytes, Smooth Muscle/physiology , Pulmonary Veins/physiology , Receptors, Purinergic P2Y2/metabolism , Vasoconstriction , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Cross-Sectional Studies , Excitation Contraction Coupling , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Myocytes, Smooth Muscle/cytology , Phospholipase C beta/metabolism , Pulmonary Veins/cytology , Rats
12.
Sci Rep ; 8(1): 3244, 2018 02 19.
Article En | MEDLINE | ID: mdl-29459735

Mechanisms underlying ectopic activity in the pulmonary vein (PV) which triggers paroxysmal atrial fibrillation are unknown. Although several studies have suggested that calcium signalling might be involved in these arrhythmias, little is known about calcium cycling in PV cardiomyocytes (CM). We found that individual PV CM showed a wide range of transverse tubular incidence and organization, going from their virtual absence, as described in atrial CM, to well transversally organised tubular systems, like in ventricular CM. These different types of CM were found in groups scattered throughout the tissue. The variability of the tubular system was associated with cell to cell heterogeneity of calcium channel (Cav1.2) localisation and, thereby, of Cav1.2-Ryanodine receptor coupling. This was responsible for multiple forms of PV CM calcium transient. Spontaneous calcium sparks and waves were not only more abundant in PV CM than in LA CM but also associated with a higher depolarising current. In conclusion, compared with either the atrium or the ventricle, PV myocardium presents marked structural and functional heterogeneity.


Calcium Signaling , Heart Atria/cytology , Myocytes, Cardiac/physiology , Pulmonary Veins/cytology , Animals , Calcium/metabolism , Calcium Channels, L-Type/analysis , Myocytes, Cardiac/enzymology , Pulmonary Veins/physiology , Rats
13.
Anat Histol Embryol ; 47(2): 145-152, 2018 Apr.
Article En | MEDLINE | ID: mdl-29314148

The myocardial sleeve of the porcine and equine pulmonary veins were histologically investigated and reconstructed three dimensionally. Moreover, the localization of neuron cell bodies at the veno-atrial junction and alongside the myocardial sleeve was light microscopically visualized to depict the organization of nerve, myocardial and fat tissue. Finally, the presence of telocytes inside the equine pulmonary veins was demonstrated by use of transmission electron microscopy. These structures are thought to play a role in the induction of atrial fibrillation, which is frequently seen in horses, while pigs are often used as a cardiovascular model in this context. This data fills in remaining gaps in the literature concerning the histological build-up of the pulmonary veins wall in pigs and horses. In-depth knowledge on the myocardial sleeve and its surrounding cell types are important to understand the possible outcome of an ablation therapy as an atrial fibrillation treatment. In pigs and horses, the layout of the pulmonary veins wall concerning these structures is comparable to humans. However, neuron cell bodies were recovered at the veno-atrial junction in both species but not alongside the myocardial sleeve in horses.


Coronary Vessels/anatomy & histology , Heart Atria/anatomy & histology , Horses/anatomy & histology , Imaging, Three-Dimensional/veterinary , Pulmonary Veins/anatomy & histology , Pulmonary Veins/innervation , Swine/anatomy & histology , Telocytes/cytology , Animals , Atrial Fibrillation/therapy , Catheter Ablation , Coronary Vessels/diagnostic imaging , Coronary Vessels/innervation , Disease Models, Animal , Female , Heart Atria/diagnostic imaging , Male , Microscopy, Electron, Transmission , Myocardium/cytology , Pulmonary Veins/cytology , Tomography, X-Ray Computed
14.
J Tissue Eng Regen Med ; 12(2): e786-e806, 2018 02.
Article En | MEDLINE | ID: mdl-27943597

Effective re-endothelialization is critical for the use of decellularized scaffolds for ex vivo lung engineering. Current approaches yield insufficiently re-endothelialized scaffolds that haemorrhage and become thrombogenic upon implantation. Herein, gravity-driven seeding coupled with bioreactor culture facilitated widespread distribution and engraftment of endothelial cells throughout rat lung scaffolds. Initially, human umbilical vein endothelial cells were seeded into the pulmonary artery by either gravity-driven, variable flow perfusion seeding or pump-driven, pulsatile flow perfusion seeding. Gravity seeding evenly distributed cells and supported cell survival and re-lining of the vascular walls while perfusion pump-driven seeding led to increased cell fragmentation and death. Using gravity seeding, rat pulmonary artery endothelial cells and rat pulmonary vein endothelial cells attached in intermediate and large vessels, while rat pulmonary microvascular endothelial cells deposited mostly in microvessels. Combination seeding of these cells led to positive vascular endothelial cadherin staining. In addition, combination seeding improved barrier function as assessed by serum albumin extravasation; however, leakage was observed in the distal portions of the re-endothelialized tissue suggesting that recellularization of the alveoli is necessary to complete barrier function of the capillary-alveolar network. Overall, these data indicate that vascular recellularization of rat lung scaffolds is achieved through gravity seeding. Copyright © 2016 John Wiley & Sons, Ltd.


Endothelial Cells/cytology , Gravitation , Lung/cytology , Tissue Scaffolds/chemistry , Animals , Apoptosis , Bioreactors , Cell Count , Cell Proliferation , Cell Shape , Cell Size , Cell Survival , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Humans , Kinetics , Lung/blood supply , Male , Neovascularization, Physiologic , Perfusion , Pulmonary Artery/cytology , Pulmonary Veins/cytology , Rats, Sprague-Dawley
15.
Adv Anat Embryol Cell Biol ; 228: 21-39, 2018.
Article En | MEDLINE | ID: mdl-29288384

Endothelium plays an important role in maintaining the vascular barrier and physiological homeostasis. Endothelium also is fundamental to the initiation and regulation of inflammation. Endothelium demonstrates phenotypic and functional heterogeneity not only among various organs but also within an organ. One of the striking examples would be the pulmonary endothelium that participates in creating blood-air barrier. Endothelium in large pulmonary blood vessels is distinct in structure and function from that lining of the pulmonary capillaries. This chapter focuses on the comparative aspects of pulmonary endothelium and highlight unique differences such as the presence of pulmonary intravascular macrophages among select species.


Blood-Air Barrier/physiology , Endothelium, Vascular/anatomy & histology , Lung/blood supply , Respiratory Mucosa/anatomy & histology , Anatomy, Comparative , Animals , Buffaloes/anatomy & histology , Buffaloes/physiology , Capillaries/cytology , Capillaries/physiology , Cattle/anatomy & histology , Cattle/physiology , Endothelium, Vascular/physiology , Horses/anatomy & histology , Horses/physiology , Lung/cytology , Lung/physiology , Pulmonary Artery/cytology , Pulmonary Artery/physiology , Pulmonary Veins/cytology , Pulmonary Veins/physiology , Respiratory Mucosa/physiology
16.
Adv Anat Embryol Cell Biol ; 228: 41-61, 2018.
Article En | MEDLINE | ID: mdl-29288385

Pulmonary blood vessels act as a well-regulated barrier to the flux of fluid and solutes between the lumen and the air space. Perturbation of the barrier function results in excessive fluid leak into the interstitium and alveoli, and impairs gas exchange. Recent studies provide deeper insight into the precise control mechanisms involved in the regulation of the barrier. This chapter will highlight these mechanisms and discuss the current understanding on the fluid and solute transport pathways across the vascular endothelial layer. In addition, the chapter summarizes the contributions of extra-endothelial structures such as pericytes and glycocalyx in regulating fluid flux across pulmonary vessels. The chapter concludes with an analysis on the impact of pulmonary endothelial heterogeneity and experimental models on current interpretations of barrier function and regulatory mechanisms.


Blood-Air Barrier/physiology , Endothelium, Vascular/physiology , Lung/physiology , Respiratory Mucosa/physiology , Animals , Biological Transport/physiology , Blood-Air Barrier/cytology , Capillaries/cytology , Capillaries/physiology , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Glycocalyx/physiology , Humans , Lung/blood supply , Lung/cytology , Models, Animal , Pericytes/physiology , Pulmonary Artery/cytology , Pulmonary Artery/physiology , Pulmonary Veins/cytology , Pulmonary Veins/physiology , Respiratory Mucosa/cytology , Water-Electrolyte Balance/physiology
17.
Anat Histol Embryol ; 46(4): 325-333, 2017 Aug.
Article En | MEDLINE | ID: mdl-28421621

Ganglia and telocytes were identified inside the porcine pulmonary veins wall near myocardial sleeve tissue at the atriopulmonary junction. These structures are reported to play a role in the initiation of pulses from outside the heart, which potentially can cause cardiac conduction disorders such as atrial fibrillation. In-depth knowledge on the fine structure of the pulmonary vein wall is a pre-requisite to better understand the underlying pathophysiology of atrial fibrillation and the origin and conduction of ectopic pulses. The importance of pulmonary vein myocardial sleeves as triggering foci for atrial fibrillation has been shown in human patients. In this context, the fine structure of the pulmonary vein wall was investigated qualitatively by light and transmission electron microscopy in the pig, which is a frequently used animal model for development of new treatment strategies. Additionally, intra and extramural ganglia, containing telocytes that create a network near the neurone cell bodies, were identified in pigs. Detailed illustration of the distribution and organization of tissues and cell types, potentially involved in the origin and propagation of ectopic stimuli originating from the pulmonary veins, might lead to a better insight on the actual composition of the tissues affected by ablation as studied in pigs.


Ganglia/cytology , Myocardium/ultrastructure , Pulmonary Veins/cytology , Swine/anatomy & histology , Telocytes/cytology , Animals , Ganglia/ultrastructure , Microscopy, Electron, Transmission/veterinary , Pulmonary Veins/ultrastructure , Telocytes/ultrastructure , Telopodes/ultrastructure
18.
J Pharmacol Sci ; 133(4): 195-202, 2017 Apr.
Article En | MEDLINE | ID: mdl-28410965

The electrophysiological properties underlying the automaticity of the guinea pig pulmonary vein myocardium were studied. About 30% of the isolated pulmonary vein tissue preparations showed spontaneous electrical activity, as shown by glass microelectrode recordings from their myocardial layer. The remaining quiescent preparations had a resting membrane potential less negative than that in the left atria. Blockade of the acetylcholine activated potassium current (IK-ACh) by tertiapin induced a depolarizing shift of the resting membrane potential and automatic electrical activity in the pulmonary vein, but not in the atria. The tertiapin-induced electrical activity, as well as the spontaneous activity, was inhibited by the application of carbachol or by chelation of intracellular Ca2+ by BAPTA. The isolated pulmonary vein cardiomyocytes had an IK-ACh density similar to that of the atrial cardiomyocytes, but a lower density of the inwardly-rectifying potassium current (IK1). Spontaneous Ca2+ transients were observed in about 30% of the isolated pulmonary vein cardiomyocytes, but not in atrial cardiomyocytes. The Ca2+ transients in the pulmonary vein cardiomyocytes were induced by tertiapin and inhibited by carbachol. These results indicate that the pulmonary vein cardiomyocytes have a reduced density of the inwardly-rectifying potassium current, which plays a permissive role in their intracellular Ca2+-dependent automaticity.


Calcium/metabolism , Myocytes, Cardiac/metabolism , Potassium/metabolism , Potassium/physiology , Pulmonary Veins/metabolism , Pulmonary Veins/physiology , Acetylcholine/antagonists & inhibitors , Acetylcholine/pharmacology , Action Potentials/drug effects , Animals , Bee Venoms/antagonists & inhibitors , Bee Venoms/pharmacology , Carbachol/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Electrophysiological Phenomena/drug effects , Guinea Pigs , In Vitro Techniques , Membrane Potentials/drug effects , Microscopy, Confocal , Potassium Channel Blockers/pharmacology , Pulmonary Veins/cytology
19.
Pflugers Arch ; 469(5-6): 703-712, 2017 06.
Article En | MEDLINE | ID: mdl-28243733

The collar of the pulmonary vein (PV) is the focal point for the initiation of atrial arrhythmias, but the mechanisms underlying how PV cells differ from neighboring left atrial tissue are unclear. We examined the biophysical and molecular properties of INa in cells isolated from the canine pulmonary sleeve and compared the properties to left atrial tissue. PV and left atrial myocytes were isolated and patch clamp techniques were used to record INa. Action potential recordings from either tissue type were made using high-resistance electrodes. mRNA was determined using quantitative RT-PCR and proteins were determined by Western blot. Analysis of the action potential characteristics showed that PV tissue had a lower Vmax compared with left atrial tissue. Fast INa showed that current density was slightly lower in PV cells compared with LA cells (-96 ± 18.7 pA/pF vs. -120 ± 6.7 pA/pF, respectively, p < 0.05). The recovery from inactivation of INa in PV cells was slightly slower but no marked difference in steady-state inactivation was noted. Analysis of late INa during a 225-ms pulse showed that late INa was significantly smaller in PV cells compared to LA cells at all measured time points into the pulse. These results suggest PV cells have lower density of both peak and late INa. Molecular analysis of Nav1.5 and the four beta subunits showed lower levels of Nav1.5 as well as Navß1 subunits, confirming the biophysical findings. These data show that a lower density of INa may lead to depression of excitability and predispose the PV collar to re-entrant circuits under pathophysiological conditions.


Action Potentials , Heart Atria/cytology , Myocytes, Cardiac/physiology , Myocytes, Smooth Muscle/physiology , Pulmonary Veins/cytology , Voltage-Gated Sodium Channels/metabolism , Animals , Cells, Cultured , Dogs , Female , Male , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , Sodium/metabolism
20.
Oncotarget ; 7(43): 69231-69242, 2016 Oct 25.
Article En | MEDLINE | ID: mdl-27713141

Fibroblast growth factor 23 (FGF23), elevated in chronic renal failure, increases atrial arrhythmogenesis and dysregulates calcium homeostasis. Late sodium currents (INa-Late) critically induces ectopic activity of pulmoanry vein (the most important atrial fibrillation trigger). This study was to investigate whether FGF23 activates the INa-Late leading to calcium dysregulation and increases PV arrhythmogenesis. Patch clamp, western blot, and confocal microscopy were used to evaluate the electrical activities, calcium homeostasis, and mitochondrial reactive oxygen species (ROS) in PV cardiomyocytes with or without FGF23 (0.1 or 1 ng/mL) incubation for 4~6 h. Compared to the control, FGF23 (1 ng/mL, but not 0.1 ng/mL)-treated PV cardiomyocytes had a faster beating rate. FGF23 (1 ng/mL)-treated PV cardiomyocytes had larger INa-Late, calcium transients, and mitochondrial ROS than controls. However, ranolazine (an inhibitor of INa-Late) attenuated FGF23 (1 ng/mL)-increased beating rates, calcium transients and mitochondrial ROS. FGF23 (1 ng/mL)-treated PV cardiomyocytes exhibited larger phosphorylation of calcium/calmodulin-dependent protein kinase II (CaMKII). Chelerythrine chloride (an inhibitor of protein kinase C) decreased INa-Late in FGF23 (1 ng/mL)-treated PV cardiomyocytes. However, KN93 (a selective CaMKII blocker) decreased INa-Late in control and FGF23 (1 ng/mL)-treated PV cardiomyocytes to a similar extent. In conclusion, FGF23 increased PV arrhythmogenesis through sodium and calcium dysregulation by acting protein kinase C signaling.


Action Potentials/drug effects , Calcium/metabolism , Fibroblast Growth Factors/pharmacology , Homeostasis/drug effects , Myocytes, Cardiac/drug effects , Sodium/metabolism , Animals , Benzophenanthridines/pharmacology , Benzylamines/pharmacology , Fibroblast Growth Factor-23 , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Patch-Clamp Techniques , Protein Kinase Inhibitors/pharmacology , Pulmonary Veins/cytology , Rabbits , Sulfonamides/pharmacology
...