Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.530
Filter
1.
Turk Kardiyol Dern Ars ; 52(4): 237-243, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38829634

ABSTRACT

OBJECTIVE: This study aims to explore the impact of sodium-glucose cotransporter-2 (SGLT-2) inhibitors, a newer class of oral antidiabetic drugs, on atrial electromechanical delay (EMD) in patients with type 2 diabetes mellitus (DM). This is particularly relevant given the significantly higher incidence of atrial fibrillation (AF) in diabetic patients compared to the general population. Atrial electromechanical delay is recognized as an important factor influencing the development of atrial fibrillation. METHODS: This study included 30 type 2 DM patients (53.3% female, mean age 60.07 ± 10.03 years), initiating treatment with SGLT-2 inhibitors. The patients were assessed using echocardiography at baseline and again at 6 months, focusing on basic echocardiographic parameters and atrial electromechanical delay times (EMD) measured via tissue Doppler imaging. RESULTS: No significant changes were observed in intra-atrial EMD times. However, significant reductions were noted in interatrial EMD times, decreasing from 15.13 ± 5.87 ms to 13.20 ± 6.12 ms (P = 0.029). Statistically significant shortening occurred in lateral pulmonary acceleration (PA) times (from 58.73 ± 6.41 ms to 54.37 ± 6.97 ms, P < 0.001), septal PA times (from 50.90 ± 6.02 ms to 48.23 ± 5), and tricuspid PA times (from 43.60 ± 6.28 ms to 41.30 ± 5.60 ms, P = 0.003). Additionally, there was a significant reduction in the E/e' ratio from 8.13 ± 4.0 to 6.50 ± 2.37 (P = 0.003). CONCLUSION: SGLT-2 inhibitors might positively influence atrial electromechanical conduction, reducing DM-related functional impairments and the risk of arrhythmias, particularly AF.


Subject(s)
Atrial Fibrillation , Diabetes Mellitus, Type 2 , Sodium-Glucose Transporter 2 Inhibitors , Humans , Female , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/complications , Middle Aged , Male , Atrial Fibrillation/drug therapy , Atrial Fibrillation/physiopathology , Heart Atria/diagnostic imaging , Heart Atria/drug effects , Heart Atria/physiopathology , Aged , Heart Conduction System/drug effects , Heart Conduction System/physiopathology , Echocardiography
2.
Cardiovasc Toxicol ; 24(6): 550-562, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38696070

ABSTRACT

Trastuzumab is widely used in HER2 breast cancer. However, it may cause left ventricular (LV) dysfunction. A decrease in LV global longitudinal strain (GLS) has been previously demonstrated to be a good predictor of subsequent cancer therapy related dysfunction (CTRCD). Left atrial morphological remodeling during Trastuzumab therapy has also been shown. The aim of this study is exploring the relationship between early changes in left atrial function and the development of Trastuzumab-induced cardiotoxicity. Consecutive patients with diagnosis of HER2+non-metastatic breast cancer treated with Trastuzumab were prospectively enrolled. A clinical, conventional, and advanced echocardiographic assessment was performed at baseline and every three months, until a one-year follow-up was reached. One-hundred-sixteen patients completed the 12 months follow-up, 10 (9%) cases of CTRCD were observed, all after the sixth month. GLS and LVEF significantly decreased in the CTRCD group at 6 months of follow-up, with an earlier (3 months) significant worsening in left atrial morpho-functional parameters. Systolic blood pressure, early peak atrial longitudinal strain (PALS), peak atrial contraction (PACS) and left atrial volume (LAVI) changes resulted independent predictors of CTRCD at multivariable logistic regression analysis. Moreover, early changes in PALS and PACS resulted good predictors of CTRCD development (AUC 0.85; p = 0.008, p < 0.001 and 0.77; p = 0.008, respectively). This prospective study emphasizes that the decline in PALS and PACS among trastuzumab-treated patients could possibly increase the accuracy in identifying future CTRCD in non-metastatic HER2 breast cancer cases, adding predictive value to conventional echocardiographic assessment.


Subject(s)
Antineoplastic Agents, Immunological , Atrial Function, Left , Breast Neoplasms , Cardiotoxicity , Receptor, ErbB-2 , Trastuzumab , Ventricular Function, Left , Humans , Trastuzumab/adverse effects , Female , Breast Neoplasms/drug therapy , Middle Aged , Receptor, ErbB-2/metabolism , Prospective Studies , Antineoplastic Agents, Immunological/adverse effects , Ventricular Function, Left/drug effects , Atrial Function, Left/drug effects , Adult , Time Factors , Risk Factors , Treatment Outcome , Aged , Predictive Value of Tests , Risk Assessment , Atrial Remodeling/drug effects , Heart Diseases/chemically induced , Heart Diseases/physiopathology , Heart Diseases/diagnostic imaging , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/diagnostic imaging , Heart Atria/drug effects , Heart Atria/physiopathology , Heart Atria/diagnostic imaging , Stroke Volume/drug effects
3.
Article in English | MEDLINE | ID: mdl-38723703

ABSTRACT

Mollusks, including snails, possess two chambered hearts. The heart and cardiomyocytes of snails have many similarities with those of mammals. Also, the biophysics and pharmacology of Ca, K, and Na ion channels resemble. Similar to mammals, in mollusks, the ventricular cardiomyocytes and K channels are often studied, which are selectively sensitive to antagonists such as 4-AP, E-4031, and TEA. Since the physiological properties of the ventricular cardiac cells of snails are well characterized, the enzymatically dissociated atrial cardiomyocytes of Cornu aspersum (Müller, 1774) were studied using the whole-cell patch-clamp technique for detailed comparisons with mice, Mus musculus. The incubation of tissues in a solution simultaneously containing two enzymes, collagenase and papain, enabled the isolation of single cells. Recordings in the atrial cardiomyocytes of snails revealed outward K+ currents closely resembling those of the ventricle. The latter was consistent, whether the voltage ramp or steps and long or short pulses were used. Interestingly, under identical conditions, the current waveforms of atrial cardiomyocytes in snails were similar to those of mice left ventricles, albeit the kinetics and the absence of inward rectifier K channel (IK1) activation. Therefore, the heart of mollusks could be used as a simple and accessible experimental model, particularly for pharmacology and toxicology studies.


Subject(s)
Heart Atria , Heart Ventricles , Myocytes, Cardiac , Animals , Heart Atria/drug effects , Heart Atria/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/cytology , Mice , Patch-Clamp Techniques , Potassium Channels/metabolism , Snails
4.
Biomed Pharmacother ; 175: 116649, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692059

ABSTRACT

BACKGROUND: Second-generation antipsychotics increase the risk of atrial fibrillation. This study explores whether the atypical antipsychotic ziprasidone triggers inflammasome signaling, leading to atrial arrhythmia. METHODS: Electromechanical and pharmacological assessments were conducted on the rabbit left atria (LA). The patch-clamp technique was used to measure ionic channel currents in single cardiomyocytes. Detection of cytosolic reactive oxygen species production was performed in atrial cardiomyocytes. RESULTS: The duration of action potentials at 50 % and 90 % repolarization was dose-dependently shortened in ziprasidone-treated LA. Diastolic tension in LA increased after ziprasidone treatment. Ziprasidone-treated LA showed rapid atrial pacing (RAP) triggered activity. PI3K inhibitor, Akt inhibitor and mTOR inhibitor abolished the triggered activity elicited by ziprasidone in LA. The NLRP3 inhibitor MCC950 suppressed the ziprasidone-induced post-RAP-triggered activity. MCC950 treatment reduced the reverse-mode Na+/Ca2+ exchanger current in ziprasidone-treated myocytes. Cytosolic reactive oxygen species production decreased in ziprasidone-treated atrial myocytes after MCC950 treatment. Protein levels of inflammasomes and proinflammatory cytokines, including NLRP3, caspase-1, IL-1ß, IL-18, and IL-6 were observed to be upregulated in myocytes treated with ziprasidone. CONCLUSIONS: Our findings suggest ziprasidone induces atrial arrhythmia, potentially through upregulation of the NLRP3 inflammasome and enhancement of reactive oxygen species production via the PI3K/Akt/mTOR pathway.


Subject(s)
Atrial Fibrillation , Inflammasomes , Myocytes, Cardiac , Piperazines , Proto-Oncogene Proteins c-akt , Reactive Oxygen Species , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Atrial Fibrillation/chemically induced , Atrial Fibrillation/metabolism , TOR Serine-Threonine Kinases/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rabbits , Reactive Oxygen Species/metabolism , Piperazines/pharmacology , Male , Phosphatidylinositol 3-Kinases/metabolism , Thiazoles/pharmacology , Heart Atria/drug effects , Heart Atria/metabolism , Action Potentials/drug effects , Antipsychotic Agents/pharmacology
5.
Aging (Albany NY) ; 16(10): 8630-8644, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38775722

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) is often associated with atrial fibrosis and oxidative stress. Neferine, a bisbenzylisoquinoline alkaloid, has been reported to exert an antiarrhythmic effect. However, its impact on Angiotensin II (Ang II) infusion-induced AF and the underlying mechanism remains unclear. This study aimed to investigate whether neferine alleviates Ang II-induced AF and explore the underlying mechanisms. METHODS: Mice subjected to Ang II infusion to induce AF were concurrently treated with neferine or saline. AF incidence, myocardial cell size, fibrosis, and oxidative stress were then examined. RESULTS: Neferine treatment inhibited Ang II-induced AF, atrial size augmentation, and atrial fibrosis. Additionally, we observed that Ang II increased reactive oxygen species (ROS) generation, induced mitochondrial membrane potential depolarization, and reduced glutathione (GSH) and superoxide dismutase (SOD) levels, which were reversed to some extent by neferine. Mechanistically, neferine activated the Nrf2/HO-1 signaling pathway and inhibited TGF-ß/p-Smad2/3 in Ang II-infused atria. Zinc Protoporphyrin (ZnPP), an HO-1 inhibitor, reduced the anti-oxidative effect of neferine to some extent and subsequently abolished the beneficial effect of neferine on Ang II-induced AF. CONCLUSIONS: These findings provide hitherto undocumented evidence that the protective role of neferine in Ang II-induced AF is dependent on HO-1.


Subject(s)
Angiotensin II , Atrial Fibrillation , Benzylisoquinolines , Fibrosis , NF-E2-Related Factor 2 , Signal Transduction , Smad3 Protein , Transforming Growth Factor beta , Animals , Angiotensin II/pharmacology , Atrial Fibrillation/chemically induced , Atrial Fibrillation/metabolism , Atrial Fibrillation/prevention & control , NF-E2-Related Factor 2/metabolism , Mice , Benzylisoquinolines/pharmacology , Signal Transduction/drug effects , Smad3 Protein/metabolism , Male , Transforming Growth Factor beta/metabolism , Oxidative Stress/drug effects , Smad2 Protein/metabolism , Up-Regulation/drug effects , Mice, Inbred C57BL , Heart Atria/drug effects , Heart Atria/metabolism , Heart Atria/pathology , Heme Oxygenase (Decyclizing)/metabolism , Membrane Proteins , Heme Oxygenase-1
6.
PeerJ ; 12: e17434, 2024.
Article in English | MEDLINE | ID: mdl-38799057

ABSTRACT

We propose a new mouse (C57Bl6/J) model combining several features of heart failure with preserved ejection fraction encountered in older women, including hypertension from Angiotensin II infusion (AngII), menopause, and advanced age. To mimic menopause, we delayed ovariectomy (Ovx) at 12 months of age. We also studied the effects of AngII infusion for 28 days in younger animals and the impact of losing gonadal steroids earlier in life. We observed that AngII effects on heart morphology were different in younger and adult mice (3- and 12-month-old; 20 and 19% increase in heart weight. P < 0.01 for both) than in older animals (24-month-old; 6%; not significant). Ovariectomy at 12 months restored the hypertrophic response to AngII in elderly females (23%, p = 0.0001). We performed a bulk RNA sequencing study of the left ventricle (LV) and left atrial gene expression in elderly animals, controls, and Ovx. AngII modulated (|Log2 fold change| ≥ 1) the LV expression of 170 genes in control females and 179 in Ovx ones, 64 being shared. In the left atrium, AngII modulated 235 genes in control females and 453 in Ovx, 140 shared. We observed many upregulated genes associated with the extracellular matrix regulation in both heart chambers. Many of these upregulated genes were shared between the ventricle and the atrium as well as in control and Ovx animals, namely for the most expressed Ankrd1, Nppb, Col3a1, Col1a1, Ctgf Col8a1, and Cilp. Several circadian clock LV genes were modulated differently by AngII between control and Ovx females (Clock, Arntl, Per2, Cry2, and Ciart). In conclusion, sex hormones, even in elderly female mice, modulate the heart's hypertrophic response to AngII. Our study identifies potential new markers of hypertensive disease in aging female mice and possible disturbances of their cardiac circadian clock.


Subject(s)
Angiotensin II , Disease Models, Animal , Hypertension , Mice, Inbred C57BL , Ovariectomy , Animals , Female , Angiotensin II/pharmacology , Mice , Hypertension/physiopathology , Aging/physiology , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Menopause , Humans , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Heart Atria/physiopathology , Heart Atria/drug effects , Heart Atria/pathology , Collagen Type III
7.
Pharmacol Rep ; 76(3): 585-599, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38619735

ABSTRACT

BACKGROUND: Amiodarone (AMIO) is an antiarrhythmic drug with the pKa in the physiological range. Here, we explored how mild extracellular pH (pHe) changes shape the interaction of AMIO with atrial tissue and impact its pharmacological properties in the classical model of sea anemone sodium channel neurotoxin type 2 (ATX) induced late sodium current (INa-Late) and arrhythmias. METHOD: Isolated atrial cardiomyocytes from male Wistar rats and human embryonic kidney cells expressing SCN5A Na+ channels were used for patch-clamp experiments. Isolated right atria (RA) and left atria (LA) tissue were used for bath organ experiments. RESULTS: A more acidophilic pHe caused negative inotropic effects on isolated RA and LA atrial tissue, without modification of the pharmacological properties of AMIO. A pHe of 7.0 changed the sodium current (INa) related components of the action potential (AP), which was enhanced in the presence of AMIO. ATXinduced arrhythmias in isolated RA and LA. Also, ATX prolonged the AP duration and enhanced repolarization dispersion in isolated cardiomyocytes in both pHe 7.4 and pHe 7.0. Pre-incubation of the isolated RA and LA and isolated atrial cardiomyocytes with AMIO prevented arrhythmias induced by ATX only at a pHe of 7.0. Moreover, AMIO was able to block INa-Late induced by ATX only at a pHe of 7.0. CONCLUSION: The pharmacological properties of AMIO concerning healthy rat atrial tissue are not dependent on pHe. However, the prevention of arrhythmias induced by INa-Late is pHe-dependent. The development of drugs analogous to AMIO with charge stabilization may help to create more effective drugs to treat arrhythmias related to the INa-Late.


Subject(s)
Action Potentials , Amiodarone , Anti-Arrhythmia Agents , Arrhythmias, Cardiac , Heart Atria , Myocytes, Cardiac , Rats, Wistar , Animals , Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Male , Humans , Rats , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Action Potentials/drug effects , Heart Atria/drug effects , Heart Atria/metabolism , Hydrogen-Ion Concentration , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/chemically induced , NAV1.5 Voltage-Gated Sodium Channel/metabolism , HEK293 Cells , Sodium/metabolism , Patch-Clamp Techniques , Cnidarian Venoms/pharmacology
8.
J Cell Physiol ; 239(5): e31248, 2024 May.
Article in English | MEDLINE | ID: mdl-38501506

ABSTRACT

The loss of semaphorin 3A (Sema3A), which is related to endothelial-to-mesenchymal transition (EndMT) in atrial fibrosis, is implicated in the pathogenesis of atrial fibrillation (AF). To explore the mechanisms by which EndMT affects atrial fibrosis and assess the potential of a Sema3A activator (naringin) to prevent atrial fibrosis by targeting transforming growth factor-beta (TGF-ß)-induced EndMT, we used human atria, isolated human atrial endocardial endothelial cells (AEECs), and used transgenic mice expressing TGF-ß specifically in cardiac tissues (TGF-ß transgenic mice). We evaluated an EndMT marker (Twist), a proliferation marker (proliferating cell nuclear antigen; PCNA), and an endothelial cell (EC) marker (CD31) through triple immunohistochemistry and confirmed that both EndMT and EC proliferation contribute to atrial endocardial fibrosis during AF in TGF-ß transgenic mice and AF patient tissue sections. Additionally, we investigated the impact of naringin on EndMT and EC proliferation in AEECs and atrial fibroblasts. Naringin exhibited an antiproliferative effect, to which AEECs were more responsive. Subsequently, we downregulated Sema3A in AEECs using small interfering RNA to clarify a correlation between the reduction in Sema3A and the elevation of EndMT markers. Naringin treatment induced the expression of Sema3A and a concurrent decrease in EndMT markers. Furthermore, naringin administration ameliorated AF and endocardial fibrosis in TGF-ß transgenic mice by stimulating Sema3A expression, inhibiting EndMT markers, reducing atrial fibrosis, and lowering AF vulnerability. This suggests therapeutic potential for naringin in AF treatment.


Subject(s)
Atrial Fibrillation , Cell Proliferation , Endothelial Cells , Epithelial-Mesenchymal Transition , Flavanones , Heart Atria , Semaphorin-3A , Transforming Growth Factor beta , Animals , Humans , Male , Mice , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Atrial Fibrillation/genetics , Atrial Fibrillation/drug therapy , Cell Proliferation/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Flavanones/pharmacology , Heart Atria/metabolism , Heart Atria/drug effects , Heart Atria/pathology , Mice, Transgenic , Semaphorin-3A/metabolism , Semaphorin-3A/genetics , Transforming Growth Factor beta/metabolism
10.
Prostaglandins Other Lipid Mediat ; 172: 106834, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38521490

ABSTRACT

Oxysterol, 25-hydroxycholesterol (25HC), is a potent regulator of immune reactions, its synthesis greatly increases by macrophages during inflammation. We hypothesize that 25HC can have cardioprotective effects by limiting consequences of excessive ß-adrenoceptor (ßAR) stimulation, particularly reactive oxygen species (ROS) production, in mouse atria. Isoproterenol, a ßAR agonist, increased extra- and intracellular levels of ROS. This enhancement of ROS production was suppressed by NADPH oxidase antagonists as well as 25HC. Inhibition of ß3ARs, Gi protein and protein kinase Cε prevented the effect of 25HC on isoproterenol-dependent ROS synthesis. Furthermore, 25HC suppressed isoproterenol-induced lipid peroxidation and mitochondrial ROS generation as well as ROS-dependent component of positive inotropic response to isoproterenol. Additionally, 25HC decreased mitochondrial ROS production and lipid peroxidation induced by antimycin A, a mitochondrial poison. Thus, 25HC exerts antioxidant properties alleviating mitochondrial dysfunction-induced and ßAR-dependent cardiac oxidative damage. In the latter case, 25HC can act via signaling mechanism engaging ß3ARs, Gi protein and protein kinase Cε.


Subject(s)
Antioxidants , Heart Atria , Hydroxycholesterols , Reactive Oxygen Species , Signal Transduction , Animals , Hydroxycholesterols/pharmacology , Hydroxycholesterols/metabolism , Mice , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Heart Atria/metabolism , Heart Atria/drug effects , Antioxidants/pharmacology , Antioxidants/metabolism , Male , Lipid Peroxidation/drug effects , Isoproterenol/pharmacology , Mice, Inbred C57BL
11.
J Cardiovasc Pharmacol ; 83(6): 621-634, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38547520

ABSTRACT

ABSTRACT: Type 2 diabetes mellitus increases the risk of cardiovascular diseases. Therefore, elucidation of the cardiovascular effects of antidiabetics is crucial. Incretin-based therapies are increasingly used for type 2 diabetes mellitus treatment as monotherapy and in combination. We aimed to study the effects of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) and sitagliptin on beating rates in isolated atria from diabetic rats. The chronotropic responses to GLP-1 RAs and sitagliptin as monotherapy and in combinations with metformin, pioglitazone, and glimepiride in isolated atria from control and diabetic rats were determined. GLP-1 (7-36), GLP-1 (9-36), and exendin-4 (1-39) produced increases in beating rates in both control and diabetic rat atria. However, sitagliptin increased the beating frequency only in the diabetic group. Exendin (9-39), nitro- l -arginine methyl ester hydrochloride, and indomethacin blocked responses to GLP-1 RAs but not the response to sitagliptin. Glibenclamide, 4-aminopyridine, apamin, charybdotoxin, superoxide dismutase, and catalase incubations did not change responses to GLP-1 RAs and sitagliptin. GLP-1 RAs increase beating rates in isolated rat atrium through GLP-1 receptor, nitric oxide, and cyclooxygenase pathways but not potassium channels and reactive oxygen radicals.


Subject(s)
Diabetes Mellitus, Experimental , Glucagon-Like Peptide-1 Receptor , Heart Atria , Heart Rate , Hypoglycemic Agents , Sitagliptin Phosphate , Animals , Sitagliptin Phosphate/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Male , Heart Atria/drug effects , Heart Atria/physiopathology , Heart Atria/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/physiopathology , Heart Rate/drug effects , Hypoglycemic Agents/pharmacology , Rats , Rats, Wistar , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/metabolism , Exenatide/pharmacology , Incretins/pharmacology , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide 1/metabolism , Pyrazines/pharmacology , Glucagon-Like Peptide-1 Receptor Agonists
12.
Hypertens Res ; 47(5): 1309-1322, 2024 May.
Article in English | MEDLINE | ID: mdl-38374239

ABSTRACT

Atrial fibrillation (AF), the most common cardiac arrhythmia, is an important contributor to mortality and morbidity. Ubquitin-specific protease 7 (USP7), one of the most abundant ubiquitin-specific proteases (USP), participated in many cellular events, such as cell proliferation, apoptosis, and tumourigenesis. However, its role in AF remains unknown. Here, the mice were treated with Ang II infusion to induce the AF model. Echocardiography was used to measure the atrial diameter. Electrical stimulation was programmed to measure the induction and duration of AF. The changes in atrial remodeling were measured using routine histologic analysis. Here, a significant increase in USP7 expression was observed in Ang II-stimulated atrial cardiomyocytes and atrial tissues, as well as in atrial tissues from patients with AF. The administration of p22077, the inhibitor of USP7, attenuated Ang II-induced inducibility and duration of AF, atrial dilatation, connexin dysfunction, atrial fibrosis, atrial inflammation, and atrial oxidase stress, and then inhibited the progression of AF. Mechanistically, the administration of p22077 alleviated Ang II-induced activation of TGF-ß/Smad2, NF-κB/NLRP3, NADPH oxidases (NOX2 and NOX4) signals, the up-regulation of CX43, ox-CaMKII, CaMKII, Kir2.1, and down-regulation of SERCA2a. Together, this study, for the first time, suggests that USP7 is a critical driver of AF and revealing USP7 may present a new target for atrial fibrillation therapeutic strategies.


Subject(s)
Angiotensin II , Atrial Fibrillation , Ubiquitin-Specific Peptidase 7 , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/chemically induced , Atrial Fibrillation/drug therapy , Atrial Fibrillation/prevention & control , Ubiquitin-Specific Peptidase 7/metabolism , Mice , Male , Mice, Inbred C57BL , Heart Atria/drug effects , Heart Atria/metabolism , Heart Atria/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Humans , Atrial Remodeling/drug effects
13.
Eur Heart J Acute Cardiovasc Care ; 13(5): 414-422, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38372622

ABSTRACT

AIMS: A recently published trial has shown no differences in outcomes between patients with new-onset supraventricular arrhythmia (SVA) in septic shock treated with either propafenone or amiodarone. However, these outcome data have not been evaluated in relation to the presence or absence of a dilated left atrium (LA). METHODS AND RESULTS: Patients with SVA and a left ventricular ejection fraction ≥ 35% were randomized to receive intravenous propafenone (70 mg bolus followed by 400-840 mg/24 h) or amiodarone (300 mg bolus followed by 600-1800 mg/24 h). They were divided into groups based on whether their end-systolic left atrial volume (LAVI) was ≥40 mL/m². The subgroup outcomes assessed were survival at ICU discharge, 1 month, 3 months, 6 months, and 12 months. Propafenone cardioverted earlier (P = 0.009) and with fewer recurrences (P = 0.001) in the patients without LA enlargement (n = 133). Patients with LAVI < 40 mL/m2 demonstrated a mortality benefit of propafenone over the follow-up of 1 year [Cox regression, hazard ratio (HR) 0.6 (95% CI 0.4; 0.9), P = 0.014]. Patients with dilated LA (n = 37) achieved rhythm control earlier in amiodarone (P = 0.05) with similar rates of recurrences (P = 0.5) compared to propafenone. The outcomes for patients with LAVI ≥ 40 mL/m2 were less favourable with propafenone compared to amiodarone at 1 month [HR 3.6 (95% CI 1.03; 12.5), P = 0.045]; however, it did not reach statistical significance at 1 year [HR 1.9 (95% CI 0.8; 4.4), P = 0.138]. CONCLUSION: Patients with non-dilated LA who achieved rhythm control with propafenone in the setting of septic shock had better short-term and long-term outcomes than those treated with amiodarone, which seemed to be more effective in patients with LAVI ≥ 40 mL/m². TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT03029169, registered on 24 January 2017.


Subject(s)
Amiodarone , Anti-Arrhythmia Agents , Heart Atria , Propafenone , Shock, Septic , Tachycardia, Supraventricular , Humans , Propafenone/therapeutic use , Propafenone/administration & dosage , Amiodarone/therapeutic use , Amiodarone/administration & dosage , Shock, Septic/drug therapy , Shock, Septic/physiopathology , Male , Female , Anti-Arrhythmia Agents/therapeutic use , Anti-Arrhythmia Agents/administration & dosage , Aged , Heart Atria/physiopathology , Heart Atria/diagnostic imaging , Heart Atria/drug effects , Tachycardia, Supraventricular/drug therapy , Tachycardia, Supraventricular/physiopathology , Treatment Outcome , Middle Aged , Stroke Volume/physiology , Stroke Volume/drug effects
14.
Transl Res ; 268: 1-12, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38244770

ABSTRACT

Interleukin (IL)-33, a cytokine involved in immune responses, can activate its receptor, suppression of tumorigenicity 2 (ST2), is elevated during atrial fibrillation (AF). However, the role of IL-33/ST2 signaling in atrial arrhythmia is unclear. This study explored the pathological effects of the IL-33/ST2 axis on atrial remodeling and arrhythmogenesis. Patch clamping, confocal microscopy, and Western blotting were used to analyze the electrical characteristics of and protein activity in atrial myocytes (HL-1) treated with recombinant IL-33 protein and/or ST2-neutralizing antibodies for 48 hrs. Telemetric electrocardiographic recordings, Masson's trichrome staining, and immunohistochemistry staining of the atrium were performed in mice receiving tail vein injections with nonspecific immunoglobulin (control), IL-33, and IL-33 combined with anti-ST2 antibody for 2 weeks. IL-33-treated HL-1 cells had a reduced action potential duration, lower L-type Ca2+ current, greater sarcoplasmic reticulum (SR) Ca2+ content, increased Na+/Ca2+ exchanger (NCX) current, elevation of K+ currents, and increased intracellular calcium transient. IL-33-treated HL-1 myocytes had greater activation of the calcium-calmodulin-dependent protein kinase II (CaMKII)/ryanodine receptor 2 (RyR2) axis and nuclear factor kappa B (NF-κB) / NLR family pyrin domain containing 3 (NLRP3) signaling than did control cells. IL-33 treated cells also had greater expression of Nav1.5, Kv1.5, NCX, and NLRP3 than did control cells. Pretreatment with neutralizing anti-ST2 antibody attenuated IL-33-mediated activation of CaMKII/RyR2 and NF-κB/NLRP3 signaling. IL-33-injected mice had more atrial ectopic beats and increased AF episodes, greater atrial fibrosis, and elevation of NF-κB/NLRP3 signaling than did controls or mice treated with IL-33 combined with anti-ST2 antibody. Thus, IL-33 recombinant protein treatment promotes atrial remodeling through ST2 signaling. Blocking the IL-33/ST2 axis might be an innovative therapeutic approach for patients with atrial arrhythmia and elevated serum IL-33.


Subject(s)
Atrial Remodeling , Interleukin-33 , Myocytes, Cardiac , Animals , Male , Mice , Action Potentials/drug effects , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/metabolism , Atrial Fibrillation/physiopathology , Atrial Fibrillation/metabolism , Atrial Remodeling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line , Heart Atria/physiopathology , Heart Atria/metabolism , Heart Atria/drug effects , Heart Atria/pathology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction
15.
Heart Rhythm ; 21(5): 622-629, 2024 May.
Article in English | MEDLINE | ID: mdl-38280622

ABSTRACT

BACKGROUND: More than 50% of patients with atrial fibrillation (AF) suffer from sleep disordered breathing (SDB). Obstructive respiratory events contribute to a transient, vagally mediated atrial arrhythmogenic substrate, which is resistant to most available antiarrhythmic drugs. OBJECTIVE: The purpose of this study was to investigate the effect of pharmacologic inhibition of the G-protein-gated acetylcholine-regulated potassium current (IK,ACh) with and without acute autonomic nervous system activation by nicotine in a pig model for obstructive respiratory events. METHODS: In 21 pigs, SDB was simulated by applying an intermittent negative upper airway pressure (INAP). AF inducibility and atrial effective refractory periods (aERPs) were determined before and during INAP by an S1S2 atrial pacing-protocol. Pigs were randomized into 3 groups-group 1: vehicle (n = 4); group 2: XAF-1407 (IK,ACh inhibitor) (n = 7); and group 3: nicotine followed by XAF-1407 (n = 10). RESULTS: In group 1, INAP shortened aERP (ΔaERP -42.6 ms; P = .004) and transiently increased AF inducibility from 0% to 31%. In group 2, XAF-1407 prolonged aERP by 25.2 ms (P = .005) during normal breathing and prevented INAP-induced aERP shortening (ΔaERP -3.6 ms; P = .3) and AF inducibility. In group 3, INAP transiently shortened aERP during nicotine perfusion (ΔaERP -33.6 ms; P = .004) and increased AF inducibility up to 61%, which both were prevented by XAF-1407. CONCLUSION: Simulated obstructive respiratory events transiently shorten aERP and increase AF inducibility, which can be prevented by the IK,ACh-inhibitor XAF-1407. XAF-1407 also prevents these arrhythmogenic changes induced by obstructive respiratory events during nicotine perfusion. Whether IK,ACh channels represent a target for SDB-related AF in humans warrants further study.


Subject(s)
Atrial Fibrillation , Disease Models, Animal , Animals , Swine , Atrial Fibrillation/physiopathology , Atrial Fibrillation/etiology , Atrial Fibrillation/prevention & control , Heart Atria/physiopathology , Heart Atria/drug effects , Heart Atria/metabolism , Acetylcholine/pharmacology , Nicotine/pharmacology , Sleep Apnea Syndromes/physiopathology , Sleep Apnea Syndromes/complications
16.
J Mol Cell Cardiol ; 183: 70-80, 2023 10.
Article in English | MEDLINE | ID: mdl-37704101

ABSTRACT

BACKGROUND: The small conductance Ca2+-activated K+ current (ISK) is a potential therapeutic target for treating atrial fibrillation. AIM: To clarify, in rabbit and human atrial cardiomyocytes, the intracellular [Ca2+]-sensitivity of ISK, and its contribution to action potential (AP) repolarisation, under physiological conditions. METHODS: Whole-cell-patch clamp, fluorescence microscopy: to record ion currents, APs and [Ca2+]i; 35-37°C. RESULTS: In rabbit atrial myocytes, 0.5 mM Ba2+ (positive control) significantly decreased whole-cell current, from -12.8 to -4.9 pA/pF (P < 0.05, n = 17 cells, 8 rabbits). By contrast, the ISK blocker apamin (100 nM) had no effect on whole-cell current, at any set [Ca2+]i (∼100-450 nM). The ISK blocker ICAGEN (1 µM: ≥2 x IC50) also had no effect on current over this [Ca2+]i range. In human atrial myocytes, neither 1 µM ICAGEN (at [Ca2+]i âˆ¼ 100-450 nM), nor 100 nM apamin ([Ca2+]i âˆ¼ 250 nM) affected whole-cell current (5-10 cells, 3-5 patients/group). APs were significantly prolonged (at APD30 and APD70) by 2 mM 4-aminopyridine (positive control) in rabbit atrial myocytes, but 1 µM ICAGEN had no effect on APDs, versus either pre-ICAGEN or time-matched controls. High concentration (10 µM) ICAGEN (potentially ISK-non-selective) moderately increased APD70 and APD90, by 5 and 26 ms, respectively. In human atrial myocytes, 1 µM ICAGEN had no effect on APD30-90, whether stimulated at 1, 2 or 3 Hz (6-9 cells, 2-4 patients/rate). CONCLUSION: ISK does not flow in human or rabbit atrial cardiomyocytes with [Ca2+]i set within the global average diastolic-systolic range, nor during APs stimulated at physiological or supra-physiological (≤3 Hz) rates.


Subject(s)
Atrial Fibrillation , Myocytes, Cardiac , Animals , Humans , Rabbits , Myocytes, Cardiac/drug effects , Apamin/pharmacology , Small-Conductance Calcium-Activated Potassium Channels , Heart Atria/drug effects , Action Potentials/drug effects
17.
Int J Mol Sci ; 24(5)2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36902177

ABSTRACT

Ergotamine (2'-methyl-5'α-benzyl-12'-hydroxy-3',6',18-trioxoergotaman) is a tryptamine-related alkaloid from the fungus Claviceps purpurea. Ergotamine is used to treat migraine. Ergotamine can bind to and activate several types of 5-HT1-serotonin receptors. Based on the structural formula of ergotamine, we hypothesized that ergotamine might stimulate 5-HT4-serotonin receptors or H2-histamine receptors in the human heart. We observed that ergotamine exerted concentration- and time-dependent positive inotropic effects in isolated left atrial preparations in H2-TG (mouse which exhibits cardiac-specific overexpression of the human H2-histamine receptor). Similarly, ergotamine increased force of contraction in left atrial preparations from 5-HT4-TG (mouse which exhibits cardiac-specific overexpression of the human 5-HT4-serotonin receptor). An amount of 10 µM ergotamine increased the left ventricular force of contraction in isolated retrogradely perfused spontaneously beating heart preparations of both 5-HT4-TG and H2-TG. In the presence of the phosphodiesterase inhibitor cilostamide (1 µM), ergotamine 10 µM exerted positive inotropic effects in isolated electrically stimulated human right atrial preparations, obtained during cardiac surgery, that were attenuated by 10 µM of the H2-histamine receptor antagonist cimetidine, but not by 10 µM of the 5-HT4-serotonin receptor antagonist tropisetron. These data suggest that ergotamine is in principle an agonist at human 5-HT4-serotonin receptors as well at human H2-histamine receptors. Ergotamine acts as an agonist on H2-histamine receptors in the human atrium.


Subject(s)
Ergotamine , Heart Atria , Receptors, Histamine H4 , Receptors, Serotonin, 5-HT4 , Serotonin 5-HT4 Receptor Agonists , Animals , Humans , Mice , Ergotamine/pharmacology , Heart Atria/drug effects , Heart Atria/metabolism , Myocardial Contraction/drug effects , Receptors, Histamine/metabolism , Receptors, Serotonin, 5-HT4/metabolism , Serotonin 5-HT4 Receptor Agonists/pharmacology , Receptors, Histamine H4/agonists
18.
Int Heart J ; 63(2): 347-355, 2022 Mar 30.
Article in English | MEDLINE | ID: mdl-35296611

ABSTRACT

Atrial inflammation and fibrosis have long been considered culprits in the development of atrial fibrillation (AF). Prior clinical studies showed that corticosteroid therapy is beneficial in patients with AF. Here we sought to determine whether prednisone treatment prevents atrial tachypacing (ATP) induced atrial fibrosis.Dogs were randomized into the sham, ATP, ATP + low-dose prednisone (ALP), and ATP + high-dose prednisone (AHP) groups. After 6 days of recovery from surgery, dogs were subjected to ATP at 400 beats per minute for 4 weeks while being treated with prednisone (15 or 40 mg/day) or a placebo. Pacemakers were not activated in the sham group.Compared with the ATP group, the expression of collagen I, collagen III, α-smooth muscle actin, transforming growth factor-ß1 and connective tissue growth factor were significantly reduced in the ALP and AHP groups. Fluorescence assays showed that reactive oxygen species formation in the right atrium was suppressed in the ALP and AHP groups compared with the ATP group. The protein level of NADPH oxidase 2 was reduced in the ALP and AHP groups' versus ATP group, while NOX4 and NOX5 were unchanged. ATP-induced downregulation of BH4 and eNOS uncoupling in the atria was partially restored in the prednisone-treated groups.Our study demonstrated that atrial fibrosis induced by ATP were suppressed by prednisone. Low-dose prednisone was also effective in suppressing the development of atrial fibrosis.


Subject(s)
Dog Diseases , Heart Atria , Inflammation , Prednisone , Animals , Atrial Fibrillation/complications , Atrial Fibrillation/veterinary , Dog Diseases/drug therapy , Dogs , Fibrosis , Heart Atria/drug effects , Heart Atria/pathology , Inflammation/drug therapy , Inflammation/veterinary , Prednisone/pharmacology , Prednisone/therapeutic use , Treatment Outcome
19.
BMC Cardiovasc Disord ; 22(1): 63, 2022 02 20.
Article in English | MEDLINE | ID: mdl-35184743

ABSTRACT

BACKGROUND: Primary malignant tumors of the heart are rare. Although preoperative histological diagnosis is difficult, it has paramount value in therapeutic strategy development and prognostic estimation. Herein, we reported 2 cases of intracardiac tumors. CASES PRESENTATION: Both patients presented to the hospital with heart-related symptoms. Echocardiography showed massive masses in the atrium and positron emission tomography-computed tomography (PET/CT) revealed hypermetabolism and invasiveness. One patient cannot take surgery due to extensive metastasis and poor condition. The other patient was primarily diagnosed with lymphoma, and surgery was not recommended. They successfully underwent intravenous atrial biopsy, and histological samples confirmed intimal sarcoma and diffuse large B cell lymphoma. Based on immunohistochemical and molecular assessments, targeted chemotherapy was administered, resulting in clinical and imaging remission at discharge. CONCLUSIONS: Percutaneous intravenous catheter biopsy as a safe invasive test provides an accurate pathological diagnosis after imaging evaluation, and offers a therapeutic direction. Nonmalignant masses and some chemo-radiosensitive malignant tumors in the atrium could have good prognosis after targeted therapy.


Subject(s)
Catheterization, Peripheral/instrumentation , Heart Atria/pathology , Heart Neoplasms/pathology , Lymphoma, Large B-Cell, Diffuse/pathology , Sarcoma/pathology , Surgical Instruments , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy , Female , Heart Atria/diagnostic imaging , Heart Atria/drug effects , Heart Neoplasms/diagnostic imaging , Heart Neoplasms/drug therapy , Humans , Lymphoma, Large B-Cell, Diffuse/diagnostic imaging , Lymphoma, Large B-Cell, Diffuse/drug therapy , Magnetic Resonance Imaging , Male , Middle Aged , Molecular Targeted Therapy , Neoplasm Staging , Positron Emission Tomography Computed Tomography , Sarcoma/diagnostic imaging , Sarcoma/drug therapy , Treatment Outcome
20.
Cardiovasc Res ; 118(1): 184-195, 2022 01 07.
Article in English | MEDLINE | ID: mdl-33098411

ABSTRACT

AIMS: Systemic inflammation and increased activity of atrial NOX2-containing NADPH oxidases have been associated with the new onset of atrial fibrillation (AF) after cardiac surgery. In addition to lowering LDL-cholesterol, statins exert rapid anti-inflammatory and antioxidant effects, the clinical significance of which remains controversial. METHODS AND RESULTS: We first assessed the impact of cardiac surgery and cardiopulmonary bypass (CPB) on atrial nitroso-redox balance by measuring NO synthase (NOS) and GTP cyclohydrolase-1 (GCH-1) activity, biopterin content, and superoxide production in paired samples of the right atrial appendage obtained before (PRE) and after CPB and reperfusion (POST) in 116 patients. The effect of perioperative treatment with atorvastatin (80 mg once daily) on these parameters, blood biomarkers, and the post-operative atrial effective refractory period (AERP) was then evaluated in a randomized, double-blind, placebo-controlled study in 80 patients undergoing cardiac surgery on CPB. CPB and reperfusion led to a significant increase in atrial superoxide production (74% CI 71-76%, n = 46 paired samples, P < 0.0001) and a reduction in atrial tetrahydrobiopterin (BH4) (34% CI 33-35%, n = 36 paired samples, P < 0.01), and in GCH-1 (56% CI 55-58%, n = 26 paired samples, P < 0.001) and NOS activity (58% CI 52-67%, n = 20 paired samples, P < 0.001). Perioperative atorvastatin treatment prevented the effect of CPB and reperfusion on all parameters but had no significant effect on the postoperative right AERP, troponin release, or NT-proBNP after cardiac surgery. CONCLUSION: Perioperative statin therapy prevents post-reperfusion atrial nitroso-redox imbalance in patients undergoing on-pump cardiac surgery but has no significant impact on postoperative atrial refractoriness, perioperative myocardial injury, or markers of postoperative LV function. CLINICAL TRIAL REGISTRATION: https://clinicaltrials.gov/ct2/show/NCT01780740.


Subject(s)
Atorvastatin/therapeutic use , Atrial Fibrillation/prevention & control , Atrial Function, Right/drug effects , Cardiac Surgical Procedures/adverse effects , Cardiopulmonary Bypass/adverse effects , Heart Atria/drug effects , Nitroso Compounds/metabolism , Refractory Period, Electrophysiological/drug effects , Action Potentials/drug effects , Atorvastatin/adverse effects , Atrial Fibrillation/diagnosis , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Biopterins/analogs & derivatives , Biopterins/metabolism , Double-Blind Method , England , Heart Atria/metabolism , Heart Atria/physiopathology , Heart Rate/drug effects , Humans , NADPH Oxidases/metabolism , Nitric Oxide Synthase/metabolism , Oxidation-Reduction , Superoxides/metabolism , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...