Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
1.
J Clin Neurosci ; 101: 239-243, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35640432

ABSTRACT

OBJECTIVE: To investigate the association between lipoprotein-associated phospholipase A2 (Lp-PLA2) concentration and the incidence of acute ischemic stroke (AIS) in patients with atrial fibrillation (AF). METHODS: A total of 257 patients admitted to the Kaifeng Central Hospital were enrolled in this study. Receiver operating characteristic (ROC) curve analysis and multivariate logistic regression analysis were used to determine the association between Lp-PLA2 and AIS in patients with AF. RESULTS: In AF group, plasma Lp-PLA2 concentrations were significantly higher in patients with AIS than in those without it (277.4 vs 155.1, p < 0.001). And in the group of AIS patients, patients with AF also had a significantly higher level of Lp-PLA2 concentration than those without (277.4 vs 204.2, p < 0.001). The analysis of the ROC curve showed a significant diagnostic value of Lp-PLA2 for the incidence of AIS in patients with AF (AUC = 0.840, 95% CI: 0.737-0.871, p < 0.001), and the optimal cut-off point was 220.5 ng/ml, with a sensitivity and specificity of 82.14% and 75.5%, respectively. All AF patients were divided into two subgroups: the high Lp-PLA2 group (≥220.5 ng/ml) and the low Lp-PLA2 group (<220.5 ng/ml). And multivariate logistic regression analysis showed that after adjustment of confounders, Lp-PLA2 (OR 12.48, 95%CI 5.73-27.16, p < 0.001) was independently associated with the incidence of AIS in patients with AF. CONCLUSIONS: Plasma Lp-PLA2 concentration was independently associated with the development of AIS in patients with AF. Lp-PLA2 is a potential biomarker for stratification of risk for AIS in patients with AF.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase , Atrial Fibrillation , Ischemic Stroke , Stroke , 1-Alkyl-2-acetylglycerophosphocholine Esterase/blood , Atrial Fibrillation/blood , Atrial Fibrillation/enzymology , Biomarkers/blood , Humans , Ischemic Stroke/blood , Ischemic Stroke/enzymology , ROC Curve , Risk Factors , Stroke/blood , Stroke/enzymology
2.
J Mol Cell Cardiol ; 168: 70-82, 2022 07.
Article in English | MEDLINE | ID: mdl-35489387

ABSTRACT

Atrial fibrillation (AF) is a major complication of type 2 diabetes mellitus (T2DM) and plays critical roles in the pathogenesis of atrial remodeling. However, the differentially expressed genes in atria during the development of AF induced by hyperglycemia have rarely been reported. Here, we showed time-dependent increased AF incidence and duration, atrial enlargement, inflammation, fibrosis, conduction time and action potential duration in db/db mice, a model of T2DM. RNA sequencing analysis showed that 2256 genes were differentially expressed in the atria at 12, 14 and 16 weeks. Gene Ontology analysis showed that these genes participate primarily in cell adhesion, cellular response to interferon-beta, immune system process, positive regulation of cell migration, ion transport and cellular response to interferon-gamma. Analysis of significant pathways revealed the IL-17 signaling pathway, TNF signaling pathway, MAPK signaling pathway, chemokine signaling pathway, and cAMP receptor signaling. Additionally, these differentially expressed genes were classified into 50 profiles by hierarchical clustering analysis. Twelve of these profiles were significant and comprised 1115 genes. Gene coexpression network analysis identified that mitogen-activated protein kinase 10 (MAPK10) was localized in the core of the gene network and was the most highly expressed gene at different time points. Knockdown of MAPK10 markedly attenuated DM-induced AF incidence, atrial inflammation, fibrosis, electrical disorder and apoptosis in db/db mice. In summary, the present findings revealed that many genes are involved in DM-induced AF and that MAPK10 plays a central role in this disease, indicating that strategies targeting MAPK10 may represent a potential therapeutic approach to treat DM-induced AF.


Subject(s)
Atrial Fibrillation , Atrial Remodeling , Diabetes Mellitus, Type 2 , Mitogen-Activated Protein Kinase 10 , Animals , Atrial Fibrillation/enzymology , Atrial Fibrillation/genetics , Atrial Fibrillation/pathology , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Fibrosis , Heart Atria/metabolism , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Mice , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 10/metabolism , RNA-Seq , Time Factors
3.
Clin Sci (Lond) ; 135(20): 2409-2422, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34386810

ABSTRACT

Oxidative stress could be a possible mechanism and a therapeutic target of atrial fibrillation (AF). However, the effects of the xanthine oxidase (XO) inhibition for AF remain to be fully elucidated. We investigated the effects of a novel XO inhibitor febuxostat on AF compared with allopurinol in hypertension rat model. Five-week-old Dahl salt-sensitive rats were fed either low-salt (LS) (0.3% NaCl) or high-salt (HS) (8% NaCl) diet. After 4 weeks of diet, HS diet rats were divided into three groups: orally administered to vehicle (HS-C), febuxostat (5 mg/kg/day) (HS-F), or allopurinol (50 mg/kg/day) (HS-A). After 4 weeks of treatment, systolic blood pressure (SBP) was significantly higher in HS-C than LS, and it was slightly but significantly decreased by treatment with each XO inhibitor. AF duration was significantly prolonged in HS-C compared with LS, and significantly suppressed in both HS-F and HS-A (LS; 5.8 ± 3.5 s, HS-C; 33.9 ± 23.7 s, HS-F; 15.0 ± 14.1 s, HS-A; 20.1 ± 11.9 s: P<0.05). Ca2+ spark frequency was obviously increased in HS-C rats and reduced in the XO inhibitor-treated rats, especially in HS-F group. Western blotting revealed that the atrial expression levels of Met281/282-oxidized Ca2+/Calmodulin-dependent kinase II (CaMKII) and Ser2814-phosphorylated ryanodine receptor 2 were significantly increased in HS-C, and those were suppressed in HS-F and HS-A. Decreased expression of gap junction protein connexin 40 in HS-C was partially restored by treatment with each XO inhibitor. In conclusion, XO inhibitor febuxostat, as well as allopurinol, could reduce hypertension-related increase in AF perpetuation by restoring Ca2+ handling and gap junction.


Subject(s)
Atrial Fibrillation/prevention & control , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Enzyme Inhibitors/pharmacology , Febuxostat/pharmacology , Hypertension/drug therapy , Myocytes, Cardiac/drug effects , Xanthine Oxidase/antagonists & inhibitors , Allopurinol/pharmacology , Animals , Atrial Fibrillation/enzymology , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Connexins/genetics , Connexins/metabolism , Disease Models, Animal , Fibrosis , Gap Junctions/drug effects , Gap Junctions/enzymology , Gap Junctions/pathology , Hypertension/enzymology , Hypertension/genetics , Hypertension/physiopathology , Male , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Oxidation-Reduction , Phosphorylation , Rats, Inbred Dahl , Ryanodine Receptor Calcium Release Channel/metabolism , Sodium Chloride, Dietary , Xanthine Oxidase/metabolism , Gap Junction alpha-5 Protein
4.
Basic Res Cardiol ; 116(1): 13, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33630168

ABSTRACT

Atrial fibrillation (AF) is associated with electrical remodeling, leading to cellular electrophysiological dysfunction and arrhythmia perpetuation. Emerging evidence suggests a key role for epigenetic mechanisms in the regulation of ion channel expression. Histone deacetylases (HDACs) control gene expression through deacetylation of histone proteins. We hypothesized that class I HDACs in complex with neuron-restrictive silencer factor (NRSF) determine atrial K+ channel expression. AF was characterized by reduced atrial HDAC2 mRNA levels and upregulation of NRSF in humans and in a pig model, with regional differences between right and left atrium. In vitro studies revealed inverse regulation of Hdac2 and Nrsf in HL-1 atrial myocytes. A direct association of HDAC2 with active regulatory elements of cardiac K+ channels was revealed by chromatin immunoprecipitation. Specific knock-down of Hdac2 and Nrsf induced alterations of K+ channel expression. Hdac2 knock-down resulted in prolongation of action potential duration (APD) in neonatal rat cardiomyocytes, whereas inactivation of Nrsf induced APD shortening. Potential AF-related triggers were recapitulated by experimental tachypacing and mechanical stretch, respectively, and exerted differential effects on the expression of class I HDACs and K+ channels in cardiomyocytes. In conclusion, HDAC2 and NRSF contribute to AF-associated remodeling of APD and K+ channel expression in cardiomyocytes via direct interaction with regulatory chromatin regions. Specific modulation of these factors may provide a starting point for the development of more individualized treatment options for atrial fibrillation.


Subject(s)
Action Potentials , Atrial Fibrillation/enzymology , Epigenesis, Genetic , Heart Atria/enzymology , Heart Rate , Histone Deacetylase 2/metabolism , Myocytes, Cardiac/enzymology , Potassium Channels/metabolism , Repressor Proteins/metabolism , Adult , Aged , Animals , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Atrial Remodeling , Case-Control Studies , Cell Line , Disease Models, Animal , Female , Heart Atria/physiopathology , Histone Deacetylase 2/genetics , Humans , Male , Middle Aged , Potassium Channels/genetics , Repressor Proteins/genetics , Sus scrofa , Time Factors
5.
Cardiovasc Res ; 117(11): 2354-2364, 2021 09 28.
Article in English | MEDLINE | ID: mdl-33483749

ABSTRACT

AIMS: Gp91-containing NADPH oxidases (NOX2) are a significant source of myocardial superoxide production. An increase in NOX2 activity accompanies atrial fibrillation (AF) induction and electrical remodelling in animal models and predicts incident AF in humans; however, a direct causal role for NOX2 in AF has not been demonstrated. Accordingly, we investigated whether myocardial NOX2 overexpression in mice (NOX2-Tg) is sufficient to generate a favourable substrate for AF and further assessed the effects of atorvastatin, an inhibitor of NOX2, on atrial superoxide production and AF susceptibility. METHODS AND RESULTS: NOX2-Tg mice showed a 2- to 2.5-fold higher atrial protein content of NOX2 compared with wild-type (WT) controls, which was associated with a significant (twofold) increase in NADPH-stimulated superoxide production (2-hydroxyethidium by HPLC) in left and right atrial tissue homogenates (P = 0.004 and P = 0.019, respectively). AF susceptibility assessed in vivo by transoesophageal atrial burst stimulation was modestly increased in NOX2-Tg compared with WT (probability of AF induction: 88% vs. 69%, respectively; P = 0.037), in the absence of significant alterations in AF duration, surface ECG parameters, and LV mass or function. Mechanistic studies did not support a role for NOX2 in promoting electrical or structural remodelling, as high-resolution optical mapping of atrial tissues showed no differences in action potential duration and conduction velocity between genotypes. In addition, we did not observe any genotype difference in markers of fibrosis and inflammation, including atrial collagen content and Col1a1, Il-1ß, Il-6, and Mcp-1 mRNA. Similarly, NOX2 overexpression did not have consistent effects on RyR2 Ca2+ leak nor did it affect PKA or CaMKII-mediated RyR2 phosphorylation. Finally, treatment with atorvastatin significantly inhibited atrial superoxide production in NOX2-Tg but had no effect on AF induction in either genotype. CONCLUSION: Together, these data indicate that while atrial NOX2 overexpression may contribute to atrial arrhythmogenesis, NOX2-derived superoxide production does not affect the electrical and structural properties of the atrial myocardium.


Subject(s)
Atrial Fibrillation/enzymology , Heart Atria/enzymology , Heart Rate , Myocytes, Cardiac/enzymology , NADPH Oxidase 2/biosynthesis , Action Potentials , Animals , Anti-Arrhythmia Agents/pharmacology , Atorvastatin/pharmacology , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Atrial Fibrillation/prevention & control , Disease Models, Animal , Enzyme Induction , Enzyme Inhibitors/pharmacology , Heart Atria/drug effects , Heart Atria/physiopathology , Mice, Transgenic , Myocytes, Cardiac/drug effects , NADPH Oxidase 2/antagonists & inhibitors , NADPH Oxidase 2/genetics , Signal Transduction , Superoxides/metabolism , Time Factors
6.
J Clin Invest ; 131(2)2021 01 19.
Article in English | MEDLINE | ID: mdl-33151911

ABSTRACT

Diabetes mellitus (DM) and atrial fibrillation (AF) are major unsolved public health problems, and diabetes is an independent risk factor for AF. However, the mechanism(s) underlying this clinical association is unknown. ROS and protein O-GlcNAcylation (OGN) are increased in diabetic hearts, and calmodulin kinase II (CaMKII) is a proarrhythmic signal that may be activated by ROS (oxidized CaMKII, ox-CaMKII) and OGN (OGN-CaMKII). We induced type 1 (T1D) and type 2 DM (T2D) in a portfolio of genetic mouse models capable of dissecting the role of ROS and OGN at CaMKII and global OGN in diabetic AF. Here, we showed that T1D and T2D significantly increased AF, and this increase required CaMKII and OGN. T1D and T2D both required ox-CaMKII to increase AF; however, we did not detect OGN-CaMKII or a role for OGN-CaMKII in diabetic AF. Collectively, our data affirm CaMKII as a critical proarrhythmic signal in diabetic AF and suggest ROS primarily promotes AF by ox-CaMKII, while OGN promotes AF by a CaMKII-independent mechanism(s). These results provide insights into the mechanisms for increased AF in DM and suggest potential benefits for future CaMKII and OGN targeted therapies.


Subject(s)
Atrial Fibrillation/enzymology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Diabetes Complications/enzymology , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 2/enzymology , Acylation , Animals , Atrial Fibrillation/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Diabetes Complications/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/genetics , Mice, Knockout , Oxidation-Reduction
7.
J Cardiovasc Transl Res ; 14(3): 492-502, 2021 06.
Article in English | MEDLINE | ID: mdl-32844365

ABSTRACT

Atrial fibrillation (AF) is common, yet there is no preventive therapy for AF. We tested the efficacy of AMP-activated protein kinase (AMPK) activators, metformin, and aspirin, in primary prevention of AF in cardiac-specific liver kinase B1 (LKB1) knockout (KO) mouse model of AF. Incidence of spontaneous AF was significantly reduced in treated KO mice with metformin (10 mg/kg/day) (8.3% in male and 10.3% in female) and aspirin (20 mg/kg/day) (29.4% in male and 21.4% in female) compared with untreated littermates (81% in male and 67% in female) at 8 weeks (p < 0.05). Prevention of AF was associated with activation of AMPK in treated mice and thereby improvement of mitochondrial function, gap junction proteins (connexin 40/43), and intra- and inter-cellular ultrastructure in atrial myocardium. Fibrosis was significantly less in treated mice atria. Pharmacological activation of AMPK is an effective upstream therapy for the primary prevention of AF in susceptible heart. Graphical abstract.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anti-Arrhythmia Agents/pharmacology , Aspirin/pharmacology , Atrial Fibrillation/prevention & control , Enzyme Activators/pharmacology , Heart Rate/drug effects , Metformin/pharmacology , Myocytes, Cardiac/drug effects , AMP-Activated Protein Kinases/genetics , Animals , Atrial Fibrillation/enzymology , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Atrial Remodeling/drug effects , Disease Models, Animal , Enzyme Activation , Female , Fibrosis , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Primary Prevention
8.
Cardiovasc Res ; 117(10): 2175-2185, 2021 08 29.
Article in English | MEDLINE | ID: mdl-33067609

ABSTRACT

Proper cardiac Ca2+ homeostasis is essential for normal excitation-contraction coupling. Perturbations in cardiac Ca2+ handling through altered kinase activity has been implicated in altered cardiac contractility and arrhythmogenesis. Thus, a better understanding of cardiac Ca2+ handling regulation is vital for a better understanding of various human disease processes. 'Striated muscle preferentially expressed protein kinase' (SPEG) is a member of the myosin light chain kinase family that is key for normal cardiac function. Work within the last 5 years has revealed that SPEG has a crucial role in maintaining normal cardiac Ca2+ handling through maintenance of transverse tubule formation and phosphorylation of junctional membrane complex proteins. Additionally, SPEG has been causally impacted in human genetic diseases such as centronuclear myopathy and dilated cardiomyopathy as well as in common acquired cardiovascular disease such as heart failure and atrial fibrillation. Given the rapidly emerging role of SPEG as a key cardiac Ca2+ regulator, we here present this review in order to summarize recent findings regarding the mechanisms of SPEG regulation of cardiac excitation-contraction coupling in both physiology and human disease. A better understanding of the roles of SPEG will be important for a more complete comprehension of cardiac Ca2+ regulation in physiology and disease.


Subject(s)
Calcium/metabolism , Excitation Contraction Coupling , Heart Diseases/enzymology , Muscle Proteins/metabolism , Myocardial Contraction , Myocytes, Cardiac/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Atrial Fibrillation/enzymology , Atrial Fibrillation/genetics , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Genetic Predisposition to Disease , Heart Diseases/genetics , Heart Diseases/pathology , Heart Diseases/physiopathology , Heart Failure/enzymology , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Homeostasis , Humans , Muscle Proteins/genetics , Mutation , Myocytes, Cardiac/pathology , Protein Serine-Threonine Kinases/genetics
9.
Arch Pharm Res ; 43(12): 1276-1296, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33245518

ABSTRACT

A wide range of histone deacetylase (HDAC) inhibitors have been studied for their therapeutic potential because the excessive activity and expression of HDACs have been implicated in the pathogenesis of cardiac diseases. An increasing number of preclinical studies have demonstrated the cardioprotective effects of numerous HDAC inhibitors, suggesting a wide variety of mechanisms by which the inhibitors protect against cardiac stress, such as the suppression of cardiac fibrosis and fetal gene expression, enhancement of angiogenesis and mitochondrial biogenesis, prevention of electrical remodeling, and regulation of apoptosis, autophagy, and cell cycle arrest. For the development of isoform-selective HDAC inhibitors with high efficacy and low toxicity, it is important to identify and understand the mechanisms responsible for the effects of the inhibitors. This review highlights the preclinical effects of HDAC inhibitors that act against Zn2+-dependent HDACs and the underlying mechanisms of their protective effects against cardiac hypertrophy, hypertension, myocardial infarction, heart failure, and atrial fibrillation.


Subject(s)
Atrial Fibrillation/drug therapy , Cardiomegaly/drug therapy , Cardiovascular Agents/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Hypertension/drug therapy , Myocardial Infarction/drug therapy , Myocardium/enzymology , Animals , Antihypertensive Agents/therapeutic use , Atrial Fibrillation/enzymology , Atrial Fibrillation/physiopathology , Blood Pressure/drug effects , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiovascular Agents/adverse effects , Fibrosis , Heart Rate/drug effects , Histone Deacetylase Inhibitors/adverse effects , Humans , Hypertension/enzymology , Hypertension/physiopathology , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Signal Transduction , Ventricular Remodeling/drug effects
10.
Circulation ; 142(25): 2443-2455, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33092403

ABSTRACT

BACKGROUND: Ibrutinib is a Bruton tyrosine kinase inhibitor with remarkable efficacy against B-cell cancers. Ibrutinib also increases the risk of atrial fibrillation (AF), which remains poorly understood. METHODS: We performed electrophysiology studies on mice treated with ibrutinib to assess inducibility of AF. Chemoproteomic analysis of cardiac lysates identified candidate ibrutinib targets, which were further evaluated in genetic mouse models and additional pharmacological experiments. The pharmacovigilance database, VigiBase, was queried to determine whether drug inhibition of an identified candidate kinase was associated with increased reporting of AF. RESULTS: We demonstrate that treatment of mice with ibrutinib for 4 weeks results in inducible AF, left atrial enlargement, myocardial fibrosis, and inflammation. This effect was reproduced in mice lacking Bruton tyrosine kinase, but not in mice treated with 4 weeks of acalabrutinib, a more specific Bruton tyrosine kinase inhibitor, demonstrating that AF is an off-target side effect. Chemoproteomic profiling identified a short list of candidate kinases that was narrowed by additional experimentation leaving CSK (C-terminal Src kinase) as the strongest candidate for ibrutinib-induced AF. Cardiac-specific Csk knockout in mice led to increased AF, left atrial enlargement, fibrosis, and inflammation, phenocopying ibrutinib treatment. Disproportionality analyses in VigiBase confirmed increased reporting of AF associated with kinase inhibitors blocking Csk versus non-Csk inhibitors, with a reporting odds ratio of 8.0 (95% CI, 7.3-8.7; P<0.0001). CONCLUSIONS: These data identify Csk inhibition as the mechanism through which ibrutinib leads to AF. Registration: URL: https://ww.clinicaltrials.gov; Unique identifier: NCT03530215.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/toxicity , Atrial Fibrillation/chemically induced , Atrial Function, Left/drug effects , CSK Tyrosine-Protein Kinase/antagonists & inhibitors , Heart Atria/drug effects , Heart Rate/drug effects , Piperidines/toxicity , Protein Kinase Inhibitors/toxicity , Action Potentials/drug effects , Adenine/toxicity , Agammaglobulinaemia Tyrosine Kinase/deficiency , Agammaglobulinaemia Tyrosine Kinase/genetics , Animals , Atrial Fibrillation/enzymology , Atrial Fibrillation/physiopathology , CSK Tyrosine-Protein Kinase/genetics , CSK Tyrosine-Protein Kinase/metabolism , Databases, Genetic , Heart Atria/enzymology , Heart Atria/physiopathology , Humans , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Risk Assessment , Risk Factors
11.
Circ Res ; 127(8): 1036-1055, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32762493

ABSTRACT

RATIONALE: Postoperative atrial fibrillation (POAF) is a common and troublesome complication of cardiac surgery. POAF is generally believed to occur when postoperative triggers act on a preexisting vulnerable substrate, but the underlying cellular and molecular mechanisms are largely unknown. OBJECTIVE: To identify cellular POAF mechanisms in right atrial samples from patients without a history of atrial fibrillation undergoing open-heart surgery. METHODS AND RESULTS: Multicellular action potentials, membrane ion-currents (perforated patch-clamp), or simultaneous membrane-current (ruptured patch-clamp) and [Ca2+]i-recordings in atrial cardiomyocytes, along with protein-expression levels in tissue homogenates or cardiomyocytes, were assessed in 265 atrial samples from patients without or with POAF. No indices of electrical, profibrotic, or connexin remodeling were noted in POAF, but Ca2+-transient amplitude was smaller, although spontaneous sarcoplasmic reticulum (SR) Ca2+-release events and L-type Ca2+-current alternans occurred more frequently. CaMKII (Ca2+/calmodulin-dependent protein kinase-II) protein-expression, CaMKII-dependent phosphorylation of the cardiac RyR2 (ryanodine-receptor channel type-2), and RyR2 single-channel open-probability were significantly increased in POAF. SR Ca2+-content was unchanged in POAF despite greater SR Ca2+-leak, with a trend towards increased SR Ca2+-ATPase activity. Patients with POAF also showed stronger expression of activated components of the NLRP3 (NACHT, LRR, and PYD domains-containing protein-3)-inflammasome system in atrial whole-tissue homogenates and cardiomyocytes. Acute application of interleukin-1ß caused NLRP3-signaling activation and CaMKII-dependent RyR2/phospholamban hyperphosphorylation in an immortalized mouse atrial cardiomyocyte cell-line (HL-1-cardiomyocytes) and enhanced spontaneous SR Ca2+-release events in both POAF cardiomyocytes and HL-1-cardiomyocytes. Computational modeling showed that RyR2 dysfunction and increased SR Ca2+-uptake are sufficient to reproduce the Ca2+-handling phenotype and indicated an increased risk of proarrhythmic delayed afterdepolarizations in POAF subjects in response to interleukin-1ß. CONCLUSIONS: Preexisting Ca2+-handling abnormalities and activation of NLRP3-inflammasome/CaMKII signaling are evident in atrial cardiomyocytes from patients who subsequently develop POAF. These molecular substrates sensitize cardiomyocytes to spontaneous Ca2+-releases and arrhythmogenic afterdepolarizations, particularly upon exposure to inflammatory mediators. Our data reveal a potential cellular and molecular substrate for this important clinical problem.


Subject(s)
Atrial Fibrillation/etiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiac Surgical Procedures/adverse effects , Heart Atria/enzymology , Heart Rate , Inflammasomes/metabolism , Myocytes, Cardiac/enzymology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Action Potentials , Aged , Animals , Atrial Fibrillation/enzymology , Atrial Fibrillation/physiopathology , Calcium Signaling , Case-Control Studies , Cell Line , Female , Heart Atria/physiopathology , Humans , Inflammation Mediators/metabolism , Male , Mice , Middle Aged , Phosphorylation , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism
12.
Circulation ; 142(13): 1261-1278, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32686471

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) is the most common heart rhythm disorder in adults and a major cause of stroke. Unfortunately, current treatments of AF are suboptimal because they are not targeted to the molecular mechanisms underlying AF. Using a highly novel gene therapy approach in a canine, rapid atrial pacing model of AF, we demonstrate that NADPH oxidase 2 (NOX2) generated oxidative injury causes upregulation of a constitutively active form of acetylcholine-dependent K+ current (IKACh), called IKH; this is an important mechanism underlying not only the genesis, but also the perpetuation of electric remodeling in the intact, fibrillating atrium. METHODS: To understand the mechanism by which oxidative injury promotes the genesis and maintenance of AF, we performed targeted injection of NOX2 short hairpin RNA (followed by electroporation to facilitate gene delivery) in atria of healthy dogs followed by rapid atrial pacing. We used in vivo high-density electric mapping, isolation of atrial myocytes, whole-cell patch clamping, in vitro tachypacing of atrial myocytes, lucigenin chemiluminescence assay, immunoblotting, real-time polymerase chain reaction, immunohistochemistry, and Masson trichrome staining. RESULTS: First, we demonstrate that generation of oxidative injury in atrial myocytes is a frequency-dependent process, with rapid pacing in canine atrial myocytes inducing oxidative injury through the induction of NOX2 and the generation of mitochondrial reactive oxygen species. We show that oxidative injury likely contributes to electric remodeling in AF by upregulating IKACh by a mechanism involving frequency-dependent activation of PKCε (protein kinase C epsilon). The time to onset of nonsustained AF increased by >5-fold in NOX2 short hairpin RNA-treated dogs. Furthermore, animals treated with NOX2 short hairpin RNA did not develop sustained AF for up to 12 weeks. The electrophysiological mechanism underlying AF prevention was prolongation of atrial effective refractory periods, at least in part attributable to the attenuation of IKACh. Attenuated membrane translocation of PKCε appeared to be a likely molecular mechanism underlying this beneficial electrophysiological remodeling. CONCLUSIONS: NOX2 oxidative injury (1) underlies the onset, and the maintenance of electric remodeling in AF, as well, and (2) can be successfully prevented with a novel, gene-based approach. Future optimization of this approach may lead to a novel, mechanism-guided therapy for AF.


Subject(s)
Atrial Fibrillation , Atrial Remodeling , Gene Expression Regulation, Enzymologic , Genetic Therapy , NADPH Oxidase 2 , RNA, Small Interfering , Animals , Atrial Fibrillation/enzymology , Atrial Fibrillation/genetics , Atrial Fibrillation/physiopathology , Atrial Fibrillation/therapy , Dogs , Heart Atria/enzymology , Heart Atria/physiopathology , NADPH Oxidase 2/biosynthesis , NADPH Oxidase 2/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
13.
Int J Mol Sci ; 21(13)2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32629939

ABSTRACT

Aging and obesity are the most prominent risk factors for onset of atrial fibrillation (AF). Nicotinamide phosphoribosyltransferase (Nampt) is the rate-limiting enzyme that catalyzes nicotinamide adenine dinucleotide (NAD) activity. Nampt and NAD are essential for maintenance of cellular redox homeostasis and modulation of cellular metabolism, and their expression levels decrease with aging and obesity. However, a role for Nampt in AF is unknown. The present study aims to test whether there is a role of Nampt/NAD axis in the pathogenesis of obesity-induced AF. Male C57BL/6J (WT) mice and heterozygous Nampt knockout (NKO) mice were fed with a normal chow diet (ND) or a high-fat diet (HFD). Electrophysiological study showed that AF inducibility was significantly increased in WT+HFD, NKO+ND, and NKO+HFD mice compared with WT+ND mice. AF duration was significantly longer in WT+HFD and NKO+ND mice and further prolonged in NKO+HFD mice compared with WT+ND mice and the calcium handling pathway was altered on molecular level. Also, treatment with nicotinamide riboside, a NAD precursor, partially restored the HFD-induced AF perpetuation. Overall, this work demonstrates that partially deletion of Nampt facilitated HFD-induced AF through increased diastolic calcium leaks. The Nampt/NAD axis may be a potent therapeutic target for AF.


Subject(s)
Atrial Fibrillation/enzymology , NAD/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Animals , Atrial Fibrillation/etiology , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Diet, High-Fat/adverse effects , Heart Atria/enzymology , Male , Mice, Knockout , Obesity/complications , Ryanodine Receptor Calcium Release Channel/metabolism
14.
Heart Vessels ; 35(8): 1116-1124, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32206865

ABSTRACT

Chymase is an angiotensin II-forming serine proteinase and elevation of its tissue activity occurs in various cardiovascular diseases. Several authors have suggested that there is an association between the renin-angiotensin system and atrial fibrillation (AF). Chymase-dependent angiotensin II-forming activity in circulating mononuclear leukocytes (CML chymase dAIIFA) was investigated in patients with AF and patients in sinus rhythm. Consecutive outpatients were recruited at our hospital. CML chymase dAIIFA was measured using a Nma/Dnp-type fluorescence-quenching substrate of modified angiotensin I in the presence or absence of a specific serine proteinase inhibitor. To search the independent contributing factor of existence of AF, the analysis between groups was carried out using multivariate analysis after univariate analysis. The patients were classified into a sinus rhythm (SR) group (n = 459) or an AF group (n = 48). CML chymase dAIIFA was significantly higher in the AF group (622 pmol/min/mg) compared with the SR group (488 pmol/min/mg) (p < 0.001). Logistic regression analysis revealed that high CML chymase dAIIFA was an independent determinant of the existence of AF (p < 0.001). Elevation of CML chymase dAIIFA was associated with AF. Activation of chymase might be linked to atrial structural and electrical remodeling.


Subject(s)
Angiotensin II/blood , Atrial Fibrillation/enzymology , Chymases/blood , Leukocytes, Mononuclear/enzymology , Action Potentials , Aged , Aged, 80 and over , Atrial Fibrillation/blood , Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Atrial Remodeling , Biomarkers/blood , Case-Control Studies , Cross-Sectional Studies , Female , Heart Rate , Humans , Male , Middle Aged
15.
BMC Cardiovasc Disord ; 20(1): 50, 2020 02 03.
Article in English | MEDLINE | ID: mdl-32013885

ABSTRACT

BACKGROUND: Previous studies demonstrated impaired lipid metabolism and augmented aerobic glycolysis in AF. The authors aimed to investigate whether the use of metformin, an AMPK activator, could reverse this metabolic remodeling in chronic AF and to explore the underlying mechanisms. METHODS: We conducted chronic AF animal models with 18 beagle dogs and divided them into SR (pacemaker implanted without pacing), AF (pacemaker implanted with sustained pacing at a frequency of 400 beats/min for 6 weeks), and metformin+AF group (daily oral administration of metformin was initiated 1 week before surgery and continued throughout the study period). After electrophysiological measurements, the left atrial appendage tissue samples were taken from the beating heart for further analysis. Protein expression, histological analysis, and biochemical measurements were conducted. RESULTS: The AF groups showed decreased expression of FAT/CD36, CPT-1, VLCAD, increased concentration of free fatty acid and triglyceride, and increased lipid deposition. The activation of AMPK/PGC-1α/PPARα pathway was decreased. The key factors of the Warburg effect, including HIF-1α, GLUT-1, PDK1, HK, and LDH, increased in AF group compared to SR group. The expression of PDH decreased significantly, accompanied by increased atrial lactate production. The extent of fibrosis increased significantly in the left atrial appendage of AF group. dERP, ∑WOV, and AF inducibility increased while ERP decreased in AF group compared to SR group. The use of metformin attenuated all these changes effectively. CONCLUSIONS: Metformin improves lipid metabolism and reverses the Warburg effect in chronic AF via AMPK activation. It attenuates atrial electrical and structural remodeling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Atrial Appendage/drug effects , Atrial Fibrillation/drug therapy , Energy Metabolism/drug effects , Enzyme Activators/pharmacology , Lipid Metabolism/drug effects , Metformin/pharmacology , Animals , Atrial Appendage/enzymology , Atrial Appendage/physiopathology , Atrial Fibrillation/enzymology , Atrial Fibrillation/physiopathology , Atrial Function, Left/drug effects , Atrial Remodeling/drug effects , Chronic Disease , Disease Models, Animal , Dogs , Enzyme Activation , Heart Rate/drug effects , Male
16.
Biochim Biophys Acta Mol Cell Res ; 1867(3): 118459, 2020 03.
Article in English | MEDLINE | ID: mdl-30880147

ABSTRACT

Perpetuation of atrial fibrillation (AF) is caused by electropathology, which is defined as impairment of electrical activation caused by structural and metabolic remodeling of cardiomyocytes. We previously dissected the molecular mechanisms underlying electropathology and identified an important role for histone deacetylases (HDACs). HDACs catalyze the removal of acetyl-groups from lysine residues within nucleosomal histone tails and many non-histone proteins. Various HDAC inhibitors are efficacious in attenuating electropathology, and improve contractile function in experimental AF. Emerging evidence reveals novel mechanisms by which HDAC inhibitors prevent cardiac electropathology and thereby benefit the heart during AF. These mechanisms include post-translational modification of contractile and structural proteins and changes in gene expression. In this review paper, we summarize recent findings on novel functions of zinc-dependent HDACs in electropathology and discuss the potential for pharmacological HDAC inhibition as a strategy to treat AF.


Subject(s)
Atrial Fibrillation/drug therapy , Heart/physiopathology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/genetics , Atrial Fibrillation/enzymology , Atrial Fibrillation/genetics , Atrial Fibrillation/pathology , Gene Expression Regulation/genetics , Heart/diagnostic imaging , Histones/genetics , Humans , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Nucleosomes/genetics , Protein Processing, Post-Translational/genetics
17.
Clin Res Cardiol ; 109(3): 324-330, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31236695

ABSTRACT

BACKGROUND: Myeloperoxidase (MPO), secreted by neutrophils under inflammatory conditions, is elevated in atrial fibrillation (AF). MPO may be involved in atrial remodeling that underpins AF progression characterized by a switch from paroxysmal to persistent AF and the formation of low-voltage areas (LVA). MPO levels are modulated by renin-angiotensin system antagonists (RAS-A), commonly used to treat AF comorbidities, and are associated with reduced AF incidence, implicating a potential link. OBJECTIVE: We investigated MPO levels in progressing AF in peripheral and left atrial (LA) blood and analyzed a potential effect of RAS-A. METHODS: Samples of AF patients were collected from the femoral vein and the LA during catheter ablation (n = 121) and at follow-up (n = 23). No-AF probands (n = 37) served as controls. MPO was determined using commercial ELISA. RESULTS: MPO levels were significantly increased in AF patients compared to controls (median, 27.7 ng/ml (IQR 14.3-66.6) versus 12.6 (IQR 9.9-17.7), p < 0.001), without differences between clinical AF progression phenotypes. MPO concentration was tenfold higher in LA than periphery (279.2 ng/ml (IQR 202.2-342.9) versus 27.7 ng/ml (IQR 14.3-65.9), p < 0.001). MPO remained increased at midterm follow-up irrespective of rhythm outcome. RAS-A was associated with significantly lower peripheral (22.2 ng/ml (IQR 12.7-48.2) versus 37.1 ng/ml (IQR 18.2-85.2), p < 0.05) MPO levels in AF patients. CONCLUSION: The pro-fibrotic enzyme MPO is generally elevated in AF patients irrespective of AF type, the presence of LVA or midterm rhythm outcome. Our data suggest that MPO may directly originate from the LA. RAS-A decrease peripheral MPO levels in AF patients.


Subject(s)
Atrial Fibrillation/physiopathology , Catheter Ablation , Peroxidase/metabolism , Renin-Angiotensin System/drug effects , Adult , Aged , Atrial Fibrillation/enzymology , Atrial Remodeling/physiology , Cohort Studies , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Humans , Male , Middle Aged
18.
J Cardiovasc Pharmacol Ther ; 24(6): 575-585, 2019 11.
Article in English | MEDLINE | ID: mdl-31159577

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) is an important and growing clinical problem. Current pharmacological treatments are unsatisfactory. Electrical remodeling has been identified as one of the principal pathophysiological mechanisms that promote AF, but there are no effective therapies to prevent or correct electrical remodeling in patients with AF. In AF, cardiac production and circulating levels of B-type natriuretic peptide (BNP) are increased. However, its functional significance in AF remains to be determined. We assessed the hypotheses that chronic BNP treatment may prevent the altered electrophysiology in AF, and preventing AF-induced activation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) may play a role. METHODS AND RESULTS: Forty-four rabbits were randomly divided into sham, rapid atrial pacing (RAP at 600 beats/min for 3 weeks), RAP/BNP, and sham/BNP groups. Rabbits in the RAP/BNP and sham/BNP groups received subcutaneous BNP (20 µg/kg twice daily) during the 3-week study period. HL-1 cells were subjected to rapid field stimulation for 24 hours in the presence or absence of BNP, KN-93 (a CaMKII inhibitor), or KN-92 (a nonactive analog of KN-93). We compared atrial electrical remodeling-related alterations in the ion channel/function/expression of these animals. We found that only in the RAP group, AF inducibility was significantly increased, atrial effective refractory periods and action potential duration were reduced, and the density of ICa, L and Ito decreased, while IK1 increased. The changes in the expressions of Cav1.2, Kv4.3, and Kir2.1 and currents showed a similar trend. In addition, in the RAP group, the activation of CaMKIIδ and phosphorylation of ryanodine receptor 2 and phospholamban significantly increased. Importantly, these changes were prevented in the RAP/BNP group, which were further validated by in vitro studies. CONCLUSIONS: Chronic BNP therapy prevents atrial electrical remodeling in AF. Inhibition of CaMKII activation plays an important role to its anti-AF efficacy in this model.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/drug therapy , Atrial Remodeling/drug effects , Heart Atria/drug effects , Heart Rate/drug effects , Myocytes, Cardiac/drug effects , Natriuretic Peptide, Brain/pharmacology , Action Potentials , Animals , Atrial Fibrillation/enzymology , Atrial Fibrillation/physiopathology , Calcium Channels, L-Type/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line , Disease Models, Animal , Heart Atria/enzymology , Heart Atria/physiopathology , Mice , Myocytes, Cardiac/enzymology , Phosphorylation , Potassium Channels, Inwardly Rectifying/metabolism , Rabbits , Ryanodine Receptor Calcium Release Channel/metabolism , Shal Potassium Channels/metabolism
19.
Med Sci Monit ; 25: 2687-2694, 2019 Apr 12.
Article in English | MEDLINE | ID: mdl-30977468

ABSTRACT

BACKGROUND Research interest in endothelial nitric oxide synthase(eNOS) polymorphisms and atrial fibrillation (AF) has grown in last recent years, but the results of individual studies are inconsistent due to their small sample sizes. MATERIAL AND METHODS We searched databases for eligible studies on eNOS and AF, extracted the relevant data, and rigorously screened them according to inclusion and exclusion criteria. Then, we evaluated the study quality according to the Newcastle-Ottawa scale score, and we pooled the odds ratios (ORs) and 95% confidence intervals (CIs) by using a random-effects model or fixed-effects model based on inter-study heterogeneity. In addition, we performed subgroup analysis and sensitivity analysis and assessed publication bias. RESULTS According to the inclusion and exclusion criteria, we finally found 8 studies in this search. The recessive (OR=0.81; 95% CI=0.67 to 0.97; p=0.988; I²=0.0%) model showed that the eNOS 786T/C polymorphism was relevant to AF. We also found that the eNOS 786T/C polymorphism decreases the risk of AF, especially in white people (OR=0.81; 95% CI=0.67 to 0.97; P=0.023 for recessive model) and in the control population (OR=0.79; 95% CI=0.65 to 0.97; P=0.022 for recessive model). We found no obvious publication bias. CONCLUSIONS The eNOS gene loci 786T/C polymorphism is relevant to the risk of AF. Our results suggest that the 786T/C polymorphism significantly decreases AF risks in white people and control populations. Larger studies are required for further evaluation.


Subject(s)
Atrial Fibrillation/enzymology , Atrial Fibrillation/genetics , Genetic Predisposition to Disease , Nitric Oxide Synthase Type III/genetics , Polymorphism, Single Nucleotide/genetics , Alleles , Case-Control Studies , Humans , Models, Genetic , Publication Bias , Risk Factors
20.
Circ Arrhythm Electrophysiol ; 12(3): e007071, 2019 03.
Article in English | MEDLINE | ID: mdl-30879335

ABSTRACT

BACKGROUND: A structural, electrical and metabolic atrial remodeling is central in the development of atrial fibrillation (AF) contributing to its initiation and perpetuation. In the heart, HDACs (histone deacetylases) control remodeling associated processes like hypertrophy, fibrosis, and energy metabolism. Here, we analyzed, whether the HDAC class I/IIa inhibitor valproic acid (VPA) is able to attenuate atrial remodeling in CREM-IbΔC-X (cAMP responsive element modulator isoform IbΔC-X) transgenic mice, a mouse model of extensive atrial remodeling with age-dependent progression from spontaneous atrial ectopy to paroxysmal and finally long-lasting AF. METHODS: VPA was administered for 7 or 25 weeks to transgenic and control mice. Atria were analyzed macroscopically and using widefield and electron microscopy. Action potentials were recorded from atrial cardiomyocytes using patch-clamp technique. ECG recordings documented the onset of AF. A proteome analysis with consecutive pathway mapping identified VPA-mediated proteomic changes and related pathways. RESULTS: VPA attenuated many components of atrial remodeling that are present in transgenic mice, animal AF models, and human AF. VPA significantly ( P<0.05) reduced atrial dilatation, cardiomyocyte enlargement, atrial fibrosis, and the disorganization of myocyte's ultrastructure. It significantly reduced the occurrence of atrial thrombi, reversed action potential alterations, and finally delayed the onset of AF by 4 to 8 weeks. Increased histone H4-acetylation in atria from VPA-treated transgenic mice verified effective in vivo HDAC inhibition. Cardiomyocyte-specific genetic inactivation of HDAC2 in transgenic mice attenuated the ultrastructural disorganization of myocytes comparable to VPA. Finally, VPA restrained dysregulation of proteins in transgenic mice that are involved in a multitude of AF relevant pathways like oxidative phosphorylation or RhoA (Ras homolog gene family, member A) signaling and disease functions like cardiac fibrosis and apoptosis of muscle cells. CONCLUSIONS: Our results suggest that VPA, clinically available, well-tolerated, and prescribed to many patients for years, has the therapeutic potential to delay the development of atrial remodeling and the onset of AF in patients at risk.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/prevention & control , Atrial Remodeling/drug effects , Heart Atria/drug effects , Histone Deacetylase Inhibitors/pharmacology , Myocytes, Cardiac/drug effects , Valproic Acid/pharmacology , Action Potentials , Animals , Atrial Fibrillation/enzymology , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Cyclic AMP Response Element Modulator/genetics , Disease Models, Animal , Heart Atria/enzymology , Heart Atria/physiopathology , Heart Atria/ultrastructure , Heart Rate , Male , Mice, Transgenic , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...