Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Proc Natl Acad Sci U S A ; 121(17): e2218204121, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38621141

ABSTRACT

Inherited arrhythmia syndromes (IASs) can cause life-threatening arrhythmias and are responsible for a significant proportion of sudden cardiac deaths (SCDs). Despite progress in the development of devices to prevent SCDs, the precise molecular mechanisms that induce detrimental arrhythmias remain to be fully investigated, and more effective therapies are desirable. In the present study, we screened a large-scale randomly mutagenized mouse library by electrocardiography to establish a disease model of IASs and consequently found one pedigree that exhibited spontaneous ventricular arrhythmias (VAs) followed by SCD within 1 y after birth. Genetic analysis successfully revealed a missense mutation (p.I4093V) of the ryanodine receptor 2 gene to be a cause of the arrhythmia. We found an age-related increase in arrhythmia frequency accompanied by cardiomegaly and decreased ventricular contractility in the Ryr2I4093V/+ mice. Ca2+ signaling analysis and a ryanodine binding assay indicated that the mutant ryanodine receptor 2 had a gain-of-function phenotype and enhanced Ca2+ sensitivity. Using this model, we detected the significant suppression of VA following flecainide or dantrolene treatment. Collectively, we established an inherited life-threatening arrhythmia mouse model from an electrocardiogram-based screen of randomly mutagenized mice. The present IAS model may prove feasible for use in investigating the mechanisms of SCD and assessing therapies.


Subject(s)
Tachycardia, Ventricular , Mice , Animals , Ryanodine Receptor Calcium Release Channel/metabolism , Arrhythmias, Cardiac/genetics , Flecainide , Mutation, Missense , Death, Sudden, Cardiac , Mutation
2.
Clin Sci (Lond) ; 136(24): 1831-1849, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36540030

ABSTRACT

Isorhamnetin, a natural flavonoid, has strong antioxidant and antifibrotic effects, and a regulatory effect against Ca2+-handling. Atrial remodeling due to fibrosis and abnormal intracellular Ca2+ activities contributes to initiation and persistence of atrial fibrillation (AF). The present study investigated the effect of isorhamnetin on angiotensin II (AngII)-induced AF in mice. Wild-type male mice (C57BL/6J, 8 weeks old) were assigned to three groups: (1) control group, (2) AngII-treated group, and (3) AngII- and isorhamnetin-treated group. AngII (1000 ng/kg/min) and isorhamnetin (5 mg/kg) were administered continuously via an implantable osmotic pump for two weeks and intraperitoneally one week before initiating AngII administration, respectively. AF induction and electrophysiological studies, Ca2+ imaging with isolated atrial myocytes and HL-1 cells, and action potential duration (APD) measurements using atrial tissue and HL-1 cells were performed. AF-related molecule expression was assessed and histopathological examination was performed. Isorhamnetin decreased AF inducibility compared with the AngII group and restored AngII-induced atrial effective refractory period prolongation. Isorhamnetin eliminated abnormal diastolic intracellular Ca2+ activities induced by AngII. Isorhamnetin also abrogated AngII-induced APD prolongation and abnormal Ca2+ loading in HL-1 cells. Furthermore, isorhamnetin strongly attenuated AngII-induced left atrial enlargement and atrial fibrosis. AngII-induced elevated expression of AF-associated molecules, such as ox-CaMKII, p-RyR2, p-JNK, p-ERK, and TRPC3/6, was improved by isorhamnetin treatment. The findings of the present study suggest that isorhamnetin prevents AngII-induced AF vulnerability and arrhythmogenic atrial remodeling, highlighting its therapeutic potential as an anti-arrhythmogenic pharmaceutical or dietary supplement.


Subject(s)
Atrial Fibrillation , Atrial Remodeling , Male , Mice , Animals , Atrial Fibrillation/drug therapy , Atrial Fibrillation/prevention & control , Calcium/metabolism , Mice, Inbred C57BL , Heart Atria/pathology , Myocytes, Cardiac/metabolism , Angiotensin II/metabolism
3.
Life Sci ; 306: 120807, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-35841977

ABSTRACT

AIMS: Dilated cardiomyopathy (DCM) remains among the most refractory heart diseases because of its complicated pathogenesis, and the key molecules that cause it remain unclear. MAIN METHODS: To elucidate the molecules and upstream pathways critical for DCM pathogenesis, we performed meta-analysis and co-expression analysis of RNA-sequencing (RNA-seq) datasets from publicly available databases. We analyzed three RNA-seq datasets containing comparisons of RNA expression in left ventricles between healthy controls and DCM patients. We extracted differentially expressed genes (DEGs) and clarified upstream regulators of cardiovascular disease-related DEGs by Ingenuity Pathway Analysis (IPA). Weighted Gene Co-expression Network Analysis (WGCNA) and Protein-Protein Interaction (PPI) analysis were also used to identify the hub gene candidates strongly associated with DCM. KEY FINDINGS: In total, 406 samples (184 healthy, 222 DCM) were used in this study. Overall, 391 DEGs [absolute fold change (FC) ≥ 1.5; P < 0.01], including 221 upregulated and 170 downregulated ones in DCM, were extracted. Seven common hub genes (LUM, COL1A2, CXCL10, FMOD, COL3A1, ADAMTS4, MRC1) were finally screened. IPA showed several upstream transcriptional regulators, including activating (NFKBIA, TP73, CALR, NFKB1, KLF4) and inhibiting (CEBPA, PPARGC1A) ones. We further validated increased expression of several common hub genes in the transverse aortic constriction-induced heart failure model. SIGNIFICANCE: In conclusion, meta-analysis and WGCNA using RNA-seq databases of DCM patients identified seven hub genes and seven upstream transcriptional regulators.


Subject(s)
Cardiomyopathy, Dilated , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Databases, Nucleic Acid , Gene Expression Profiling , Gene Regulatory Networks , Humans , RNA/genetics , Transcription Factors/genetics
4.
Front Cardiovasc Med ; 9: 1075358, 2022.
Article in English | MEDLINE | ID: mdl-36741841

ABSTRACT

Background: There is growing evidence indicating a close relationship between inflammation and atrial fibrillation (AF). Although underlying inflammatory atrial cardiomyopathy may contribute to the development of AF, the arrhythmogenic remodeling caused by atrial inflammation has not been elucidated in detail. Herein, we examined electrical, structural, and autonomic changes in the atria in a mouse model of autoimmune myocarditis. Methods: BALB/c mice were immunized with cardiac myosin peptide (MyHC-α614-629) conjugated with complete Freund's adjuvant on days 0 and 7. Susceptibility to AF was assessed using right-atrial burst pacing. Results: The mice immunized with MyHC-α614-629 showed an inflammatory atrial cardiomyopathy phenotype, with enlarged atria; a high degree of inflammatory cell infiltration primarily consisting of CD4+ T cells, CD8+ T cells, Ly6GlowCD11b+ macrophages, and CD11c+ dendritic cells; and severe interstitial fibrosis with collagen deposition. These mice demonstrated significantly enhanced susceptibility to AF, as indicated by their increased AF induction rate and duration. In addition, the expression of potassium channels (Kcnh2, Kcnd3, and Kcnj2) and calcium handling-associated genes (Cacna1c, Camk2, Ryr2, and Atp2a2) was downregulated. Connexin 40 expression was significantly downregulated, leading to frequent lateralization to the inflamed atrium. Sympathetic and parasympathetic innervation and neurotrophin expression (nerve growth factor and brain-derived neurotrophic factor) were upregulated in the inflamed atria. Conclusion: Inflammatory atrial cardiomyopathy promotes susceptibility to AF via arrhythmogenic electrical, structural, and autonomic remodeling of the atria.

6.
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
7.
Int J Mol Sci ; 22(3)2021 Jan 31.
Article in English | MEDLINE | ID: mdl-33572655

ABSTRACT

Programmed death ligand 2 (PD-L2) is the second ligand of programmed death 1 (PD-1) protein. In autoimmune myocarditis, the protective roles of PD-1 and its first ligand programmed death ligand 1 (PD-L1) have been well documented; however, the role of PD-L2 remains unknown. In this study, we report that PD-L2 deficiency exacerbates myocardial inflammation in mice with experimental autoimmune myocarditis (EAM). EAM was established in wild-type (WT) and PD-L2-deficient mice by immunization with murine cardiac myosin peptide. We found that PD-L2-deficient mice had more serious inflammatory infiltration in the heart and a significantly higher myocarditis severity score than WT mice. PD-L2-deficient dendritic cells (DCs) enhanced CD4+ T cell proliferation in the presence of T cell receptor and CD28 signaling. These data suggest that PD-L2 on DCs protects against autoreactive CD4+ T cell expansion and severe inflammation in mice with EAM.


Subject(s)
Autoimmune Diseases/immunology , B7-H1 Antigen/immunology , Myocarditis/immunology , Programmed Cell Death 1 Ligand 2 Protein/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Autoimmune Diseases/pathology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Dendritic Cells/immunology , Male , Mice , Myocarditis/pathology
8.
Int J Mol Sci ; 22(2)2021 Jan 13.
Article in English | MEDLINE | ID: mdl-33450865

ABSTRACT

Neutrophils are recruited into the heart at an early stage following a myocardial infarction (MI). These secrete several proteases, one of them being neutrophil elastase (NE), which promotes inflammatory responses in several disease models. It has been shown that there is an increase in NE activity in patients with MI; however, the role of NE in MI remains unclear. Therefore, the present study aimed to investigate the role of NE in the pathogenesis of MI in mice. NE expression peaked on day 1 in the infarcted hearts. In addition, NE deficiency improved survival and cardiac function post-MI, limiting fibrosis in the noninfarcted myocardium. Sivelestat, an NE inhibitor, also improved survival and cardiac function post-MI. Flow cytometric analysis showed that the numbers of heart-infiltrating neutrophils and inflammatory macrophages (CD11b+F4/80+CD206low cells) were significantly lower in NE-deficient mice than in wild-type (WT) mice. At the border zone between intact and necrotic areas, the number of terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive apoptotic cells was lower in NE-deficient mice than in WT mice. Western blot analyses revealed that the expression levels of insulin receptor substrate 1 and phosphorylation of Akt were significantly upregulated in NE-knockout mouse hearts, indicating that NE deficiency might improve cardiac survival by upregulating insulin/Akt signaling post-MI. Thus, NE may enhance myocardial injury by inducing an excessive inflammatory response and suppressing Akt signaling in cardiomyocytes. Inhibition of NE might serve as a novel therapeutic target in the treatment of MI.


Subject(s)
Leukocyte Elastase/deficiency , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardium/metabolism , Neutrophils/enzymology , Animals , Apoptosis/genetics , Biomarkers , Biopsy , Disease Models, Animal , Heart Function Tests , Insulins/metabolism , Leukocyte Elastase/metabolism , Mice , Mice, Knockout , Myocardial Infarction/mortality , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Ventricular Remodeling/genetics
9.
J Cell Mol Med ; 24(24): 14481-14490, 2020 12.
Article in English | MEDLINE | ID: mdl-33140535

ABSTRACT

Macrophages are fundamental components of inflammation in post-myocardial infarction (MI) and contribute to adverse cardiac remodelling and heart failure. However, the regulatory mechanisms in macrophage activation have not been fully elucidated. Previous studies showed that myeloid-associated immunoglobulin-like receptor II (MAIR-II) is involved in inflammatory responses in macrophages. However, its role in MI is unknown. Thus, this study aimed to determine a novel role and mechanism of MAIR-II in MI. We first identified that MAIR-II-positive myeloid cells were abundant from post-MI days 3 to 5 in infarcted hearts of C57BL/6J (WT) mice induced by permanent left coronary artery ligation. Compared to WT, MAIR-II-deficient (Cd300c2-/- ) mice had longer survival, ameliorated cardiac remodelling, improved cardiac function and smaller infarct sizes. Moreover, we detected lower pro-inflammatory cytokine and fibrotic gene expressions in Cd300c2-/- -infarcted hearts. These mice also had less infiltrating pro-inflammatory macrophages following MI. To elucidate a novel molecular mechanism of MAIR-II, we considered macrophage activation by Toll-like receptor (TLR) 9-mediated inflammation. In vitro, we observed that Cd300c2-/- bone marrow-derived macrophages stimulated by a TLR9 agonist expressed less pro-inflammatory cytokines compared to WT. In conclusion, MAIR-II may enhance inflammation via TLR9-mediated macrophage activation in MI, leading to adverse cardiac remodelling and poor prognosis.


Subject(s)
Macrophage Activation/genetics , Macrophages/metabolism , Myocardial Infarction/complications , Receptors, Polymeric Immunoglobulin/deficiency , Toll-Like Receptor 9/metabolism , Ventricular Remodeling/genetics , Animals , Biomarkers , Biopsy , Cytokines/metabolism , DNA, Mitochondrial , Disease Models, Animal , Disease Susceptibility , Echocardiography , Inflammation Mediators/metabolism , Kaplan-Meier Estimate , Macrophages/immunology , Mice , Mice, Knockout , Myocardial Infarction/etiology , Myocardial Infarction/mortality , Myocardium/pathology , Prognosis
10.
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
12.
JACC Clin Electrophysiol ; 5(3): 354-363, 2019 03.
Article in English | MEDLINE | ID: mdl-30898239

ABSTRACT

OBJECTIVES: This study sought to determine the feasibility of a novel simplified ablation protocol targeting only the pulmonary vein antrum using the radiofrequency hot-balloon catheter in patients with paroxysmal atrial fibrillation. BACKGROUND: Radiofrequency hot-balloon (RHB) catheter has been recently introduced into clinical practice for pulmonary vein isolation (PVI). The authors hypothesized that a novel simplified ablation protocol targeting only the PV antrum with energy application for a longer time (single-shot technique) could be an alternative approach to achieve PVI, while avoiding unnecessary energy application at the PV ostium. METHODS: A total of 61 consecutive paroxysmal atrial fibrillation patients (age 64.1 ± 10.9 years, 48 male) who underwent antrum RHB-PVI were enrolled. Energy applications were performed following the pre-specified protocol only targeting the PV antrum. If the PVI was not achieved after 2 energy applications using the RHB, a touch-up ablation was performed. RESULTS: Of 241 PV, including 3 left common PV, 194 (80%) were isolated exclusively using the RHB. The target PVI average per group of 15 consecutive procedures improved from 75% (initial 15) to 89% (last 16) of patients. The injected volume was greatest in the right superior PV (13.1 ± 2.0 ml) and the smallest in the left inferior PV (10.8 ± 1.1 ml), and 23 PV (9.5%) required over 15 ml (estimated balloon diameter of 30 mm). Periprocedural complications were noted in 3 patients (4.9%), but phrenic nerve injury was not observed. Sinus rhythm maintenance at 12-month follow-up was achieved in 57 patients (93%). CONCLUSIONS: A novel simplified antrum RHB-PVI appears to be a feasible technique for the treatment of paroxysmal atrial fibrillation.


Subject(s)
Atrial Fibrillation/surgery , Catheter Ablation , Aged , Cardiac Catheters , Catheter Ablation/adverse effects , Catheter Ablation/instrumentation , Catheter Ablation/methods , Catheter Ablation/statistics & numerical data , Female , Humans , Male , Middle Aged , Pulmonary Veins/surgery , Retrospective Studies , Treatment Outcome
13.
Intern Med ; 57(11): 1605-1609, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29434120

ABSTRACT

A 71-year-old woman was admitted with dyspnea. An electrocardiogram revealed ST-segment elevation, and echocardiography showed akinesis in the left ventricular apex with hyperkinesis of the base. Coronary angiography revealed no stenosis, and left ventriculography indicated ballooning of the left ventricular apex and apical ventricular septal perforation. We diagnosed the patient with Takotsubo syndrome complicated by ventricular septal perforation, which was surgically repaired. Although ventricular septal perforation is recognized as a life-threatening complication after acute myocardial infarction, it can also occur after Takotsubo syndrome. The early recognition and management of this condition can help prevent morbidity and mortality.


Subject(s)
Takotsubo Cardiomyopathy/complications , Ventricular Septal Rupture/etiology , Aged , Coronary Angiography , Echocardiography , Female , Humans , Takotsubo Cardiomyopathy/diagnosis , Ventricular Septal Rupture/diagnosis
14.
Heart Vessels ; 32(8): 916-925, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28220240

ABSTRACT

Although a previous randomized study showed that the use of atrial natriuretic peptide (carperitide) improved the long-term prognosis of patients with heart failure, its effect on short-term prognosis remains unclear. We retrospectively identified patients who were admitted to our tertiary-care center with acute decompensated heart failure (ADHF) between April 2009 and December 2013.We divided the eligible patients into two groups: patients who started receiving carperitide on the day of admission (carperitide group) and those who did not receive carperitide during hospitalization (control group). We compared the in-hospital mortality between the two groups using propensity scores derived from 40 baseline variables. We identified 879 eligible patients (mean age, 75.2 years; male, 56.7%), including 336 (38.2%) in the carperitide group and 543 (61.8%) in the control group. One-to-one propensity score matching created 177 pairs. Although the unmatched analysis found a significantly lower in-hospital mortality in the carperitide group than in the control group (3.3% vs. 7.9%, respectively, p = 0.005), the propensity score-matched analysis found no significant difference in in-hospital mortality between the two groups [4.0% vs. 5.1%, p = 0.609; risk difference, -1.1%, 95% confidence interval (CI), -5.5-3.2%]. Logistic regression analysis with adjustment for propensity scores also found no significant association between carperitide use and in-hospital mortality (adjusted odds ratio, 0.61; 95% CI, 0.29 to 1.28; p = 0.605). The present retrospective study showed that carperitide use as the initial treatment was not significantly associated with lower in-hospital mortality in patients with ADHF.


Subject(s)
Atrial Natriuretic Factor/administration & dosage , Heart Failure/drug therapy , Patient Admission/trends , Propensity Score , Aged , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Heart Failure/mortality , Heart Failure/physiopathology , Hospital Mortality/trends , Humans , Japan/epidemiology , Male , Prognosis , Retrospective Studies , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...