Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.634
Filter
1.
Mol Cell Biol ; 44(4): 149-163, 2024.
Article in English | MEDLINE | ID: mdl-38725392

ABSTRACT

Phenylacetylglutamine (PAGln), a gut metabolite is substantially elevated in heart failure (HF). The increase of PAGln in plasma is associated with atrial fibrillation (AF), and contributes to AF pathogenesis. However, the role of PAGln in AF with HF remains uncertain. Therefore, this study aimed to determine the effect of PAGln on AF after HF. Thoracic aortic coarctation (TAC) created overpressure-induced HF mice for 4 weeks. Histopathology, biochemical, echocardiographic for assessment of cardiac function, and electrophysiological examination of several electrophysiological indexes (ERP, SNRT, and the occurrence rate of AF) were performed at the end of the HF mice model. We found that plasma PAGln levels were significantly elevated in PAGln-treated HF mice and that PAGln aggravated maladaptive structural remodeling and electrical remodeling, which aggravated the vulnerability of AF, shortened the ERP duration, prolonged the SNRT, increased the occurrence rate of AF in HF mice. Mechanistically, PAGln exacerbated ROS accumulation and increased the levels of phosphorylated PLB and CAMK II. Overall, PAGln played a vital role in promoting the occurrence of AF in HF mice by activating the CAMK II signaling pathway.


Subject(s)
Atrial Fibrillation , Heart Failure , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/etiology , Mice , Heart Failure/etiology , Heart Failure/metabolism , Male , Mice, Inbred C57BL , Disease Models, Animal , Glutamine/metabolism , Glutamine/analogs & derivatives , Glutamine/pharmacology , Signal Transduction/drug effects , Reactive Oxygen Species/metabolism
2.
Front Biosci (Landmark Ed) ; 29(5): 201, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38812314

ABSTRACT

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.


Subject(s)
Adenine , Atrial Fibrillation , Autophagy , Connexin 43 , Connexins , Phosphatidylinositol 3-Kinases , Piperidines , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Humans , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine/adverse effects , TOR Serine-Threonine Kinases/metabolism , Autophagy/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Piperidines/pharmacology , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Connexin 43/metabolism , Connexin 43/genetics , Female , Atrial Fibrillation/metabolism , Atrial Fibrillation/chemically induced , Connexins/metabolism , Connexins/genetics , Male , Aged , Middle Aged , Gap Junction alpha-5 Protein , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/chemically induced
3.
Genes (Basel) ; 15(5)2024 May 16.
Article in English | MEDLINE | ID: mdl-38790263

ABSTRACT

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.


Subject(s)
Atrial Fibrillation , Toll-Like Receptor 4 , Humans , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Transcriptome , Signal Transduction/genetics , Computational Biology/methods , Gene Expression Profiling , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Adaptor Proteins, Vesicular Transport
4.
Int J Mol Sci ; 25(10)2024 May 20.
Article in English | MEDLINE | ID: mdl-38791605

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate gene expression by binding to target messenger RNAs (mRNAs). miRNAs have been implicated in a variety of cardiovascular and neurological diseases, such as myocardial infarction, cardiomyopathies of various geneses, rhythmological diseases, neurodegenerative illnesses and strokes. Numerous studies have focused on the expression of miRNA patterns with respect to atrial fibrillation (AF) or acute ischemic stroke (AIS) However, only a few studies have addressed the expression pattern of miRNAs in patients with AF and AIS in order to provide not only preventive information but also to identify therapeutic potentials. Therefore, the aim of this review is to summarize 18 existing manuscripts that have dealt with this combined topic of AF and associated AIS in detail and to shed light on the most frequently mentioned miRNAs-1, -19, -21, -145 and -146 with regard to their molecular mechanisms and targets on both the heart and the brain. From this, possible diagnostic and therapeutic consequences for the future could be derived.


Subject(s)
Atrial Fibrillation , Biomarkers , MicroRNAs , Stroke , Humans , Atrial Fibrillation/genetics , Atrial Fibrillation/therapy , Atrial Fibrillation/metabolism , MicroRNAs/genetics , Stroke/genetics , Stroke/metabolism , Stroke/therapy , Gene Expression Regulation , Animals
5.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731908

ABSTRACT

In atrial fibrillation (AF), multifactorial pathologic atrial alterations are manifested by structural and electrophysiological changes known as atrial remodeling. AF frequently develops in the context of underlying cardiac abnormalities. A critical mechanistic role played by atrial stretch is played by abnormal substrates in a number of conditions that predispose to AF, including obesity, heart failure, hypertension, and sleep apnea. The significant role of overweight and obesity in the development of AF is known; however, the differential effect of overweight, obesity, cardiovascular comorbidities, lifestyle, and other modifiable risk factors on the occurrence and recurrence of AF remains to be determined. Reverse remodeling of the atrial substrate and subsequent reduction in the AF burden by conversion into a typical sinus rhythm has been associated with weight loss through lifestyle changes or surgery. This makes it an essential pillar in the management of AF in obese patients. According to recently published research, microRNAs (miRs) may function as post-transcriptional regulators of genes involved in atrial remodeling, potentially contributing to the pathophysiology of AF. The focus of this review is on their modulation by both weight loss and catheter ablation interventions to counteract atrial remodeling in AF. Our analysis outlines the experimental and clinical evidence supporting the synergistic effects of weight loss and catheter ablation (CA) in reversing atrial electrical and structural remodeling in AF onset and in recurrent post-ablation AF by attenuating pro-thrombotic, pro-inflammatory, pro-fibrotic, arrhythmogenic, and male-sex-associated hypertrophic remodeling pathways. Furthermore, we discuss the promising role of miRs with prognostic potential as predictive biomarkers in guiding approaches to AF recurrence prevention.


Subject(s)
Atrial Fibrillation , Biomarkers , Catheter Ablation , MicroRNAs , Weight Loss , Atrial Fibrillation/metabolism , Atrial Fibrillation/genetics , Atrial Fibrillation/etiology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Catheter Ablation/methods , Recurrence , Atrial Remodeling , Animals , Obesity/metabolism , Obesity/complications
6.
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
7.
Sci Rep ; 14(1): 12470, 2024 05 30.
Article in English | MEDLINE | ID: mdl-38816374

ABSTRACT

Atrial fibrosis serves as an arrhythmogenic substrate in atrial fibrillation (AF) and contributes to AF persistence. Treating atrial fibrosis is challenging because atrial fibroblast activity is multifactorial. We hypothesized that the primary cilium regulates the profibrotic response of AF atrial fibroblasts, and explored therapeutic potentials of targeting primary cilia to treat fibrosis in AF. We included 25 patients without AF (non-AF) and 26 persistent AF patients (AF). Immunohistochemistry using a subset of the patients (non-AF: n = 10, AF: n = 10) showed less ciliated fibroblasts in AF versus non-AF. Acetylated α-tubulin protein levels were decreased in AF, while the gene expressions of AURKA and NEDD9 were highly increased in AF patients' left atrium. Loss of primary cilia in human atrial fibroblasts through IFT88 knockdown enhanced expression of ECM genes, including FN1 and COL1A1. Remarkably, restoration or elongation of primary cilia by an AURKA selective inhibitor or lithium chloride, respectively, prevented the increased expression of ECM genes induced by different profibrotic cytokines in atrial fibroblasts of AF patients. Our data reveal a novel mechanism underlying fibrotic substrate formation via primary cilia loss in AF atrial fibroblasts and suggest a therapeutic potential for abrogating atrial fibrosis by restoring primary cilia.


Subject(s)
Atrial Fibrillation , Aurora Kinase A , Cilia , Fibroblasts , Fibrosis , Heart Atria , Humans , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Atrial Fibrillation/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Cilia/metabolism , Cilia/pathology , Heart Atria/metabolism , Heart Atria/pathology , Male , Female , Middle Aged , Aurora Kinase A/metabolism , Aurora Kinase A/genetics , Aurora Kinase A/antagonists & inhibitors , Aged , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Tubulin/metabolism , Cells, Cultured , Tumor Suppressor Proteins
8.
Cell Rep Med ; 5(5): 101559, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38744275

ABSTRACT

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.


Subject(s)
Adipocytes , Adipose Tissue , Arrhythmias, Cardiac , Leptin , Myocytes, Cardiac , Neuropeptide Y , Pericardium , Humans , Animals , Pericardium/metabolism , Pericardium/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neuropeptide Y/metabolism , Leptin/metabolism , Adipocytes/metabolism , Male , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Neurons/metabolism , Neurons/pathology , Sodium-Calcium Exchanger/metabolism , Female , Receptors, Neuropeptide Y/metabolism , Middle Aged , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Atrial Fibrillation/pathology , Sympathetic Nervous System/metabolism , Mice , Epicardial Adipose Tissue
9.
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
10.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38673963

ABSTRACT

Accurate etiologic diagnosis provides an appropriate secondary prevention and better prognosis in ischemic stroke (IS) patients; still, 45% of IS are cryptogenic, urging us to enhance diagnostic precision. We have studied the transcriptomic content of plasma extracellular vesicles (EVs) (n = 21) to identify potential biomarkers of IS etiologies. The proteins encoded by the selected genes were measured in the sera of IS patients (n = 114) and in hypertensive patients with (n = 78) and without atrial fibrillation (AF) (n = 20). IGFBP-2, the most promising candidate, was studied using immunohistochemistry in the IS thrombi (n = 23) and atrium of AF patients (n = 13). In vitro, the IGFBP-2 blockade was analyzed using thromboelastometry and endothelial cell cultures. We identified 745 differentially expressed genes among EVs of cardioembolic, atherothrombotic, and ESUS groups. From these, IGFBP-2 (cutoff > 247.6 ng/mL) emerged as a potential circulating biomarker of embolic IS [OR = 8.70 (1.84-41.13) p = 0.003], which was increased in patients with AF vs. controls (p < 0.001) and was augmented in cardioembolic vs. atherothrombotic thrombi (p < 0.01). Ex vivo, the blockage of IGFBP-2 reduced clot firmness (p < 0.01) and lysis time (p < 0.001) and in vitro, diminished endothelial permeability (p < 0.05) and transmigration (p = 0.06). IGFBP-2 could be a biomarker of embolic IS and a new therapeutic target involved in clot formation and endothelial dysfunction.


Subject(s)
Biomarkers , Extracellular Vesicles , Insulin-Like Growth Factor Binding Protein 2 , Ischemic Stroke , Thrombosis , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Biomarkers/blood , Male , Female , Aged , Thrombosis/metabolism , Thrombosis/etiology , Thrombosis/blood , Ischemic Stroke/metabolism , Ischemic Stroke/blood , Ischemic Stroke/genetics , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor Binding Protein 2/blood , Middle Aged , Gene Expression Profiling , Transcriptome , Atrial Fibrillation/metabolism , Atrial Fibrillation/genetics , Atrial Fibrillation/complications , Atrial Fibrillation/blood
11.
Eur J Histochem ; 68(2)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38656259

ABSTRACT

This study investigated the impact of resveratrol on abnormal metabolic remodeling in atrial fibrillation (AF) and explored potential molecular mechanisms. An AF cell model was established by high-frequency electrical stimulation of HL-1 atrial muscle cells. Resveratrol concentrations were optimized using CCK-8 and flow cytometry. AF-induced increases in ROS and mitochondrial calcium, along with decreased adenosine triphosphate (ATP) and mitochondrial membrane potential, were observed. Resveratrol mitigated these changes and maintained normal mitochondrial morphology. Moreover, resveratrol acted through the SIRT3-dependent pathway, as evidenced by its ability to suppress AF-induced acetylation of key metabolic enzymes. SIRT3 overexpression controls acetylation modifications, suggesting its regulatory role. In conclusion, resveratrol's SIRT3-dependent pathway intervenes in AF-induced mitochondrial dysfunction, presenting a potential therapeutic avenue for AF-related metabolic disorders. This study sheds light on the role of resveratrol in mitigating AF-induced mitochondrial remodeling and highlights its potential as a novel treatment for AF.


Subject(s)
Atrial Fibrillation , Resveratrol , Sirtuin 3 , Resveratrol/pharmacology , Sirtuin 3/metabolism , Atrial Fibrillation/metabolism , Atrial Fibrillation/drug therapy , Animals , Mice , Cell Line , Signal Transduction/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Reactive Oxygen Species/metabolism
12.
J Biol Chem ; 300(5): 107294, 2024 May.
Article in English | MEDLINE | ID: mdl-38636665

ABSTRACT

Exenatide, a promising cardioprotective agent, protects against cardiac structural remodeling and diastolic dysfunction. Combined blockade of sodium and potassium channels is valuable for managing atrial fibrillation (AF). Here, we explored whether exenatide displayed anti-AF effects by inhibiting human Kv1.5 and Nav1.5 channels. We used the whole-cell patch-clamp technique to investigate the effects of exenatide on hKv1.5 and hNav1.5 channels expressed in human embryonic kidney 293 cells and studied the effects of exenatide on action potential (AP) and other cardiac ionic currents in rat atrial myocytes. Additionally, an electrical mapping system was used to explore the effects of exenatide on electrical properties and AF activity in isolated rat hearts. Finally, a rat AF model, established using acetylcholine and calcium chloride, was employed to evaluate the anti-AF potential of exenatide in rats. Exenatide reversibly suppressed IKv1.5 with IC50 of 3.08 µM, preferentially blocked the hKv1.5 channel in its closed state, and positively shifted the voltage-dependent activation curve. Exenatide also reversibly inhibited INav1.5 with IC50 of 3.30 µM, negatively shifted the voltage-dependent inactivation curve, and slowed its recovery from inactivation with significant use-dependency at 5 and 10 Hz. Furthermore, exenatide prolonged AP duration and suppressed the sustained K+ current (Iss) and transient outward K+ current (Ito), but without inhibition of L-type Ca2+ current (ICa,L) in rat atrial myocytes. Exenatide prevented AF incidence and duration in rat hearts and rats. These findings demonstrate that exenatide inhibits IKv1.5 and INav1.5in vitro and reduces AF susceptibility in isolated rat hearts and rats.


Subject(s)
Action Potentials , Atrial Fibrillation , Exenatide , Kv1.5 Potassium Channel , Myocytes, Cardiac , NAV1.5 Voltage-Gated Sodium Channel , Animals , Exenatide/pharmacology , Atrial Fibrillation/metabolism , Atrial Fibrillation/drug therapy , Atrial Fibrillation/pathology , Atrial Fibrillation/prevention & control , Humans , Kv1.5 Potassium Channel/metabolism , Kv1.5 Potassium Channel/antagonists & inhibitors , Kv1.5 Potassium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , Rats , Action Potentials/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , HEK293 Cells , Male , Rats, Sprague-Dawley
13.
J Mol Cell Cardiol ; 190: 1-12, 2024 May.
Article in English | MEDLINE | ID: mdl-38514002

ABSTRACT

BACKGROUND: Overexpression of the CREM (cAMP response element-binding modulator) isoform CREM-IbΔC-X in transgenic mice (CREM-Tg) causes the age-dependent development of spontaneous AF. PURPOSE: To identify key proteome signatures and biological processes accompanying the development of persistent AF through integrated proteomics and bioinformatics analysis. METHODS: Atrial tissue samples from three CREM-Tg mice and three wild-type littermates were subjected to unbiased mass spectrometry-based quantitative proteomics, differential expression and pathway enrichment analysis, and protein-protein interaction (PPI) network analysis. RESULTS: A total of 98 differentially expressed proteins were identified. Gene ontology analysis revealed enrichment for biological processes regulating actin cytoskeleton organization and extracellular matrix (ECM) dynamics. Changes in ITGAV, FBLN5, and LCP1 were identified as being relevant to atrial fibrosis and structural based on expression changes, co-expression patterns, and PPI network analysis. Comparative analysis with previously published datasets revealed a shift in protein expression patterns from ion-channel and metabolic regulators in young CREM-Tg mice to profibrotic remodeling factors in older CREM-Tg mice. Furthermore, older CREM-Tg mice exhibited protein expression patterns reminiscent of those seen in humans with persistent AF. CONCLUSIONS: This study uncovered distinct temporal changes in atrial protein expression patterns with age in CREM-Tg mice consistent with the progressive evolution of AF. Future studies into the role of the key differentially abundant proteins identified in this study in AF progression may open new therapeutic avenues to control atrial fibrosis and substrate development in AF.


Subject(s)
Atrial Fibrillation , Cyclic AMP Response Element Modulator , Fibrosis , Heart Atria , Mice, Transgenic , Proteomics , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/genetics , Cyclic AMP Response Element Modulator/metabolism , Cyclic AMP Response Element Modulator/genetics , Proteomics/methods , Heart Atria/metabolism , Heart Atria/pathology , Mice , Gene Expression Regulation , Protein Interaction Maps , Proteome/metabolism , Disease Models, Animal , Gene Expression Profiling , Extracellular Matrix/metabolism , Male
14.
Physiol Rep ; 12(6): e15957, 2024 03.
Article in English | MEDLINE | ID: mdl-38546216

ABSTRACT

Epicardial adipose tissue (EAT) is an active endocrine organ that is closely associated with occurrence of atrial fibrillation (AF). However, the role of EAT in the development of postoperative AF (POAF) remains unclear. We aimed to investigate the association between EAT profile and POAF occurrence in patients who underwent cardiovascular surgery. We obtained EAT samples from 53 patients to evaluate gene expression, histological changes, mitochondrial oxidative phosphorylation (OXPHOS) capacity in the EAT, and protein secretion in EAT-conditioned medium. EAT volume was measured using computed tomography scan. Eighteen patients (34%) experienced POAF within 7 days after surgery. Although no significant difference was observed in EAT profile between patients with and without POAF, logistic regression analysis identified that the mRNA expression levels of tumor necrosis factor-alpha (TNF-α) were positively correlated and adipocyte size in the EAT was inversely correlated with onset of POAF, respectively. Mitochondrial OXPHOS capacity in the EAT was not associated with POAF occurrence; however, it showed an inverse correlation with adipocyte size and a positive correlation with adiponectin secretion. In conclusion, changes in the secretory profile and adipocyte morphology of the EAT, which represent qualitative aspects of the adipose tissue, were present before the onset of AF.


Subject(s)
Atrial Fibrillation , Humans , Atrial Fibrillation/metabolism , Epicardial Adipose Tissue , Adipocytes/metabolism , Adipose Tissue/metabolism , Inflammation/metabolism , Pericardium/metabolism
15.
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
16.
J Transl Med ; 22(1): 259, 2024 Mar 09.
Article in English | MEDLINE | ID: mdl-38461346

ABSTRACT

BACKGROUND: Amino acids (AAs) are one of the primary metabolic substrates for cardiac work. The correlation between AAs and both atrial fibrillation (AF) and aging has been documented. However, the relationship between AAs and age-related AF remains unclear. METHODS: Initially, the plasma AA levels of persistent AF patients and control subjects were assessed, and the correlations between AA levels, age, and other clinical indicators were explored. Subsequently, the age-related AF mouse model was constructed and the untargeted myocardial metabolomics was conducted to detect the level of AAs and related metabolites. Additionally, the gut microbiota composition associated with age-related AF was detected by a 16S rDNA amplicon sequencing analysis on mouse fecal samples. RESULTS: Higher circulation levels of lysine (Student's t-test, P = 0.001), tyrosine (P = 0.002), glutamic acid (P = 0.008), methionine (P = 0.008), and isoleucine (P = 0.014), while a lower level of glycine (P = 0.003) were observed in persistent AF patients. The feature AAs identified by machine learning algorithms were glutamic acid and methionine. The association between AAs and age differs between AF and control subjects. Distinct patterns of AA metabolic profiles were observed in the myocardial metabolites of aged AF mice. Aged AF mice had lower levels of Betaine, L-histidine, L-alanine, L-arginine, L-Pyroglutamic acid, and L-Citrulline compared with adult AF mice. Aged AF mice also presented a different gut microbiota pattern, and its functional prediction analysis showed AA metabolism alteration. CONCLUSION: This study provided a comprehensive network of AA disturbances in age-related AF from multiple dimensions, including plasma, myocardium, and gut microbiota. Disturbances of AAs may serve as AF biomarkers, and restoring their homeostasis may have potential benefits for the management of age-related AF.


Subject(s)
Amino Acids , Atrial Fibrillation , Adult , Humans , Animals , Mice , Aged , Amino Acids/metabolism , Atrial Fibrillation/metabolism , Metabolomics/methods , Methionine , Glutamates
17.
J Biol Chem ; 300(3): 105735, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38336298

ABSTRACT

One of the independent risk factors for atrial fibrillation is diabetes mellitus (DM); however, the underlying mechanisms causing atrial fibrillation in DM are unknown. The underlying mechanism of Atrogin-1-mediated SK2 degradation and associated signaling pathways are unclear. The aim of this study was to elucidate the relationship among reactive oxygen species (ROS), the NF-κB signaling pathway, and Atrogin-1 protein expression in the atrial myocardia of DM mice. We found that SK2 expression was downregulated comitant with increased ROS generation and enhanced NF-κB signaling activation in the atrial cardiomyocytes of DM mice. These observations were mimicked by exogenously applicating H2O2 and by high glucose culture conditions in HL-1 cells. Inhibition of ROS production by diphenyleneiodonium chloride or silencing of NF-κB by siRNA decreased the protein expression of NF-κB and Atrogin-1 and increased that of SK2 in HL-1 cells with high glucose culture. Moreover, chromatin immunoprecipitation assay demonstrated that NF-κB/p65 directly binds to the promoter of the FBXO32 gene (encoding Atrogin-1), regulating the FBXO32 transcription. Finally, we evaluated the therapeutic effects of curcumin, known as a NF-κB inhibitor, on Atrogin-1 and SK2 expression in DM mice and confirmed that oral administration of curcumin for 4 weeks significantly suppressed Atrogin-1 expression and protected SK2 expression against hyperglycemia. In summary, the results from this study indicated that the ROS/NF-κB signaling pathway participates in Atrogin-1-mediated SK2 regulation in the atria of streptozotocin-induced DM mice.


Subject(s)
Diabetes Mellitus, Experimental , Heart Atria , Muscle Proteins , NF-kappa B , Reactive Oxygen Species , SKP Cullin F-Box Protein Ligases , Signal Transduction , Small-Conductance Calcium-Activated Potassium Channels , Animals , Mice , Atrial Fibrillation/etiology , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Atrial Fibrillation/physiopathology , Cell Line , Chromatin Immunoprecipitation , Curcumin/pharmacology , Curcumin/therapeutic use , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Gene Expression Regulation/drug effects , Glucose/pharmacology , Heart Atria/metabolism , Heart Atria/physiopathology , Hydrogen Peroxide/pharmacology , Hyperglycemia/genetics , Hyperglycemia/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocardium , Myocytes, Cardiac , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Proteolysis , Reactive Oxygen Species/metabolism , RNA, Small Interfering , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Small-Conductance Calcium-Activated Potassium Channels/genetics , Small-Conductance Calcium-Activated Potassium Channels/metabolism
18.
Circulation ; 149(18): 1419-1434, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38357791

ABSTRACT

BACKGROUND: Clonal hematopoiesis of indeterminate potential (CHIP), a common age-associated phenomenon, associates with increased risk of both hematological malignancy and cardiovascular disease. Although CHIP is known to increase the risk of myocardial infarction and heart failure, the influence of CHIP in cardiac arrhythmias, such as atrial fibrillation (AF), is less explored. METHODS: CHIP prevalence was determined in the UK Biobank, and incident AF analysis was stratified by CHIP status and clone size using Cox proportional hazard models. Lethally irradiated mice were transplanted with hematopoietic-specific loss of Tet2, hematopoietic-specific loss of Tet2 and Nlrp3, or wild-type control and fed a Western diet, compounded with or without NLRP3 (NLR [NACHT, LRR {leucine rich repeat}] family pyrin domain containing protein 3) inhibitor, NP3-361, for 6 to 9 weeks. Mice underwent in vivo invasive electrophysiology studies and ex vivo optical mapping. Cardiomyocytes from Ldlr-/- mice with hematopoietic-specific loss of Tet2 or wild-type control and fed a Western diet were isolated to evaluate calcium signaling dynamics and analysis. Cocultures of pluripotent stem cell-derived atrial cardiomyocytes were incubated with Tet2-deficient bone marrow-derived macrophages, wild-type control, or cytokines IL-1ß (interleukin 1ß) or IL-6 (interleukin 6). RESULTS: Analysis of the UK Biobank showed individuals with CHIP, in particular TET2 CHIP, have increased incident AF. Hematopoietic-specific inactivation of Tet2 increases AF propensity in atherogenic and nonatherogenic mouse models and is associated with increased Nlrp3 expression and CaMKII (Ca2+/calmodulin-dependent protein kinase II) activation, with AF susceptibility prevented by inactivation of Nlrp3. Cardiomyocytes isolated from Ldlr-/- mice with hematopoietic inactivation of Tet2 and fed a Western diet have impaired calcium release from the sarcoplasmic reticulum into the cytosol, contributing to atrial arrhythmogenesis. Abnormal sarcoplasmic reticulum calcium release was recapitulated in cocultures of cardiomyocytes with the addition of Tet2-deficient macrophages or cytokines IL-1ß or IL-6. CONCLUSIONS: We identified a modest association between CHIP, particularly TET2 CHIP, and incident AF in the UK Biobank population. In a mouse model of AF resulting from hematopoietic-specific inactivation of Tet2, we propose altered calcium handling as an arrhythmogenic mechanism, dependent on Nlrp3 inflammasome activation. Our data are in keeping with previous studies of CHIP in cardiovascular disease, and further studies into the therapeutic potential of NLRP3 inhibition for individuals with TET2 CHIP may be warranted.


Subject(s)
Atrial Fibrillation , Clonal Hematopoiesis , DNA-Binding Proteins , Dioxygenases , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Proto-Oncogene Proteins , Animals , Dioxygenases/metabolism , Dioxygenases/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Atrial Fibrillation/metabolism , Atrial Fibrillation/etiology , Atrial Fibrillation/genetics , Atrial Fibrillation/pathology , Inflammasomes/metabolism , Humans , Mice , Clonal Hematopoiesis/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Male , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Middle Aged , Mice, Knockout , Risk Factors
19.
Stem Cell Res ; 76: 103336, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38341987

ABSTRACT

Gain-of-function mutations in the KCNQ1 gene can cause atrial fibrillation. In this study, we generated an induced stem cell line (GRCHJUi001) from one member of an atrial fibrillation family line, whom had heterozygous mutation in the KCNQ1 gene c.625 T > C (p.Ser209Pro), and the cell line showed maintenance of stem cells characterized by morphology, normal karyotype, and pluripotency.


Subject(s)
Atrial Fibrillation , Induced Pluripotent Stem Cells , Humans , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Cell Line
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...