Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 235
Filter
1.
Int J Mol Sci ; 24(13)2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37445678

ABSTRACT

Atrial fibrillation (AF) is the most frequent persistent arrhythmia. Many genes have been reported as a genetic background for AF. However, most transcriptome analyses of AF are limited to the atrial samples and have not been evaluated by multiple cardiac regions. In this study, we analyzed the expression levels of protein-coding and long noncoding RNAs (lncRNAs) in six cardiac regions by RNA-seq. Samples were donated from six subjects with or without persistent AF for left atria, left atrial appendages, right atria, sinoatrial nodes, left ventricles, right ventricles, and pulmonary veins (PVs), and additional four right atrial appendages samples were collected from patients undergoing mitral valve replacement. In total, 23 AF samples were compared to 23 non-AF samples. Surprisingly, the most influenced heart region in gene expression by AF was the PV, not the atria. The ion channel-related gene set was significantly enriched upon analysis of these significant genes. In addition, some significant genes are cancer-related lncRNAs in PV in AF. A co-expression network analysis could detect the functional gene clusters. In particular, the cancer-related lncRNA, such as SAMMSON and FOXCUT, belong to the gene network with the cancer-related transcription factor FOXC1. Thus, they may also play an aggravating role in the pathogenesis of AF, similar to carcinogenesis. In the least, this study suggests that (1) RNA alteration is most intense in PVs and (2) post-transcriptional gene regulation by lncRNA may contribute to the progression of AF. Through the screening analysis across the six cardiac regions, the possibility that the PV region can play a role other than paroxysmal triggering in the pathogenesis of AF was demonstrated for the first time. Future research with an increase in the number of PV samples will lead to a novel understanding of the pathophysiology of AF.


Subject(s)
Atrial Fibrillation , Catheter Ablation , Neoplasms , Pulmonary Veins , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Pulmonary Veins/metabolism , Heart Atria/pathology , Computational Biology , Neoplasms/metabolism
2.
J Cardiothorac Surg ; 18(1): 146, 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37069582

ABSTRACT

BACKGROUND: Although the pressure of pulmonary vein increases before pulmonary artery in pulmonary hypertension due to left heart disease (PH-LHD), only a few studies have assessed pulmonary vein smooth muscle cells (PVSMCs) because of the lack of a simple and feasible isolation method. METHODS: In this study, we introduced a simple method to obtain PVSMCs. Primary pulmonary veins were removed by puncture needle cannula guidance. Then, PVSMCs were cultured by the tissue explant method and purified by the differential adhesion method. The cells were characterized by hematoxylin-eosin (HE) staining, immunohistochemistry, western blotting, and immunofluorescence to observe the morphology and verify the expression of alpha-smooth muscle actin (α-SMA). RESULTS: The HE staining results showed that the pulmonary vein media was thinner than the pulmonary artery, the intima and adventitia of the pulmonary vein were removed by this method, and the obtained cells with good activity exhibited morphological characteristics of smooth muscle cells. In addition, higher α-SMA expression was observed in the cells obtained by our isolation method than in the traditional method. CONCLUSION: This study established a simple and feasible method to isolate and culture PVSMCs that might facilitate the cytological experiments for PH-LHD.


Subject(s)
Hypertension, Pulmonary , Pulmonary Veins , Rats , Animals , Pulmonary Veins/metabolism , Hypertension, Pulmonary/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery , Immunohistochemistry , Cells, Cultured
3.
Int J Mol Sci ; 24(5)2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36902298

ABSTRACT

Pulmonary vein stenosis (PVS) causes a rare type of pulmonary hypertension (PH) by impacting the flow and pressure within the pulmonary vasculature, resulting in endothelial dysfunction and metabolic changes. A prudent line of treatment in this type of PH would be targeted therapy to relieve the pressure and reverse the flow-related changes. We used a swine model in order to mimic PH after PVS using pulmonary vein banding (PVB) of the lower lobes for 12 weeks to mimic the hemodynamic profile associated with PH and investigated the molecular alterations that provide an impetus for the development of PH. Our current study aimed to employ unbiased proteomic and metabolomic analyses on both the upper and lower lobes of the swine lung to identify regions with metabolic alterations. We detected changes in the upper lobes for the PVB animals mainly pertaining to fatty acid metabolism, reactive oxygen species (ROS) signaling and extracellular matrix (ECM) remodeling and small, albeit, significant changes in the lower lobes for purine metabolism.


Subject(s)
Hypertension, Pulmonary , Pulmonary Veins , Swine , Animals , Hypertension, Pulmonary/metabolism , Proteomics , Lung/metabolism , Metabolomics , Pulmonary Veins/metabolism
4.
Biomolecules ; 12(5)2022 05 20.
Article in English | MEDLINE | ID: mdl-35625651

ABSTRACT

Ectopic excitability in pulmonary veins (PVs) is the major cause of atrial fibrillation. We previously reported that the inositol trisphosphate receptor in rat PV cardiomyocytes cooperates with the Na+-Ca2+ exchanger to provoke ectopic automaticity in response to norepinephrine. Here, we focused on adenylyl cyclase (AC) as another effector of norepinephrine stimulation. RT-PCR, immunohistochemistry, and Western blotting revealed that the abundant expression of Ca2+-stimulable AC3 was restricted to the supraventricular area, including the PVs. All the other AC isotypes hardly displayed any region-specific expressions. Immunostaining of isolated cardiomyocytes showed an enriched expression of AC3 along the t-tubules in PV myocytes. The cAMP-dependent response of L-type Ca2+ currents in the PV and LA cells is strengthened by the 0.1 mM intracellular Ca2+ condition, unlike in the ventricular cells. The norepinephrine-induced automaticity of PV cardiomyocytes was reversibly suppressed by 100 µM SQ22536, an adenine-like AC inhibitor. These findings suggest that the specific expression of AC3 along t-tubules may contribute to arrhythmogenic automaticity in rat PV cardiomyocytes.


Subject(s)
Atrial Fibrillation , Pulmonary Veins , Adenylyl Cyclases/metabolism , Animals , Myocytes, Cardiac/metabolism , Norepinephrine/metabolism , Norepinephrine/pharmacology , Pulmonary Veins/metabolism , Rats , Sodium-Calcium Exchanger/metabolism
5.
Bull Exp Biol Med ; 172(6): 671-675, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35501645

ABSTRACT

Cardiac-specific microRNA miR-133a-3p modulates adrenergic signaling. Adrenergic receptors and their intracellular pathways are the key players in proarrhythmic ectopy derived from the myocardial sleeves of the pulmonary veins. We studied the effect of miR-133a-3p on ectopy induced by norepinephrine in myocardial tissue of rat pulmonary veins. Using microelectrode technique, we revealed facilitation of proarrhythmic pattern of spontaneous bursts of action potentials induced by norepinephrine in tissue preparations of the pulmonary veins isolated from rats in 24 h after injection of a transfection mixture containing miR-133a-3p (1 mg/kg) in vivo. According to ELISA data, the cAMP level in the pulmonary vein myocardium of rats receiving miR-133a-3p was 2-fold higher than in control animals. Bioinformatic analysis showed that mRNA of protein phosphatases and some phosphodiesterases are most probable targets of miR-133a-3p. The proarrhythmic effect of miR-133a-3p can be related to inhibition of the expression of phosphodiesterases accompanied by cAMP accumulation and increased intracellular ß-adrenergic signaling.


Subject(s)
Cyclic AMP , MicroRNAs , Myocardium , Pulmonary Veins , Animals , Cyclic AMP/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardium/metabolism , Norepinephrine/pharmacology , Phosphoric Diester Hydrolases/metabolism , Pulmonary Veins/metabolism , Rats , Receptors, Adrenergic, beta/metabolism
6.
J Mol Cell Cardiol ; 161: 86-97, 2021 12.
Article in English | MEDLINE | ID: mdl-34375616

ABSTRACT

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.


Subject(s)
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
8.
J Cardiovasc Pharmacol ; 78(2): 280-287, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34050090

ABSTRACT

ABSTRACT: Aquaporins (AQPs) are a group of membrane proteins related to water permeability. Studies have shown that AQPs play a vital role in various diseases. Whether AQPs participate in regulating vascular permeability after sepsis and whether the subtype of AQPs is related are unknown. Ss-31, as a new antioxidant, had protective effects on a variety of diseases. However, whether Ss-31 has a protective effect on pulmonary vascular permeability in sepsis and whether its effect is related to AQPs are unclear. Using the cecum ligation perforation-induced septic rat and LPS-treated pulmonary vein endothelial cells, the role of AQPs in the regulation of the permeability of pulmonary vascular and its relationship to Ss-31 were studied. The results showed that the pulmonary vascular permeability significantly increased after sepsis, meanwhile the expressions of AQP3, 4, and 12 increased. Among those, the AQP3 was closely correlated with pulmonary vascular permeability. The inhibition of AQP3 antagonized the increase of the permeability of monolayer pulmonary vein endothelial cells. Further study showed that the expression of caveolin-1 (Cav-1) increased and occludin decreased after sepsis. The inhibition of AQP3 antagonized the decrease of Cav-1 and the increase of occludin in sepsis. Antioxidant Ss-31 decreased the expression of AQP3 and ROS levels. At the same time, Ss-31 improved pulmonary vascular permeability and prolonged survival of sepsis rats. In conclusion, AQP3 participates in the regulation of pulmonary vascular permeability after sepsis, and the antioxidant Ss-31 has a protective effect on pulmonary vascular permeability by downregulating the expression of AQP3 and inhibiting ROS production.


Subject(s)
Antioxidants/pharmacology , Aquaporin 3/metabolism , Capillary Permeability/drug effects , Endothelial Cells/drug effects , Oligopeptides/pharmacology , Pulmonary Veins/drug effects , Sepsis/drug therapy , Animals , Aquaporin 3/genetics , Caveolin 1/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Female , Lipopolysaccharides/toxicity , Male , Occludin/metabolism , Oxidative Stress/drug effects , Pulmonary Veins/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sepsis/genetics , Sepsis/metabolism , Sepsis/microbiology , Signal Transduction
9.
J Cardiothorac Surg ; 16(1): 127, 2021 May 10.
Article in English | MEDLINE | ID: mdl-33971931

ABSTRACT

BACKGROUND: This study aimed to explore whether the mechanical stretching-induced expression of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in pulmonary veins occurred through the stretch-activated channel (SAC)/ mitogen-activated protein kinases (MAPKs) pathway. METHODS: Sixty male Sprague-Dawley rats were divided into three sham groups and seven model groups. A metal clip was placed on the ascending aorta in the model group to establish PH-LHD rat model. The sham group received a similar operation without ascending aorta clamped. On day 25, pulmonary vein was given mechanical stretching with 0 g, 2.0 g tension in two model groups and two sham groups. Another four model groups were given 2.0 g tension after MAPKs pathway inhibitors soaked. The last sham group and model group rats' pulmonary veins, pulmonary artery and lung tissues were obtained on day 35. Pulmonary vein, pulmonary artery and lung tissue were evaluated by echocardiography, HE staining, immunohistochemistry and western blotting respectively. RESULTS: On day 25, left heart weight, right ventricular pressure (35.339 cmH2O) and left atrial pressure (13.657 cmH2O) were increased in model group than those in sham group. Echocardiography showed left heart failure in the PH-LHD group (Interventrieular septum dimension 1.716 mm, left ventricular internal end diastolic dimension 4.888 mm, left ventricular posterior wall thickness in diastole 1.749 mm, ejection fraction 76.917%). But there was no difference in lung tissue between the sham group and PH-LHD group as showed by HE staining. Our results showed that the expression of IL-6 and TNF-α was highly expressed in PH-LHD rats' serum and pulmonary vein, which were further increased after 2.0 g tension was given and were decreased after SAC/MAPKs inhibitors treatment. Meanwhile, on day 25, immunohistochemistry analysis showed the expression of IL-6 and TNF-α was higher in the PH-LHD rats' pulmonary vein than that in pulmonary artery and lung tissue, and these expressions in pulmonary vein of PH-LHD group were also higher than that in sham group. However, on day 35, IL-6 and TNF-α were all increased in the pulmonary veins, arteries and lung tissues. Besides, our results uncovered that SAC/MAPKs pathway were upregulating in PH-LHD rats' pulmonary vein. CONCLUSION: In conclusion, pulmonary vein mechanical stretching exacerbated PH-LHD possibly through the SAC/MAPKs pathway and upregulating expression of IL-6 and TNF-α.


Subject(s)
Hypertension, Pulmonary/etiology , Interleukin-6/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Pulmonary Veins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Remodeling/physiology , Ventricular Dysfunction, Left/physiopathology , Animals , Biomarkers/metabolism , Biomechanical Phenomena , Blotting, Western , Echocardiography , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Immunohistochemistry , Male , Pulmonary Veins/diagnostic imaging , Pulmonary Veins/pathology , Pulmonary Veins/physiopathology , Random Allocation , Rats , Rats, Sprague-Dawley , Signal Transduction , Up-Regulation
10.
Transl Res ; 229: 24-37, 2021 03.
Article in English | MEDLINE | ID: mdl-33045409

ABSTRACT

Despite significant morbidity among infants with single ventricle heart disease (SVHD), clinical monitoring is limited by poor understanding of the underlying pathobiology. Proteomics can identify novel biomarkers and important pathways in complex disease. No prior study has evaluated whether the proteome of SVHD infants differs from healthy controls, how it shifts after stage 2 palliation, or whether differences can predict post-operative outcomes. We present a prospective cohort study of cardiovascular proteomic phenotyping in infants with SVHD undergoing stage 2 palliation. Twenty-nine pre-stage-2 SVHD infants and 25 healthy controls were enrolled. Outcomes included postoperative hypoxemia and endotracheal intubation time. Serum samples were drawn pre-operatively (systemic and pulmonary vein) and at 24 hours postoperation. Targeted cardiovascular proteomic analysis included 184 proteins. Partial least squares discriminant analysis distinguished cases from controls (Accuracy = 0.98, R2 = 0.93, Q2 = 0.81) with decreased inflammatory mediators and increased modulators of vascular tone. Partial least squares discriminant analysis also distinguished cases pre-operation vs. post-operation (Accuracy=0.98, R2=0.99, Q2 = 0.92) with postoperative increase in both inflammatory and vascular tone mediators. Pre-operation pulmonary vein tissue inhibitor of metalloproteinase-1 (1.8x-fold, p=1.6 × 10-4) and nidogen-1 (1.5x-fold, p=1.7 × 10-4) were higher in subjects with longer endotracheal intubation time. Postoperation matrix metalloproteinase 7 levels were higher in subjects with greater postoperative hypoxemia (1.5x-fold, P= 1.97 × 10-5). Proteomic analysis identifies significant changes among SVHD infants pre- and post-stage 2, and healthy controls. Tissue inhibitor of metalloproteinase-1, nidogen-1, and matrix metalloproteinase 7 levels are higher in SVHD cases with greater morbidity suggesting an important role for regulation of extracellular matrix production. Proteomic profiling may identify high-risk SVHD infants.


Subject(s)
Blood Proteins/analysis , Fontan Procedure/adverse effects , Biomarkers/blood , Cardiac Catheterization , Case-Control Studies , Female , Humans , Hypoxia/blood , Hypoxia/etiology , Infant , Male , Matrix Metalloproteinase 7/blood , Palliative Care/methods , Postoperative Complications/blood , Postoperative Complications/etiology , Preoperative Period , Prospective Studies , Proteomics , Pulmonary Veins/metabolism , Treatment Outcome , Univentricular Heart/surgery
11.
PLoS One ; 15(9): e0235824, 2020.
Article in English | MEDLINE | ID: mdl-32881898

ABSTRACT

Pulmonary hypertension due to left heart disease (PH-LHD) is a momentous pulmonary hypertension disease, and left heart disease is the most familiar cause. Mechanical stretching may be a crucial cause of vascular remodeling. While, the underlining mechanism of mechanical stretching-induced in remodeling of pulmonary vein in the early stage of PH-LHD has not been completely elucidated. In our study, the PH-LHD model rats were successfully constructed. After 25 days, doppler echocardiography and hemodynamic examination were performed. In addition, after treatment, the levels of matrix metalloproteinase-9 (MMP-9) and transforming growth factor-ß1 (TGF-ß1) were determined by ELISA, immunohistochemistry and western blot assays in the pulmonary veins. Moreover, the pathological change of pulmonary tissues was evaluated by H&E staining. Our results uncovered that left ventricular insufficiency and interventricular septal shift could be observed in PH-LHD model rats, and the right ventricular systolic pressure (RVSP) and mean left atrial pressure (mLAP) were also elevated in PH-LHD model rats. Meanwhile, we found that MMP-9 and TGF-ß1 could be highly expressed in PH-LHD model rats. Besides, we revealed that stretch-activated channel (SAC)/mitogen-activated protein kinases (MAPKs) signaling pathway could be involved in the upregulations of MMP-9 and TGF-ß1 mediated by mechanical stretching in pulmonary vein. Therefore, current research revealed that mechanical stretching induced the increasing expressions of MMP-9 and TGF-ß1 in pulmonary vein, which could be mediated by activation of SAC/MAPKs signaling pathway in the early stage of PH-LHD.


Subject(s)
Hypertension, Pulmonary/physiopathology , MAP Kinase Signaling System , Matrix Metalloproteinase 9/metabolism , Pulmonary Veins/physiopathology , Transforming Growth Factor beta1/metabolism , Ventricular Dysfunction, Left/physiopathology , Animals , Biomechanical Phenomena , Disease Models, Animal , Enzyme Activation , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Male , Pulmonary Veins/metabolism , Rats , Rats, Sprague-Dawley , Stress, Mechanical , Vascular Remodeling , Ventricular Dysfunction, Left/complications , Ventricular Dysfunction, Left/metabolism
12.
Article in English | MEDLINE | ID: mdl-32673988

ABSTRACT

Pulmonary hypertension (PH) is a progressive and life-threating lung disorder characterized by elevated pulmonary artery pressure and vascular remodeling. PH is classified into five groups, and one of the most common and lethal forms, PH Group-III is defined as PH due to lung diseases and/or hypoxia. Due to the lack of studies in this group, PH-specific drug therapies including prostacyclin (PGI2) analogues have not been approved or recommended for use in these patients. PGI2 is synthesized by the PGI2 synthase (PGIS) enzyme, and its production is determined by measuring its stable metabolite, 6-keto-PGF1α. An impaired PGI2 pathway has been observed in PH animal models and in PH Group-I patients; however, there are contradictory results. The aim of this study is to determine whether PH Group-III is associated with altered expression of PGIS and production of PGI2 in humans. To explore this hypothesis, we measured PGIS expression (by western blot) and PGI2 production (by ELISA) in a large variety of preparations from the pulmonary circulation including human pulmonary artery, pulmonary vein, distal lung tissue, pulmonary artery smooth muscle cells (hPASMC), and bronchi in PH Group-III (n = 35) and control patients (n = 32). Our results showed decreased PGIS expression and/or 6-keto-PGF1α levels in human pulmonary artery, hPASMC, and distal lung tissue derived from PH Group-III patients. Moreover, the production of 6-keto-PGF1α from hPASMC positively correlated with PGIS expression and was inversely correlated with mean pulmonary artery pressure. On the other hand, PH Group-III pulmonary veins and bronchi did not show altered PGI2 production compared to controls. The deficit in PGIS expression and/or PGI2 production observed in pulmonary artery and distal lung tissue in PH Group-III patients may have important implications in the pathogenesis and treatment of PH Group-III.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Epoprostenol/metabolism , Hypertension, Pulmonary/metabolism , Intramolecular Oxidoreductases/metabolism , Pulmonary Artery/metabolism , Bronchi/enzymology , Bronchi/metabolism , Cell Hypoxia/physiology , Cells, Cultured , Dinoprost/metabolism , Down-Regulation , Female , Humans , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/physiopathology , Lung/enzymology , Lung/metabolism , Male , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/enzymology , Pulmonary Veins/enzymology , Pulmonary Veins/metabolism
13.
J Pharmacol Sci ; 143(4): 325-329, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32487451

ABSTRACT

The effects of class I antiarrhythmic drugs on the automaticity of isolated guinea pig pulmonary vein myocardia were investigated using microelectrode and voltage clamp methods. All of the drugs examined reduced the maximum rate of rise of automatic action potentials. The firing frequency and rate of diastolic depolarization were decreased by aprindine, flecainide and propafenone, but not by cibenzoline, disopyramide and pilsicainide, which correlated with blockade of the sodium current component induced by ramp depolarization mimicking the diastolic depolarization. In conclusion, class I antiarrhythmic drugs which block the diastolic sodium current component inhibit the automaticity of the pulmonary vein myocardium.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Pulmonary Veins/drug effects , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/classification , Guinea Pigs , In Vitro Techniques , Microelectrodes , Patch-Clamp Techniques , Pulmonary Veins/metabolism , Sodium/metabolism
14.
Pflugers Arch ; 472(8): 1019-1029, 2020 08.
Article in English | MEDLINE | ID: mdl-32556635

ABSTRACT

In rat pulmonary vein (PV) cardiomyocytes (CM), norepinephrine (NE) induces an automatic activity consisting of bursts of slow action potentials which depend on Ca2+ (upstroke) and Na+ (inter-burst) channels. Our objective was to characterize low voltage-activated (LVA) currents in rat PVCM susceptible to trigger this activity. Whole-cell ICa (5 mM Ca2+) was recorded from - 100 mV with classical Na+- and K+-free solutions. A fast LVA ICa (FLVA-ICa), present in ≈ 56% of PVCM between ~ - 50 to - 20 mV, was blocked by 10 µM TTX and markedly increased by addition of NaCl (1 or 3 mM) or KCl (5 or 10 mM). Permeability ratios P'Ca/PNa and P'Ca/PK calculated for bi-ionic conditions were respectively 2.25 ± 0.51 and 1.88 ± 0.25, and not different from a value of 2. FLVA-ICa was increased by 10 µM NE and 300 nM BayK8644, decreased by 5 µM nifedipine but not blocked by ranolazine (10 µM). NiCl2 (40 µM) and TTA-A2 (10 or 100 nM) increased FLVA-ICa. Similar results were obtained in left atrial (LA) CM. Neither Ba2+ nor Sr2+ alone could permeate the FLVA channel or block Ca2+ influx but revealed a large slower activating and inactivating LVA Ca2+ current (SLVA-ICa), present in 10 out of 80 PVCM, absent in LACM, and partially inhibited by 100 nM TTA-A2. Therefore, the ionic channel underlying FLVA-ICa is likely a fast voltage-gated non-selective channel with a dihydropyridine binding site. SLVA-ICa might correspond to Ca2+ influx through Cav3.x channels and contribute to triggering NE-induced automatic activity in the PV myocardial sleeve.


Subject(s)
Action Potentials/physiology , Calcium Channels, T-Type/metabolism , Cations/metabolism , Myocytes, Cardiac/metabolism , Pulmonary Veins/metabolism , Animals , Male , Patch-Clamp Techniques/methods , Rats , Rats, Wistar
15.
Physiol Rep ; 8(9): e14432, 2020 05.
Article in English | MEDLINE | ID: mdl-32401431

ABSTRACT

The electrophysiological properties of pulmonary vein (PV)-cardiomyocytes, and their responses to the sympathetic neurotransmitter, noradrenaline (NA), are thought to differ from those of the left atrium (LA) and contribute to atrial ectopy. The aim of this study was to examine rat PV cardiomyocyte electrophysiology and responses to NA in comparison with LA cells. LA and PV cardiomyocytes were isolated from adult male Wistar rat hearts, and membrane potentials and ion currents recorded at 36°C using whole-cell patch-clamp techniques. PV and LA cardiomyocytes did not differ in size. In control, there were no differences between the two cell-types in zero-current potential or action potential duration (APD) at 1 Hz, although the incidence of early afterdepolarizations (EADs) was greater in PV than LA cardiomyocytes. The L-type Ca2+ current (ICaL ) was ~×1.5 smaller (p = .0029, Student's t test) and the steady-state K+ current (IKss ) was ~×1.4 larger (p = .0028, Student's t test) in PV than in LA cardiomyocytes. PV cardiomyocyte inward-rectifier current (IK1 ) was slightly smaller than LA cardiomyocyte IK1 . In LA cardiomyocytes, NA significantly prolonged APD30 . In PV cells, APD30 responses to 1 µM NA were heterogeneous: while the mean percentage change in APD30 was not different from 0 (16.5 ± 9.7%, n cells/N animals = 12/10, p = .1177, one-sample t test), three cells showed shortening (-18.8 ± 6.0%) whereas nine showed prolongation (28.3 ± 10.1%, p = .008, Student's t test). NA had no effect on IK1 in either cell-type but inhibited PV IKss by 41.9 ± 4.1% (n/N = 23/11 p < .0001), similar to LA cells. NA increased ICaL in most PV cardiomyocytes (median × 2.2-increase, p < .0001, n/N = 32/14, Wilcoxon-signed-rank test), although in 7/32 PV cells ICaL was decreased following NA. PV cardiomyocytes differ from LA cells and respond heterogeneously to NA.


Subject(s)
Ion Channels/physiology , Myocytes, Cardiac/physiology , Norepinephrine/pharmacology , Pulmonary Veins/physiology , Action Potentials/physiology , Adrenergic alpha-Agonists/pharmacology , Animals , Heart Atria/cytology , Heart Atria/drug effects , Heart Atria/metabolism , Heart Atria/physiopathology , Ion Channels/metabolism , Male , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques/methods , Pulmonary Veins/drug effects , Pulmonary Veins/metabolism , Rats , Rats, Wistar
16.
PLoS One ; 15(5): e0233176, 2020.
Article in English | MEDLINE | ID: mdl-32421724

ABSTRACT

INTRODUCTION: Levosimendan is approved for acute heart failure. Within this context, pulmonary hypertension represents a frequent co-morbidity. Hence, the effects of levosimendan on segmental pulmonary vascular resistance (PVR) are relevant. So far, this issue has been not studied. Beyond that the relaxant effects of levosimendan in human pulmonary vessel are unknown. We addressed these topics in rats' isolated perfused lungs (IPL) and human precision-cut lung slices (PCLS). MATERIAL AND METHODS: In IPL, levosimendan (10 µM) was perfused in untreated and endothelin-1 pre-contracted lungs. The pulmonary arterial pressure (PPA) was continuously recorded and the capillary pressure (Pcap) was determined by the double-occlusion method. Thereafter, segmental PVR, expressed as precapillary (Rpre) and postcapillary resistance (Rpost) and PVR were calculated. Human PCLS were prepared from patients undergoing lobectomy. Levosimendan-induced relaxation was studied in naïve and endothelin-1 pre-contracted PAs and PVs. In endothelin-1 pre-contracted PAs, the role of K+-channels was studied by inhibition of KATP-channels (glibenclamide), BKCa2+-channels (iberiotoxin) and Kv-channels (4-aminopyridine). All changes of the vascular tone were measured by videomicroscopy. In addition, the increase of cAMP/GMP due to levosimendan was measured by ELISA. RESULTS: Levosimendan did not relax untreated lungs or naïve PAs and PVs. In IPL, levosimendan attenuated the endothelin-1 induced increase of PPA, PVR, Rpre and Rpost. In human PCLS, levosimendan relaxed pre-contracted PAs or PVs to 137% or 127%, respectively. In pre-contracted PAs, the relaxant effect of levosimendan was reduced, if KATP- and Kv-channels were inhibited. Further, levosimendan increased cGMP in PAs/PVs, but cAMP only in PVs. DISCUSSION: Levosimendan reduces rats' segmental PVR and relaxes human PAs or PVs, if the pulmonary vascular tone is enhanced by endothelin-1. Regarding levosimendan-induced relaxation, the activation of KATP- and Kv-channels is of impact, as well as the formation of cAMP and cGMP. In conclusion, our results suggest that levosimendan improves pulmonary haemodynamics, if PVR is increased as it is the case in pulmonary hypertension.


Subject(s)
Hypertension, Pulmonary , Lung , Pulmonary Artery , Pulmonary Veins , Simendan/pharmacology , Vascular Resistance/drug effects , Animals , Female , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Lung/blood supply , Lung/metabolism , Lung/physiopathology , Male , Perfusion , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Pulmonary Veins/metabolism , Pulmonary Veins/physiopathology , Rats , Rats, Wistar
17.
Heart Vessels ; 35(7): 1025-1035, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32270355

ABSTRACT

Atrial fibrillation (AF) is one of the most prevalent arrhythmias. Myocardial sleeves of the pulmonary vein are critical in the occurrence of AF. Our study aims to investigate the effect of synthetic vascular smooth muscle cells (SMCs) on gap junction proteins in cardiomyocytes. (1) Extraction of vascular SMCs from the pulmonary veins of Norway rats. TGF-ß1 was used to induce the vascular SMCs switching to the synthetic phenotype and 18-α-GA was used to inhibit gap junctions of SMCs. The contractile and synthetic phenotype vascular SMCs were cocultured with HL-1 cells; (2) Western blotting was used to detect the expression of Cx43, Cx40 and Cx45 in HL-1 cells, and RT-PCR to test microRNA 27b in vascular SMCs or in HL-1 cells; (3) Lucifer yellow dye transfer experiment was used to detect the function of gap junctions. (1) TGF- ß1 induced the vascular SMCs switching to synthetic phenotype; (2) Cx43 was significantly increased, and Cx40 and Cx45 were decreased in HL-1 cocultured with synthetic SMCs; (3) The fluorescence intensity of Lucifer yellow was higher in HL-1 cocultured with synthetic SMCs than that in the cells cocultured with contractile SMCs, which was inhibited by18-α-GA; (4) the expression of microRNA 27b was increased in HL-1 cocultured with synthetic SMCs, which was attenuated markedly by 18-α-GA. (5) the expression of ZFHX3 was decreased in HL-1 cocultured with synthetic SMCs, which was reversed by 18-α-GA. The gap junction proteins of HL-1 were regulated by pulmonary venous SMCs undergoing phenotypic transition in this study, accompanied with the up-regulation of microRNA 27b and the down-regulation of ZFHX3 in HL-1 cells, which was associated with heterocellular gap junctions between HL-1 and pulmonary venous SMCs.


Subject(s)
Cell Communication , Cell Plasticity , Connexins/metabolism , Gap Junctions/metabolism , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Cell Communication/drug effects , Cell Line , Cell Plasticity/drug effects , Coculture Techniques , Connexin 43/genetics , Connexin 43/metabolism , Connexins/genetics , Gap Junctions/drug effects , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , MicroRNAs/genetics , Muscle, Smooth, Vascular/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Smooth Muscle/drug effects , Phenotype , Pulmonary Veins/metabolism , Rats , Signal Transduction , Transforming Growth Factor beta1/pharmacology , Gap Junction alpha-5 Protein
18.
Sci Rep ; 10(1): 5390, 2020 03 25.
Article in English | MEDLINE | ID: mdl-32214185

ABSTRACT

Pulmonary veins (PV) are involved in the pathophysiology of paroxysmal atrial fibrillation. In the rat, left atrium (LA) and PV cardiomyocytes have different reactions to α1-adrenergic receptor activation. In freely beating atria-PV preparations, we found that electrical field potential (EFP) originated from the sino-atrial node propagated through the LA and the PV. The α1-adrenergic receptor agonist cirazoline induced a progressive loss of EFP conduction in the PV whereas it was maintained in the LA. This could be reproduced in preparations electrically paced at 5 Hz in LA. During pacing at 10 Hz in the PV where high firing rate ectopic foci can occur, cirazoline stopped EFP conduction from the PV to the LA, which allowed the sino-atrial node to resume its pace-making function. Loss of conduction in the PV was associated with depolarization of the diastolic membrane potential of PV cardiomyocytes. Adenosine, which reversed the cirazoline-induced depolarization of the diastolic membrane potential of PV cardiomyocytes, restored full over-shooting action potentials and EFP conduction in the PV. In conclusion, selective activation of α1-adrenergic receptors results in the abolition of electrical conduction within the PV. These results highlight a potentially novel pharmacological approach to treat paroxysmal atrial fibrillation by targeting directly the PV myocardium.


Subject(s)
Atrial Fibrillation/physiopathology , Pulmonary Veins/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Action Potentials/physiology , Adrenergic alpha-1 Receptor Antagonists/metabolism , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Animals , Atrial Fibrillation/metabolism , Electric Conductivity , Heart Atria/physiopathology , Heart Conduction System/physiopathology , Heart Rate , Male , Membrane Potentials , Myocardium/pathology , Myocytes, Cardiac/pathology , Pulmonary Veins/physiology , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1/physiology , Sinoatrial Node/physiopathology
19.
Biomolecules ; 10(1)2020 01 17.
Article in English | MEDLINE | ID: mdl-31963453

ABSTRACT

Retinoic acid (RA) is a key molecular player in embryogenesis and adult tissue homeostasis. In embryo development, RA plays a crucial role in the formation of different organ systems, namely, the respiratory system. During lung development, there is a spatiotemporal regulation of RA levels that assures the formation of a fully functional organ. RA signaling influences lung specification, branching morphogenesis, and alveolarization by regulating the expression of particular target genes. Moreover, cooperation with other developmental pathways is essential to shape lung organogenesis. This review focuses on the events regulated by retinoic acid during lung developmental phases and pulmonary vascular development; also, it aims to provide a snapshot of RA interplay with other well-known regulators of lung development.


Subject(s)
Lung/blood supply , Lung/growth & development , Tretinoin/metabolism , Animals , Gene Expression Regulation, Developmental , Humans , Lung/embryology , Lung/metabolism , Lung Diseases/etiology , Lung Diseases/metabolism , Pulmonary Artery/embryology , Pulmonary Artery/growth & development , Pulmonary Artery/metabolism , Pulmonary Veins/embryology , Pulmonary Veins/growth & development , Pulmonary Veins/metabolism , Signal Transduction , Tretinoin/analysis
20.
Hypertens Res ; 43(4): 271-280, 2020 04.
Article in English | MEDLINE | ID: mdl-31853041

ABSTRACT

Chronic hypoxia (CH) causes remodeling not only in pulmonary arteries but also in pulmonary veins. Pulmonary vascular remodeling stems from increased pulmonary vascular myocyte proliferation. However, the pathogenesis of CH-induced proliferation of pulmonary venous smooth muscle cells (PVSMCs) remains unknown. The present study aimed to explore the mechanisms by which CH affects PVSMCs proliferation. PVSMCs were isolated from rat distal pulmonary veins and exposed to CH (4% O2 for 60 h). The expression of calcium sensing receptor (CaSR) was determined by immunofluorescence, real-time quantitative PCR and Western blotting. Cell proliferation was assessed by cell counting, CCK-8 assay, and BrdU incorporation. Apoptosis analysis was examined by flow cytometry. In rat distal PVSMCs, CH increased the cell number and cell viability and enhanced DNA synthesis, which is accompanied by upregulated mRNA and protein expression levels of CaSR. Two negative CaSR modulators (NPS2143, NPS2390) not only attenuated CH-induced CaSR upregulation but also inhibited CH-induced increases in cell number, cell viability and the proliferation index of PVSMCs, whereas two positive modulators (spermine, R568) not only amplified CH-induced CaSR upregulation but also intensified CH-induced increases in cell number, cell viability and the proliferation index of PVSMCs. Silencing CaSR with siRNA similarly attenuated the CH-induced enhancement of cell number, cell viability and DNA synthesis in PVSMCs. Neither CH nor downregulation of CaSR with siRNA had an effect on apoptosis in PVSMCs. These results suggest that CaSR mediating excessive proliferation is a new pathogenic mechanism involved in the initiation and progression of distal PVSMCs proliferation under CH conditions.


Subject(s)
Cell Proliferation/physiology , Hypoxia/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Veins/metabolism , Receptors, Calcium-Sensing/metabolism , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Hypoxia/pathology , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Naphthalenes/pharmacology , Pulmonary Veins/drug effects , Pulmonary Veins/pathology , Quinoxalines/pharmacology , Rats , Rats, Wistar , Up-Regulation/drug effects , Vascular Remodeling/drug effects , Vascular Remodeling/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...