Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Sci Rep ; 14(1): 15422, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965264

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is an inherited disorder characterized by left ventricular hypertrophy and diastolic dysfunction, and increases the risk of arrhythmias and heart failure. Some patients with HCM develop a dilated phase of hypertrophic cardiomyopathy (D-HCM) and have poor prognosis; however, its pathogenesis is unclear and few pathological models exist. This study established disease-specific human induced pluripotent stem cells (iPSCs) from a patient with D-HCM harboring a mutation in MYBPC3 (c.1377delC), a common causative gene of HCM, and investigated the associated pathophysiological mechanisms using disease-specific iPSC-derived cardiomyocytes (iPSC-CMs). We confirmed the expression of pluripotent markers and the ability to differentiate into three germ layers in D-HCM patient-derived iPSCs (D-HCM iPSCs). D-HCM iPSC-CMs exhibited disrupted myocardial sarcomere structures and an increased number of damaged mitochondria. Ca2+ imaging showed increased abnormal Ca2+ signaling and prolonged decay time in D-HCM iPSC-CMs. Cell metabolic analysis revealed increased basal respiration, maximal respiration, and spare-respiratory capacity in D-HCM iPSC-CMs. RNA sequencing also showed an increased expression of mitochondrial electron transport system-related genes. D-HCM iPSC-CMs showed abnormal Ca2+ handling and hypermetabolic state, similar to that previously reported for HCM patient-derived iPSC-CMs. Although further studies are required, this is expected to be a useful pathological model for D-HCM.


Subject(s)
Calcium , Cardiomyopathy, Hypertrophic , Carrier Proteins , Frameshift Mutation , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Induced Pluripotent Stem Cells/metabolism , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Calcium/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Calcium Signaling , Cell Differentiation , Male
2.
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
3.
Circ J ; 88(5): 751-759, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38462534

ABSTRACT

BACKGROUND: In the present study, we aimed to investigate whether early cardiac biomarker alterations and echocardiographic parameters, including left atrial (LA) strain, can predict anthracycline-induced cardiotoxicity (AIC) and thus develop a predictive risk score.Methods and Results: The AIC registry is a prospective, observational cohort study designed to gather serial echocardiographic and biomarker data before and after anthracycline chemotherapy. Cardiotoxicity was defined as a reduction in left ventricular ejection fraction (LVEF) ≥10 percentage points from baseline and <55%. In total, 383 patients (93% women; median age, 57 [46-66] years) completed the 2-year follow-up; 42 (11.0%) patients developed cardiotoxicity (median time to onset, 292 [175-440] days). Increases in cardiac troponin T (TnT) and B-type natriuretic peptide (BNP) and relative reductions in the left ventricular global longitudinal strain (LV GLS) and LA reservoir strain [LASr] at 3 months after anthracycline administration were independently associated with subsequent cardiotoxicity. A risk score containing 2 clinical variables (smoking and prior cardiovascular disease), 2 cardiac biomarkers at 3 months (TnT ≥0.019 ng/mL and BNP ≥31.1 pg/mL), 2 echocardiographic variables at 3 months (relative declines in LV GLS [≥6.5%], and LASr [≥7.5%]) was generated. CONCLUSIONS: Early decline in LASr was independently associated with subsequent cardiotoxicity. The AIC risk score may provide useful prognostication in patients receiving anthracyclines.


Subject(s)
Anthracyclines , Cardiotoxicity , Natriuretic Peptide, Brain , Humans , Anthracyclines/adverse effects , Middle Aged , Female , Male , Prospective Studies , Aged , Natriuretic Peptide, Brain/blood , Biomarkers/blood , Troponin T/blood , Echocardiography , Registries , Early Diagnosis
4.
Ann Surg Oncol ; 30(2): 711-721, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36273057

ABSTRACT

BACKGROUND: Perioperative atrial fibrillation is a common postoperative complication. Adverse consequences associated with POAF include hemodynamic instability, increased risk of stroke, extended hospital stays, and increased mortality. METHODS: To determine the risk factors for POAF and to investigate the outcomes of POAF for patients with cancer, a systematic search of the PubMed and Cochrane Library databases was conducted from inception of the study to 1 September 2021. The inclusion criteria specified studies reporting the prevalence of POAF among patients with cancer. The study excluded articles not written in English, review articles, case reports, letters, commentaries, systematic reviews, meta-analyses, and conference abstracts. RESULTS: The search identified 49 studies with 201,081 patients, and the pooled prevalence of POAF was 13.5% (95% confidence interval [CI], 11.6-15.7%). Meta-analyses showed that the incidence of POAF among patients with cancer was associated with age (mean difference [MD], 4.31; 95%CI, 3.16-5.47), male sex (odds ratio [OR], 1.39; 95% CI, 1.19-1.62), chronic obstructive pulmonary disease (OR, 2.47; 95% CI, 1.71-3.56), hypertension (OR, 1.47; 95% CI, 1.23-1.75), intraoperative blood transfusion (OR, 4.58; 95% CI, 2.31-9.10), and open surgery (OR, 1.51; 95% CI, 1.26-1.81). Patients with POAF had significantly higher in-hospital mortality (OR, 4.25; 95% CI, 2.79-6.45), longer hospital stays (MD, 3.07; 95% CI, 1.63-4.51), and higher incidences of pneumonia (OR, 3.32; 95% CI, 2.85-3.86), stroke (OR, 6.57; 95% CI, 1.56-26.00), and myocardial infarction (OR, 3.00; 95% CI, 1.45-6.20) than those without POAF. CONCLUSIONS: For patients with cancer, POAF is associated with an increased burden of comorbidities and worse outcomes.


Subject(s)
Atrial Fibrillation , Neoplasms , Humans , Male , Atrial Fibrillation/complications , Atrial Fibrillation/epidemiology , Hospitals , Neoplasms/surgery , Postoperative Complications/epidemiology , Risk Factors , Stroke/epidemiology , Female
6.
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
8.
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
9.
Stem Cell Res ; 62: 102793, 2022 07.
Article in English | MEDLINE | ID: mdl-35500377

ABSTRACT

Dilated cardiomyopathy (DCM) is a refractory heart disease characterized by dilation of the left ventricle and systolic dysfunction. LMNA, the gene encoding lamin A/C (a nuclear envelope protein), is the second leading causative gene associated with familial DCM. LMNA-related DCM is likely to develop severe heart failure, various types of arrhythmias, and poor prognosis. We established a human induced pluripotent stem cell line, derived from a patient with DCM carrying a nonsense mutation in LMNA. This line should be a useful resource for elucidating disease mechanisms and developing fundamental treatments for LMNA-related DCM.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Arrhythmias, Cardiac , Cardiomyopathy, Dilated/genetics , Codon, Nonsense , Heart , Humans , Induced Pluripotent Stem Cells/metabolism , Lamin Type A/genetics , Mutation
10.
Heart Vessels ; 37(4): 609-618, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34562143

ABSTRACT

Historically, a right bundle branch block has been considered a benign finding in asymptomatic individuals. However, this conclusion is based on a few old studies with small sample sizes. We examined the association between a complete right bundle branch block (CRBBB) and subsequent cardiovascular mortality in the general population in Japan. In this large community-based cohort study, data of 90,022 individuals (mean age, 58.5 ± 10.2 years; 66.2% women) who participated in annual community-based health check-ups were assessed. Subjects were followed up from 1993 to the end of 2016. Cox proportional hazards' models and log-rank tests were used for the data analysis. CRBBB was documented in 1,344 participants (1.5%). Among all included participants, CRBBB was associated with an increased risk of cardiovascular mortality after adjustment for all potential confounders (hazard ratio [HR] 1.21; 95% confidence interval [CI] 1.06-1.38). The increased risk of cardiovascular mortality was particularly evident in women aged < 65 years (HR 2.00; 95% CI 1.34-2.98) and men aged ≥ 65 years (HR 1.28; 95% CI 1.06-1.55). CRBBB is associated with an increased risk of cardiovascular mortality in women aged < 65 years and men aged ≥ 65 years. Clinicians should be aware of the presence of CRBBB in young women and elderly men, even if they exhibit no symptoms.


Subject(s)
Bundle-Branch Block , Cardiovascular System , Aged , Bundle-Branch Block/complications , Cohort Studies , Electrocardiography , Female , Humans , Japan/epidemiology , Male , Middle Aged
11.
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.

13.
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
14.
J Cardiol ; 78(6): 509-516, 2021 12.
Article in English | MEDLINE | ID: mdl-34275691

ABSTRACT

BACKGROUND: Extended-duration work shifts (EDWSs) might affect the health of physician residents, causing autonomic alteration. Skin sympathetic nerve activity (SKNA) recorded by noninvasive neuro-electrocardiography (neuECG) is used to estimate cardiac sympathetic tone. In this study, we aim to evaluate the impact of EDWSs on nocturnal SKNA assessed in resident doctors. METHODS: Twenty-four residents working EDWSs and 12 PhD students not working nightshift schedules were prospectively recruited. The neuECG was performed between 12 am and 6 am for 5 consecutive nights. SKNA was filtered from neuECG recorded signals. The questionnaires regarding work stress and sleep quality, blood pressure, and salivary alpha-amylase and cortisol levels were administered. RESULTS: The hours of weekly working and sleep opportunities were similar between residents and students, while residents reported more work stress and worse sleep quality. In residents, SKNA at 6 am (SKNA6am) was significantly higher than SKNA2am during the precall night, revealing a dipping pattern. However, the SKNA dipping disappeared during the on-call night and prominently flattened during the first postcall night, the full recovery of which was delayed until the second postcall nights. The morning blood pressure and salivary alpha-amylase and cortisol levels were similar between the precall and postcall days. In contrast, SKNA in students exhibited a constant dipping profile for all recorded nights. CONCLUSIONS: In healthy young adults, SKNA presents a dip night. The SKNA dip is impaired by working a nightshift, with a delayed recovery. The neuECG might serve as a useful tool to detect subclinical autonomic disturbances in shiftworkers.


Subject(s)
Sleep Quality , Sympathetic Nervous System , Electrocardiography , Heart Rate , Humans , Skin , Young Adult
15.
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
16.
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
17.
J Interv Cardiol ; 2020: 7417912, 2020.
Article in English | MEDLINE | ID: mdl-33177963

ABSTRACT

BACKGROUND: Premature ventricular complexes (PVCs) exhibit circadian fluctuation. We determine if PVCs of different origin exhibit specific circadian patterns. METHODS: We analyzed Holter recordings from patients with monomorphic PVCs who underwent catheter ablation. PVC circadian patterns were classified as fast-heart rate- (HR-) dependent (F-PVC), slow-HR-dependent (S-PVC), or HR-independent (I-PVC). PVC origins were determined intraprocedurally. RESULTS: In a retrospective cohort of 407 patients, F-PVC and S-PVC typically exhibited diurnal and nocturnal predominance, respectively. Despite decreased circadian fluctuation, I-PVC generally had heavier nocturnal than diurnal burden. PVCs of left anterior fascicle origin were predominantly S-PVC, while those of posterior hemibranch origin were mostly F-PVC. PVCs originating from the aortic sinus of Valsalva (ASV) were predominantly I-PVC, while most PVCs arising from the left ventricular outflow tract (LVOT) were F-PVC. Using a diurnal/nocturnal PVC burden ratio of 0.92 as the cutoff value to distinguish LVOT from ASV origin achieved 97% sensitivity and, as further verification, an accuracy of 89% (16/18) in a prospective cohort of patients with PVCs originating from either ASV or LVOT. In contrast, PVCs originating from right ventricles, such as right ventricular outflow tract, did not show distinct circadian patterns. CONCLUSIONS: The circadian patterns exhibit origin specificity for PVCs arising from left ventricles. An analysis of Holter monitoring provides useful information on PVC localization in ablation procedure planning.


Subject(s)
Catheter Ablation/methods , Circadian Rhythm/physiology , Electrocardiography, Ambulatory/methods , Ventricular Premature Complexes , Female , Heart Rate , Heart Ventricles/physiopathology , Humans , Male , Middle Aged , Patient Care Planning , Retrospective Studies , Ventricular Premature Complexes/diagnosis , Ventricular Premature Complexes/physiopathology , Ventricular Premature Complexes/surgery
18.
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
19.
Front Cell Dev Biol ; 8: 578197, 2020.
Article in English | MEDLINE | ID: mdl-33117805

ABSTRACT

Cardiac hypertrophy and fibrosis are major pathophysiologic disorders that lead to serious cardiovascular diseases (CVDs), such as heart failure and arrhythmia. It is well known that transforming growth factor ß (TGFß) signaling pathways play a major role in the proliferation of cardiac hypertrophy and fibrosis, which is mainly stimulated by angiotensin II (AgII). This study aimed to investigate the cardioprotective potential of isorhamnetin (ISO) in human amniotic epithelial stem cells (hAESCs) through global gene expression analysis and to confirm its beneficial effects on cardiac hypertrophy and fibrosis in the AgII-induced in vivo model. In vitro, biological processes including TGFß, collagen-related functions, and inflammatory processes were significantly suppressed in ISO pretreated hAESCs. In vivo, continuous AgII infusion using an osmotic pump induced significant pathological fibrosis and myocardial hypertrophy, which were remarkably suppressed by ISO pretreatment. ISO was found to reverse the enhanced TGFß and Collagen type I alpha 1 mRNA expression induced by AgII exposure, which causes cardiovascular remodeling in ventricular tissue. These findings indicate that ISO could be a potential agent against cardiac hypertrophy and fibrosis.

20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...