Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.651
Filtrer
1.
PLoS One ; 19(7): e0306323, 2024.
Article de Anglais | MEDLINE | ID: mdl-38976680

RÉSUMÉ

Left atrial strain (LAS) measured by two-dimensional speckle tracking echocardiography (2DSTE) is considered to be a marker of LA structural remodeling, but it remains unsettled. We investigated the potential usefulness and clinical relevance of LAS to detect atrial remodeling including fibrosis by analyzing gene expression in cardiovascular surgery patients. Preoperative 2DSTE was performed in 131 patients (92 patients with sinus rhythm [SR] patients including paroxysmal AF [PAF], 39 atrial fibrillation [AF]) undergoing cardiovascular surgery. Atrial samples were obtained from the left atrial appendages, and mRNA expression level was analyzed by real-time reverse transcription polymerase chain reaction (RT-PCR) in 59 cases (24 PAF, 35 AF). Mean value of left atrial reservoir strain (mLASr) correlated with left atrial volume index (LAVI), and left atrial conduit strain (mLAScd). mLASr also correlated with left atrial contractile strain (mLASct) in SR patients including PAF. mLASr was significantly lower, and LAVI was higher, in the AF group, compared with SR patients including PAF. The expression of COL1A1 mRNA encoding collagen type I α1 significantly increased in AF patients (p = 0.031). mLASr negatively correlated with COL1A1 expression level, and multivariate regression analysis showed that mLASr was an independent predictor of atrial COL1A1 expression level, even after adjusting for age, sex, and BMI. But, neither mLAScd / mLASct nor LAVI (bp) correlated with COL1A1 gene expression. The expression level of COL1A1 mRNA strongly correlated with ECM-related genes (COL3A1, FN1). It also correlated ECM degradation-related genes (MMP2, TIMP1, and TIMP2), pro-fibrogenic cytokines (TGFB1 encoding TGFß1, END1, PDGFD, CTGF), oxidant stress-related genes (NOX2, NOX4), ACE, inflammation-related genes (NLRP, IL1B, MCP-1), and apoptosis (BAX). Among the fibrosis-related genes examined, univariable regression analysis showed that log (COL1A1) was associated with log (TGFB1) (adjusted R2 = 0.685, p<0.001), log (NOX4) (adjusted R2 = 0.622, p<0.001), log (NOX2) (adjusted R2 = 0.611, p<0.001), suggesting that TGFB1 and NOX4 was the potent independent determinants of COL1A1 expression level. mLASr negatively correlated with the ECM-related genes, and fibrosis-related gene expression level including TGFB1, NOX2, and NLRP3 in PAF patients. PAF patients with low mLASr had higher expression of the fibrosis-related gene expression, compared with those with high mLASr. These results suggest that LASr correlates with atrial COL1A1 gene expression associated with fibrosis-related gene expression. Patients with low LASr exhibit increased atrial fibrosis-related gene expression, even those with PAF, highlighting the utility of LAS as a marker for LA fibrosis in cardiovascular surgery patients.


Sujet(s)
Fibrillation auriculaire , Remodelage auriculaire , Fibrose , Atrium du coeur , Humains , Mâle , Femelle , Remodelage auriculaire/génétique , Sujet âgé , Adulte d'âge moyen , Atrium du coeur/métabolisme , Atrium du coeur/anatomopathologie , Fibrillation auriculaire/génétique , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/chirurgie , Fibrillation auriculaire/anatomopathologie , Fibrillation auriculaire/physiopathologie , Collagène de type I/génétique , Collagène de type I/métabolisme , Échocardiographie , Chaine alpha-1 du collagène de type I , Marqueurs biologiques/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Fonction auriculaire gauche
3.
Circ Arrhythm Electrophysiol ; 17(7): e012452, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39012929

RÉSUMÉ

BACKGROUND: Aging is one of the most potent risk determinants for the onset of atrial fibrillation (AF). Sirts (sirtuins) have been implicated in the pathogenesis of cardiovascular disease, and their expression declines with aging. However, whether Sirts involved in age-related AF and its underlying mechanisms remain unknown. The present study aims to explore the role of Sirts in age-related AF and delineate the underlying molecular mechanisms. METHODS: Sirt1 levels in the atria of both elderly individuals and aging rats were evaluated using quantitative real-time polymerase chain reaction and Western blot analysis. Mice were engineered to specifically knockout Sirt1 in the atria and right ventricle (Sirt1mef2c/mef2c). Various techniques, such as echocardiography, atrial electrophysiology, and protein acetylation modification omics were employed. Additionally, coimmunoprecipitation was utilized to substantiate the interaction between Sirt1 and RIPK1 (receptor-interacting protein kinase 1). RESULTS: We discerned that among the diverse subtypes of sirtuin proteins, only Sirt1 expression was significantly diminished in the atria of elderly people and aged rats. The Sirt1mef2c/mef2c mice exhibited an enlarged atrial diameter and heightened vulnerability to AF. Acetylated proteomics and cell experiments identified that Sirt1 deficiency activated atrial necroptosis through increasing RIPK1 acetylation and subsequent pseudokinase MLKL (mixed lineage kinase domain-like protein) phosphorylation. Consistently, necroptotic inhibitor necrosulfonamide mitigated atrial necroptosis and diminished both the atrial diameter and AF susceptibility of Sirt1mef2c/mef2c mice. Resveratrol prevented age-related AF in rats by activating atrial Sirt1 and inhibiting necroptosis. CONCLUSIONS: Our findings first demonstrated that Sirt1 exerts significant efficacy in countering age-related AF by impeding atrial necroptosis through regulation of RIPK1 acetylation, highlighting that the activation of Sirt1 or the inhibition of necroptosis could potentially serve as a therapeutic strategy for age-related AF.


Sujet(s)
Fibrillation auriculaire , Modèles animaux de maladie humaine , Atrium du coeur , Souris knockout , Nécroptose , Receptor-Interacting Protein Serine-Threonine Kinases , Sirtuine-1 , Animaux , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Acétylation , Atrium du coeur/métabolisme , Atrium du coeur/anatomopathologie , Atrium du coeur/physiopathologie , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Receptor-Interacting Protein Serine-Threonine Kinases/génétique , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/génétique , Fibrillation auriculaire/physiopathologie , Fibrillation auriculaire/prévention et contrôle , Fibrillation auriculaire/anatomopathologie , Mâle , Humains , Rats , Vieillissement/métabolisme , Vieillissement/anatomopathologie , Souris , Rat Sprague-Dawley , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Transduction du signal , Facteurs âges , Sujet âgé , Souris de lignée C57BL , Femelle
4.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167274, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38838411

RÉSUMÉ

This study aims to investigate the role of claudin-5 (Cldn5) in cardiac structural integrity. Proteomic analysis was performed to screen the protein profiles in enlarged left atrium from atrial fibrillation (AF) patients. Cldn5 shRNA adeno-associated virus (AAV) or siRNA was injected into the mouse left ventricle or added into HL1 cells respectively to knockdown Cldn5 in cardiomyocytes to observe whether the change of Cldn5 influences cardiac morphology and function, and affects those protein expressions stem from the proteomic analysis. Mitochondrial density and membrane potential were also measured by Mitotracker staining and JC-1 staining under the confocal microscope in HL1 cells. Cldn5 was reduced in cardiomyocytes from the left atrial appendage of AF patients compared to non-AF donors. Proteomic analysis showed 83 proteins were less abundant and 102 proteins were more abundant in AF patients. KEGG pathway analysis showed less abundant CACNA2D2, CACNB2, MYL2 and MAP6 were highly associated with dilated cardiomyopathy. Cldn5 shRNA AAV injection caused severe cardiac atrophy, dilation and myocardial dysfunction in mice. The decreases in mitochondrial numbers and mitochondrial membrane potentials in HL1 cells were observed after Cldn5 knockdown. We demonstrated for the first time the mechanism of Cldn5 downregulation-induced myocyte atrophy and myocardial dysfunction might be associated with the downregulation of CACNA2D2, CACNB2, MYL2 and MAP6, and mitochondrial dysfunction in cardiomyocytes.


Sujet(s)
Fibrillation auriculaire , Claudine-5 , Myocytes cardiaques , Animaux , Femelle , Humains , Mâle , Souris , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/anatomopathologie , Fibrillation auriculaire/génétique , Lignée cellulaire , Claudine-5/métabolisme , Claudine-5/génétique , Potentiel de membrane mitochondriale/génétique , Souris de lignée C57BL , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Protéomique/méthodes
5.
Cell Mol Life Sci ; 81(1): 264, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38878214

RÉSUMÉ

Atrial fibrillation (AF) is the most common arrhythmia, and atrial fibrosis is a pathological hallmark of structural remodeling in AF. Prostaglandin I2 (PGI2) can prevent the process of fibrosis in various tissues via cell surface Prostaglandin I2 receptor (IP). However, the role of PGI2 in AF and atrial fibrosis remains unclear. The present study aimed to clarify the role of PGI2 in angiotensin II (Ang II)-induced AF and the underlying molecular mechanism. PGI2 content was decreased in both plasma and atrial tissue from patients with AF and mice treated with Ang II. Treatment with the PGI2 analog, iloprost, reduced Ang II-induced AF and atrial fibrosis. Iloprost prevented Ang II-induced atrial fibroblast collagen synthesis and differentiation. RNA-sequencing analysis revealed that iloprost significantly attenuated transcriptome changes in Ang II-treated atrial fibroblasts, especially mitogen-activated protein kinase (MAPK)-regulated genes. We demonstrated that iloprost elevated cAMP levels and then activated protein kinase A, resulting in a suppression of extracellular signal-regulated kinase1/2 and P38 activation, and ultimately inhibiting MAPK-dependent interleukin-6 transcription. In contrast, cardiac fibroblast-specific IP-knockdown mice had increased Ang II-induced AF inducibility and aggravated atrial fibrosis. Together, our study suggests that PGI2/IP system protects against atrial fibrosis and that PGI2 is a therapeutic target for treating AF.The prospectively registered trial was approved by the Chinese Clinical Trial Registry. The trial registration number is ChiCTR2200056733. Data of registration was 2022/02/12.


Sujet(s)
Angiotensine-II , Fibrillation auriculaire , Remodelage auriculaire , Prostacycline , Souris de lignée C57BL , Transduction du signal , Animaux , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/anatomopathologie , Fibrillation auriculaire/induit chimiquement , Fibrillation auriculaire/prévention et contrôle , Souris , Humains , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Remodelage auriculaire/effets des médicaments et des substances chimiques , Prostacycline/métabolisme , Fibrose , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/anatomopathologie , Atrium du coeur/métabolisme , Atrium du coeur/anatomopathologie , Atrium du coeur/effets des médicaments et des substances chimiques , Iloprost/pharmacologie , Récepteurs de l'époprosténol/métabolisme , Récepteurs de l'époprosténol/génétique , Femelle
6.
Int J Mol Sci ; 25(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38892223

RÉSUMÉ

The high incidence of atrial fibrillation (AFib) following cardiac surgery (postoperative atrial fibrillation, POAF) relies on specific surgical features. However, in the setting of POAF, the role of the microbiome in the modulation of cardiac fibrosis is still not clear. This study aimed to analyze the effect of the microbiome and its main metabolic product (trimethylamine-N-oxide, TMAO) in the fibrosis of myocardial tissue, to investigate its role in POAF. Patients undergoing elective cardiac surgery with cardiopulmonary bypass, central atrio-caval cannulation and no history of AFib, were included. A fragment of the right atrium was analyzed for qualitative and mRNA-quantitative evaluation. A preoperative blood sample was analyzed with enzyme-linked immunosorbent assay (ELISA). A total of 100 patients have been included, with POAF occurring in 38%. Histologically, a higher degree of fibrosis, angiogenesis and inflammation has been observed in POAF. Quantitative evaluation showed increased mRNA expression of collagen-1, collagen-3, fibronectin, and transforming growth factor beta (TGFb) in the POAF group. ELISA analysis showed higher levels of TMAO, lipopolysaccharide and TGFb in POAF, with similar levels of sP-selectin and zonulin. TMAO ≥ 61.8 ng/mL (odds ratio, OR 2.88 [1.35-6.16], p = 0.006), preoperative hemoglobin < 13.1 g/dL (OR 2.37 [1.07-5.24], p = 0.033) and impaired right ventricular function (OR 2.38 [1.17-4.83], p = 0.017) were independent predictors of POAF. Also, TMAO was significantly associated with POAF by means of increased fibrosis. Gut microbiome product TMAO is crucial for myocardial fibrosis, which is a key factor for POAF. Patients in preoperative sinus rhythm who will develop POAF have increased genetic expression of pro-fibrotic genes and enhanced fibrosis in histological staining. Elevated TMAO level (≥61.8 ng/mL) is an independent risk factor for POAF.


Sujet(s)
Fibrillation auriculaire , Fibrose , Microbiome gastro-intestinal , Myocarde , Humains , Fibrillation auriculaire/étiologie , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/anatomopathologie , Mâle , Femelle , Sujet âgé , Adulte d'âge moyen , Myocarde/métabolisme , Myocarde/anatomopathologie , Complications postopératoires/étiologie , Complications postopératoires/microbiologie , Méthylamines/sang , Méthylamines/métabolisme
8.
Sci Rep ; 14(1): 13976, 2024 06 17.
Article de Anglais | MEDLINE | ID: mdl-38886511

RÉSUMÉ

Stroke is an acute cerebrovascular disease in which blood flow to the brain is suddenly disrupted, causing damage to nerve cells. It involves complex and diverse pathophysiological processes and the treatment strategies are also diverse. The treatment for patients with stroke and atrial fibrillation (AF) is aimed at suppressing thrombus formation and migration. However, information regarding the protein networking involved in different thrombus formation pathways in patients with AF and stroke is insufficient. We performed protein profiling of patients with ischemic stroke with and without AF to investigate the mechanisms of thrombus formation and its pathophysiological association while providing helpful information for treating and managing patients with AF. These two groups were compared to identify the protein networks related to thrombus formation in AF. We observed that patients with ischemic stroke and AF had activated inflammatory responses induced by C-reactive protein, lipopolysaccharide-binding protein, and alpha-1-acid glycoprotein 1. In contrast, thyroid hormones were increased due to a decrease in transthyretin and retinol-binding protein 4 levels. The mechanism underlying enhanced cardiac activity, vasodilation, and the resulting thrombosis pathway were confirmed in AF. These findings will play an essential role in improving the prevention and treatment of AF-related stroke.


Sujet(s)
Fibrillation auriculaire , Thrombose , Humains , Fibrillation auriculaire/métabolisme , Thrombose/métabolisme , Mâle , Femelle , Sujet âgé , Adulte d'âge moyen , Protéines du sang/métabolisme , Accident vasculaire cérébral/métabolisme , Accident vasculaire cérébral/sang , Accident vasculaire cérébral ischémique/métabolisme , Cartes d'interactions protéiques , Protéomique/méthodes
9.
Eur J Pharmacol ; 977: 176714, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38849043

RÉSUMÉ

OBJECTIVE: This study aimed to elucidate the molecular mechanisms by which BRD4 play a role in atrial fibrillation (AF). METHODS AND RESULTS: We used a discovery-driven approach to detect BRD4 expression in the atria of patients with AF and in various murine models of atrial fibrosis. We used a BRD4 inhibitor (JQ1) and atrial fibroblast (aFB)-specific BRD4-knockout mice to elucidate the role of BRD4 in AF. We further examined the underlying mechanisms using RNA-seq and ChIP-seq analyses in vitro, to identify key downstream targets of BRD4. We found that BRD4 expression is significantly increased in patients with AF, with accompanying atrial fibrosis and aFB differentiation. We showed that JQ1 treatment and shRNA-based molecular silencing of BRD4 blocked ANG-II-induced extracellular matrix production and cell-cycle progression in aFBs. BRD4-related RNA-seq and ChIP-seq analyses in aFBs demonstrated enrichment of a subset of promoters related to the expression of profibrotic and proliferation-related genes. The pharmacological inhibition of BRD4 in vivo or in aFB-specific BRD4-knockout in mice limited ANG-II-induced atrial fibrosis, atrial enlargement, and AF susceptibility. CONCLUSION: Our findings suggest that BRD4 plays a key role in pathological AF, at least partially by activating aFB proliferation and ECM synthesis. This study provides mechanistic insights into the development of BRD4 inhibitors as targeted antiarrhythmic therapies.


Sujet(s)
Fibrillation auriculaire , Azépines , Protéines du cycle cellulaire , Fibrose , Atrium du coeur , Souris knockout , Facteurs de transcription , Triazoles , Fibrillation auriculaire/génétique , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/anatomopathologie , Fibrillation auriculaire/traitement médicamenteux , Animaux , Facteurs de transcription/génétique , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Atrium du coeur/anatomopathologie , Atrium du coeur/effets des médicaments et des substances chimiques , Atrium du coeur/métabolisme , Humains , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/métabolisme , Souris , Azépines/pharmacologie , Azépines/usage thérapeutique , Mâle , Triazoles/pharmacologie , Triazoles/usage thérapeutique , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/antagonistes et inhibiteurs , Modèles animaux de maladie humaine , Souris de lignée C57BL , Angiotensine-II/pharmacologie , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Thérapie moléculaire ciblée , Protéines contenant un bromodomaine
10.
Cells ; 13(12)2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38920645

RÉSUMÉ

This manuscript explores the intricate role of acetylcholine-activated inward rectifier potassium (KACh) channels in the pathogenesis of atrial fibrillation (AF), a common cardiac arrhythmia. It delves into the molecular and cellular mechanisms that underpin AF, emphasizing the vital function of KACh channels in modulating the atrial action potential and facilitating arrhythmogenic conditions. This study underscores the dual nature of KACh activation and its genetic regulation, revealing that specific variations in potassium channel genes, such as Kir3.4 and K2P3.1, significantly influence the electrophysiological remodeling associated with AF. Furthermore, this manuscript identifies the crucial role of the KACh-mediated current, IKACh, in sustaining arrhythmia through facilitating shorter re-entry circuits and stabilizing the re-entrant circuits, particularly in response to vagal nerve stimulation. Experimental findings from animal models, which could not induce AF in the absence of muscarinic activation, highlight the dependency of AF induction on KACh channel activity. This is complemented by discussions on therapeutic interventions, where KACh channel blockers have shown promise in AF management. Additionally, this study discusses the broader implications of KACh channel behavior, including its ubiquitous presence across different cardiac regions and species, contributing to a comprehensive understanding of AF dynamics. The implications of these findings are profound, suggesting that targeting KACh channels might offer new therapeutic avenues for AF treatment, particularly in cases resistant to conventional approaches. By integrating genetic, cellular, and pharmacological perspectives, this manuscript offers a holistic view of the potential mechanisms and therapeutic targets in AF, making a significant contribution to the field of cardiac arrhythmia research.


Sujet(s)
Fibrillation auriculaire , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/physiopathologie , Fibrillation auriculaire/génétique , Humains , Animaux , Canaux potassiques rectifiants entrants couplés aux protéines G/métabolisme , Canaux potassiques rectifiants entrants couplés aux protéines G/génétique , Potentiels d'action , Acétylcholine/métabolisme
11.
Europace ; 26(6)2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38788213

RÉSUMÉ

AIMS: Human induced pluripotent stem cell-derived atrial cardiomyocytes (hiPSC-aCM) could be a helpful tool to study the physiology and diseases of the human atrium. To fulfil this expectation, the electrophysiology of hiPSC-aCM should closely resemble the situation in the human atrium. Data on the contribution of the slowly activating delayed rectifier currents (IKs) to repolarization are lacking for both human atrium and hiPSC-aCM. METHODS AND RESULTS: Human atrial tissues were obtained from patients with sinus rhythm (SR) or atrial fibrillation (AF). Currents were measured in human atrial cardiomyocytes (aCM) and compared with hiPSC-aCM and used to model IKs contribution to action potential (AP) shape. Action potential was recorded by sharp microelectrodes. HMR-1556 (1 µM) was used to identify IKs and to estimate IKs contribution to repolarization. Less than 50% of hiPSC-aCM and aCM possessed IKs. Frequency of occurrence, current densities, activation/deactivation kinetics, and voltage dependency of IKs did not differ significantly between hiPSC-aCM and aCM, neither in SR nor AF. ß-Adrenoceptor stimulation with isoprenaline did not increase IKs neither in aCM nor in hiPSC-aCM. In tissue from SR, block of IKs with HMR-1556 did not lengthen the action potential duration, even when repolarization reserve was reduced by block of the ultra-rapid repolarizing current with 4-aminopyridine or the rapidly activating delayed rectifier potassium outward current with E-4031. CONCLUSION: I Ks exists in hiPSC-aCM with biophysics not different from aCM. As in adult human atrium (SR and AF), IKs does not appear to relevantly contribute to repolarization in hiPSC-aCM.


Sujet(s)
Potentiels d'action , Fibrillation auriculaire , Canaux potassiques rectifiants retardés , Atrium du coeur , Cellules souches pluripotentes induites , Myocytes cardiaques , Humains , Myocytes cardiaques/physiologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Cellules souches pluripotentes induites/métabolisme , Atrium du coeur/physiopathologie , Canaux potassiques rectifiants retardés/métabolisme , Fibrillation auriculaire/physiopathologie , Fibrillation auriculaire/métabolisme , Femelle , Cellules cultivées , Mâle , Adulte d'âge moyen , Cinétique , Sujet âgé , Différenciation cellulaire , Modèles cardiovasculaires , Inhibiteurs des canaux potassiques/pharmacologie
12.
Front Biosci (Landmark Ed) ; 29(5): 201, 2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38812314

RÉSUMÉ

BACKGROUND: Ibrutinib could increase the risk of atrial fibrillation (AF) in chronic lymphocytic leukemia (CLL) patients. However, the precise mechanism underlying ibrutinib-induced AF remains incompletely elucidated. METHODS: We investigated the proportion of ibrutinib-treated CLL patients with new-onset AF. Optical mapping was conducted to reveal the proarrhythmic effect of ibrutinib on HL-1 cells. Fluorescence staining and western blot were used to compare connexins 43 and 40 expression in ibrutinib-treated and control groups. To identify autophagy phenotypes, we used western blot to detect autophagy-related proteins, transmission electron microscopy to picture autophagosomes, and transfected mCherry-GFP-LC3 virus to label autophagosomes and lysosomes. Hydroxychloroquine as an autophagy inhibitor was administered to rescue ibrutinib-induced Cx43 and Cx40 degradation. RESULTS: About 2.67% of patients developed atrial arrhythmias after ibrutinib administration. HL-1 cells treated with ibrutinib exhibited diminished conduction velocity and a higher incidence of reentry-like arrhythmias compared to controls. Cx43 and Cx40 expression reduced along with autophagy markers increased in HL-1 cells treated with ibrutinib. Inhibiting autophagy upregulated Cx43 and Cx40. CONCLUSIONS: The off-target effect of ibrutinib on the PI3K-AKT-mTOR signaling pathway caused connexin degradation and atrial arrhythmia via promoting autophagy. CLINICAL TRIAL REGISTRATION: ChiCTR2100046062, https://clin.larvol.com/trial-detail/ChiCTR2100046062.


Sujet(s)
Adénine , Fibrillation auriculaire , Autophagie , Connexine 43 , Connexines , Phosphatidylinositol 3-kinases , Pipéridines , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Humains , Adénine/analogues et dérivés , Adénine/pharmacologie , Adénine/effets indésirables , Sérine-thréonine kinases TOR/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Pipéridines/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases/métabolisme , Connexine 43/métabolisme , Connexine 43/génétique , Femelle , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/induit chimiquement , Connexines/métabolisme , Connexines/génétique , Mâle , Sujet âgé , Adulte d'âge moyen , , Troubles du rythme cardiaque/métabolisme , Troubles du rythme cardiaque/induit chimiquement
13.
Genes (Basel) ; 15(5)2024 05 16.
Article de Anglais | MEDLINE | ID: mdl-38790263

RÉSUMÉ

In the present study, we have explored the involvement of Toll-like Receptor 4 (TLR4) in atrial fibrillation (AF), by using a meta-analysis of publicly available human transcriptomic data. The meta-analysis revealed 565 upregulated and 267 downregulated differentially expressed genes associated with AF. Pathway enrichment analysis highlighted a significant overrepresentation in immune-related pathways for the upregulated genes. A significant overlap between AF differentially expressed genes and TLR4-modulated genes was also identified, suggesting the potential role of TLR4 in AF-related transcriptional changes. Additionally, the analysis of other Toll-like receptors (TLRs) revealed a significant association with TLR2 and TLR3 in AF-related gene expression patterns. The examination of MYD88 and TICAM1, genes associated with TLR4 signalling pathways, indicated a significant yet nonspecific enrichment of AF differentially expressed genes. In summary, this study offers novel insights into the molecular aspects of AF, suggesting a pathophysiological role of TLR4 and other TLRs. By targeting these specific receptors, new treatments might be designed to better manage AF, offering hope for improved outcomes in affected patients.


Sujet(s)
Fibrillation auriculaire , Récepteur de type Toll-4 , Humains , Fibrillation auriculaire/génétique , Fibrillation auriculaire/métabolisme , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme , Facteur de différenciation myéloïde-88/génétique , Facteur de différenciation myéloïde-88/métabolisme , Transcriptome , Transduction du signal/génétique , Biologie informatique/méthodes , Analyse de profil d'expression de gènes , Récepteur de type Toll-2/génétique , Récepteur de type Toll-2/métabolisme , Protéines adaptatrices du transport vésiculaire
14.
Cell Rep Med ; 5(5): 101559, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38744275

RÉSUMÉ

Dysfunction of the sympathetic nervous system and increased epicardial adipose tissue (EAT) have been independently associated with the occurrence of cardiac arrhythmia. However, their exact roles in triggering arrhythmia remain elusive. Here, using an in vitro coculture system with sympathetic neurons, cardiomyocytes, and adipocytes, we show that adipocyte-derived leptin activates sympathetic neurons and increases the release of neuropeptide Y (NPY), which in turn triggers arrhythmia in cardiomyocytes by interacting with the Y1 receptor (Y1R) and subsequently enhancing the activity of the Na+/Ca2+ exchanger (NCX) and calcium/calmodulin-dependent protein kinase II (CaMKII). The arrhythmic phenotype can be partially blocked by a leptin neutralizing antibody or an inhibitor of Y1R, NCX, or CaMKII. Moreover, increased EAT thickness and leptin/NPY blood levels are detected in atrial fibrillation patients compared with the control group. Our study provides robust evidence that the adipose-neural axis contributes to arrhythmogenesis and represents a potential target for treating arrhythmia.


Sujet(s)
Adipocytes , Tissu adipeux , Troubles du rythme cardiaque , Leptine , Myocytes cardiaques , Neuropeptide Y , Péricarde , Humains , Animaux , Péricarde/métabolisme , Péricarde/anatomopathologie , Tissu adipeux/métabolisme , Tissu adipeux/anatomopathologie , Troubles du rythme cardiaque/métabolisme , Troubles du rythme cardiaque/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Neuropeptide Y/métabolisme , Leptine/métabolisme , Adipocytes/métabolisme , Mâle , Calcium-Calmodulin-Dependent Protein Kinase Type 2/métabolisme , Neurones/métabolisme , Neurones/anatomopathologie , Échangeur sodium-calcium/métabolisme , Femelle , Récepteur neuropeptide Y/métabolisme , Adulte d'âge moyen , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/physiopathologie , Fibrillation auriculaire/anatomopathologie , Système nerveux sympathique/métabolisme , Souris ,
15.
Biomed Pharmacother ; 175: 116649, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38692059

RÉSUMÉ

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.


Sujet(s)
Fibrillation auriculaire , Inflammasomes , Myocytes cardiaques , Pipérazines , Protéines proto-oncogènes c-akt , Espèces réactives de l'oxygène , Transduction du signal , Sérine-thréonine kinases TOR , Animaux , Fibrillation auriculaire/induit chimiquement , Fibrillation auriculaire/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Inflammasomes/métabolisme , Inflammasomes/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Lapins , Espèces réactives de l'oxygène/métabolisme , Pipérazines/pharmacologie , Mâle , Phosphatidylinositol 3-kinases/métabolisme , Thiazoles/pharmacologie , Atrium du coeur/effets des médicaments et des substances chimiques , Atrium du coeur/métabolisme , Potentiels d'action/effets des médicaments et des substances chimiques , Neuroleptiques/pharmacologie
16.
Aging (Albany NY) ; 16(10): 8630-8644, 2024 05 21.
Article de Anglais | MEDLINE | ID: mdl-38775722

RÉSUMÉ

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.


Sujet(s)
Angiotensine-II , Fibrillation auriculaire , Benzylisoquinoléines , Fibrose , Facteur-2 apparenté à NF-E2 , Transduction du signal , Protéine Smad-3 , Facteur de croissance transformant bêta , Animaux , Angiotensine-II/pharmacologie , Fibrillation auriculaire/induit chimiquement , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/prévention et contrôle , Facteur-2 apparenté à NF-E2/métabolisme , Souris , Benzylisoquinoléines/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Protéine Smad-3/métabolisme , Mâle , Facteur de croissance transformant bêta/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Protéine Smad2/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Atrium du coeur/effets des médicaments et des substances chimiques , Atrium du coeur/métabolisme , Atrium du coeur/anatomopathologie , Heme oxygenase (decyclizing)/métabolisme , Protéines membranaires , Heme oxygenase-1
18.
J Mol Cell Cardiol ; 193: 11-24, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38797242

RÉSUMÉ

Atrial fibrillation (AF) is the most common cardiac rhythm disorder, often occurring in the setting of atrial distension and elevated myocardialstretch. While various mechano-electrochemical signal transduction pathways have been linked to AF development and progression, the underlying molecular mechanisms remain poorly understood, hampering AF therapies. In this review, we describe different aspects of stretch-induced electro-anatomical remodeling as seen in animal models and in patients with AF. Specifically, we focus on cellular and molecular mechanisms that are responsible for mechano-electrochemical signal transduction and the development of ectopic beats triggering AF from pulmonary veins, the most common source of paroxysmal AF. Furthermore, we describe structural changes caused by stretch occurring before and shortly after the onset of AF as well as during AF progression, contributing to longstanding forms of AF. We also propose mechanical stretch as a new dimension to the concept "AF begets AF", in addition to underlying diseases. Finally, we discuss the mechanisms of these electro-anatomical alterations in a search for potential therapeutic strategies and the development of novel antiarrhythmic drugs targeted at the components of mechano-electrochemical signal transduction not only in cardiac myocytes, but also in cardiac non-myocyte cells.


Sujet(s)
Fibrillation auriculaire , Humains , Animaux , Fibrillation auriculaire/physiopathologie , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/anatomopathologie , Remodelage auriculaire , Atrium du coeur/physiopathologie , Atrium du coeur/anatomopathologie , Atrium du coeur/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Transduction du signal , Veines pulmonaires/anatomopathologie , Veines pulmonaires/métabolisme , Veines pulmonaires/physiopathologie , Phénomènes électrophysiologiques
19.
Circulation ; 150(2): 111-127, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38726666

RÉSUMÉ

BACKGROUND: G protein-coupled receptors play a critical role in atrial fibrillation (AF). Spexin is a novel ligand of galanin receptors (GALRs). In this study, we investigated the regulation of spexin and GALRs on AF and the underlying mechanisms. METHODS: Global spexin knockout (SPX-KO) and cardiomyocyte-specific GALRs knockout (GALR-cKO) mice underwent burst pacing electrical stimulation. Optical mapping was used to determine atrial conduction velocity and action potential duration. Atrial myocyte action potential duration and inward rectifying K+ current (IK1) were recorded using whole-cell patch clamps. Isolated cardiomyocytes were stained with Fluo-3/AM dye, and intracellular Ca2+ handling was examined by CCD camera. A mouse model of AF was established by Ang-II (angiotensin II) infusion. RESULTS: Spexin plasma levels in patients with AF were lower than those in subjects without AF, and knockout of spexin increased AF susceptibility in mice. In the atrium of SPX-KO mice, potassium inwardly rectifying channel subfamily J member 2 (KCNJ2) and sarcolipin (SLN) were upregulated; meanwhile, IK1 current was increased and Ca2+ handling was impaired in isolated atrial myocytes of SPX-KO mice. GALR2-cKO mice, but not GALR1-cKO and GALR3-cKO mice, had a higher incidence of AF, which was associated with higher IK1 current and intracellular Ca2+ overload. The phosphorylation level of CREB (cyclic AMP responsive element binding protein 1) was upregulated in atrial tissues of SPX-KO and GALR2-cKO mice. Chromatin immunoprecipitation confirmed the recruitment of p-CREB to the proximal promoter regions of KCNJ2 and SLN. Finally, spexin treatment suppressed CREB signaling, decreased IK1 current and decreased intracellular Ca2+ overload, which thus reduced the inducibility of AF in Ang-II-infused mice. CONCLUSIONS: Spexin reduces atrial fibrillation susceptibility by inhibiting CREB phosphorylation and thus downregulating KCNJ2 and SLN transcription by GALR2 receptor. The spexin/GALR2/CREB signaling pathway represents a novel therapeutic avenue in the development of agents against atrial fibrillation.


Sujet(s)
Fibrillation auriculaire , Souris knockout , Myocytes cardiaques , Hormones peptidiques , Récepteur de la galanine de type 2 , Animaux , Fibrillation auriculaire/métabolisme , Hormones peptidiques/métabolisme , Souris , Myocytes cardiaques/métabolisme , Récepteur de la galanine de type 2/métabolisme , Récepteur de la galanine de type 2/génétique , Humains , Potentiels d'action/effets des médicaments et des substances chimiques , Mâle , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Modèles animaux de maladie humaine , Souris de lignée C57BL , Femelle , Transduction du signal
20.
Circ Arrhythm Electrophysiol ; 17(6): e012486, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38690652

RÉSUMÉ

BACKGROUND: 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME) has shown potential in protecting against heart disease, but its relationship with atrial fibrillation (AF) remains unknown. METHODS: Coronary sinus (CS) and femoral vein blood samplings were synchronously collected from AF and non-AF subjects (paroxysmal supraventricular tachycardia or idiopathic premature ventricular complexes) who underwent catheter ablation. First, untargeted metabolomic profiling was performed in a discovery cohort (including 12 AF and 12 non-AF subjects) to identify the most promising CS or femoral vein metabolite. Then, the selected metabolite was further measured in a validation cohort (including 119 AF and 103 non-AF subjects) to confirm its relationship with left atrium remodeling and 1-year postablation recurrence of AF. Finally, the biological function of the selected metabolite was validated in a rapid-paced cultured HL-1 atrial cardiomyocytes model. RESULTS: Metabolomic analysis identified CS 12,13-diHOME as the most pronounced change metabolite correlated with left atrium remodeling in the discovery cohort. In the validation cohort, CS 12,13-diHOME was significantly lower in patients with AF than non-AF controls (84.32±20.13 versus 96.24±23.56 pg/mL; P<0.01), and associated with worse structural, functional, and electrical remodeling of left atrium. Multivariable regression analyses further demonstrated that decreased CS 12,13-diHOME was an independent predictor of 1-year postablation recurrence of AF (odds ratio, 0.754 [95% CI, 0.648-0.920]; P=0.005). Biological function validations showed that 12,13-diHOME treatment significantly protect the cell viability, improved the expression of MHC (myosin heavy chain) and Cav1.2 (L-type calcium channel α1c), and attenuated mitochondrial damage in the rapid-paced cultured HL-1 cardiomyocytes model. CONCLUSIONS: CS metabolite 12,13-diHOME is decreased in patients with AF and can serve as a novel biomarker for left atrium remodeling.


Sujet(s)
Fibrillation auriculaire , Remodelage auriculaire , Marqueurs biologiques , Ablation par cathéter , Sinus coronaire , Fibrillation auriculaire/physiopathologie , Fibrillation auriculaire/chirurgie , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/diagnostic , Humains , Mâle , Femelle , Marqueurs biologiques/sang , Marqueurs biologiques/métabolisme , Adulte d'âge moyen , Sinus coronaire/métabolisme , Sinus coronaire/physiopathologie , Métabolomique , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Animaux , Sujet âgé , Études cas-témoins , Récidive , Fonction auriculaire gauche , Atrium du coeur/physiopathologie , Atrium du coeur/métabolisme , Valeur prédictive des tests
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE