Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 274
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(17): e2218204121, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38621141

RESUMO

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.


Assuntos
Taquicardia Ventricular , Camundongos , Animais , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Arritmias Cardíacas/genética , Flecainida , Mutação de Sentido Incorreto , Morte Súbita Cardíaca , Mutação
2.
Circ Res ; 133(10): 861-876, 2023 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-37818671

RESUMO

BACKGROUND: The membrane components of cardiomyocytes are rich in polyunsaturated fatty acids, which are easily oxidized. Thus, an efficient glutathione-based lipid redox system is essential for maintaining cellular functions. However, the relationship between disruption of the redox system during ischemia-reperfusion (IR), oxidized lipid production, and consequent cell death (ferroptosis) remains unclear. We investigated the mechanisms underlying the disruption of the glutathione-mediated reduction system related to ferroptosis during IR and developed intervention strategies to suppress ferroptosis. METHODS: In vivo fluctuations of both intra- and extracellular metabolite levels during IR were explored via microdialysis and tissue metabolome analysis. Oxidized phosphatidylcholines were assessed using liquid chromatography high-resolution mass spectrometry. The areas at risk following IR were assessed using triphenyl-tetrazolium chloride/Evans blue stain. RESULTS: Metabolomic analysis combined with microdialysis revealed a significant release of glutathione from the ischemic region into extracellular spaces during ischemia and after reperfusion. The release of glutathione into extracellular spaces and a concomitant decrease in intracellular glutathione concentrations were also observed during anoxia-reperfusion in an in vitro cardiomyocyte model. This extracellular glutathione release was prevented by chemical inhibition or genetic suppression of glutathione transporters, mainly MRP1 (multidrug resistance protein 1). Treatment with MRP1 inhibitor reduced the intracellular reactive oxygen species levels and lipid peroxidation, thereby inhibiting cell death. Subsequent in vivo evaluation of endogenously oxidized phospholipids following IR demonstrated the involvement of ferroptosis, as levels of multiple oxidized phosphatidylcholines were significantly elevated in the ischemic region 12 hours after reperfusion. Inhibition of the MRP1 transporter also alleviated intracellular glutathione depletion in vivo and significantly reduced the generation of oxidized phosphatidylcholines. Administration of MRP1 inhibitors significantly attenuated infarct size after IR injury. CONCLUSIONS: Glutathione was released continuously during IR, primarily in an MRP1-dependent manner, and induced ferroptosis. Suppression of glutathione release attenuated ferroptosis and reduced myocardial infarct size following IR.


Assuntos
Ferroptose , Miócitos Cardíacos , Humanos , Miócitos Cardíacos/metabolismo , Reperfusão , Isquemia/metabolismo , Glutationa/metabolismo , Fosfolipídeos/metabolismo , Fosfatidilcolinas
3.
Cell ; 142(3): 375-86, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20691899

RESUMO

The reprogramming of fibroblasts to induced pluripotent stem cells (iPSCs) raises the possibility that a somatic cell could be reprogrammed to an alternative differentiated fate without first becoming a stem/progenitor cell. A large pool of fibroblasts exists in the postnatal heart, yet no single "master regulator" of direct cardiac reprogramming has been identified. Here, we report that a combination of three developmental transcription factors (i.e., Gata4, Mef2c, and Tbx5) rapidly and efficiently reprogrammed postnatal cardiac or dermal fibroblasts directly into differentiated cardiomyocyte-like cells. Induced cardiomyocytes expressed cardiac-specific markers, had a global gene expression profile similar to cardiomyocytes, and contracted spontaneously. Fibroblasts transplanted into mouse hearts one day after transduction of the three factors also differentiated into cardiomyocyte-like cells. We believe these findings demonstrate that functional cardiomyocytes can be directly reprogrammed from differentiated somatic cells by defined factors. Reprogramming of endogenous or explanted fibroblasts might provide a source of cardiomyocytes for regenerative approaches.


Assuntos
Diferenciação Celular , Fibroblastos/citologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Animais , Separação Celular , Fibroblastos/metabolismo , Fator de Transcrição GATA4/metabolismo , Perfilação da Expressão Gênica , Fatores de Transcrição MEF2 , Camundongos , Contração Muscular , Miócitos Cardíacos/metabolismo , Fatores de Regulação Miogênica/metabolismo , Proteínas com Domínio T/metabolismo
4.
Semin Cell Dev Biol ; 122: 21-27, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34210577

RESUMO

Cardiovascular diseases are a common cause of death worldwide. Adult cardiomyocytes have limited regenerative capacity after injury, and there is growing interest in cardiac regeneration as a new therapeutic strategy. There are several limitations of induced pluripotent stem cell-based transplantation therapy with respect to efficiency and risks of tumorigenesis. Direct reprogramming enables the conversion of terminally differentiated cells into target cell types using defined factors. In most cardiac diseases, activated fibroblasts proliferate in the damaged heart and contribute to the progression of heart failure. In vivo cardiac reprogramming, in which resident cardiac fibroblasts are converted into cardiomyocytes in situ, is expected to become a new cardiac regenerative therapy. Indeed, we and other groups have demonstrated that in vivo reprogramming improves cardiac function and reduces fibrosis after myocardial infarction. In this review, we summarize recent discoveries and developments related to in vivo reprogramming. In addition, issues that need to be resolved for clinical application are described.


Assuntos
Reprogramação Celular/fisiologia , Cardiopatias/terapia , Miócitos Cardíacos/metabolismo , Medicina Regenerativa/métodos , Animais , Humanos , Camundongos
5.
Circulation ; 147(3): 223-238, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36503256

RESUMO

BACKGROUND: Because adult cardiomyocytes have little regenerative capacity, resident cardiac fibroblasts (CFs) synthesize extracellular matrix after myocardial infarction (MI) to form fibrosis, leading to cardiac dysfunction and heart failure. Therapies that can regenerate the myocardium and reverse fibrosis in chronic MI are lacking. The overexpression of cardiac transcription factors, including Mef2c/Gata4/Tbx5/Hand2 (MGTH), can directly reprogram CFs into induced cardiomyocytes (iCMs) and improve cardiac function under acute MI. However, the ability of in vivo cardiac reprogramming to repair chronic MI with established scars is undetermined. METHODS: We generated a novel Tcf21iCre/reporter/MGTH2A transgenic mouse system in which tamoxifen treatment could induce both MGTH and reporter expression in the resident CFs for cardiac reprogramming and fibroblast lineage tracing. We first tested the efficacy of this transgenic system in vitro and in vivo for acute MI. Next, we analyzed in vivo cardiac reprogramming and fusion events under chronic MI using Tcf21iCre/Tomato/MGTH2A and Tcf21iCre/mTmG/MGTH2A mice, respectively. Microarray and single-cell RNA sequencing were performed to determine the mechanism of cardiac repair by in vivo reprogramming. RESULTS: We confirmed the efficacy of transgenic in vitro and in vivo cardiac reprogramming for acute MI. In chronic MI, in vivo cardiac reprogramming converted ≈2% of resident CFs into iCMs, in which a majority of iCMs were generated by means of bona fide cardiac reprogramming rather than by fusion with cardiomyocytes. Cardiac reprogramming significantly improved myocardial contraction and reduced fibrosis in chronic MI. Microarray analyses revealed that the overexpression of MGTH activated cardiac program and concomitantly suppressed fibroblast and inflammatory signatures in chronic MI. Single-cell RNA sequencing demonstrated that resident CFs consisted of 7 subclusters, in which the profibrotic CF population increased under chronic MI. Cardiac reprogramming suppressed fibroblastic gene expression in chronic MI by means of conversion of profibrotic CFs to a quiescent antifibrotic state. MGTH overexpression induced antifibrotic effects partly by suppression of Meox1, a central regulator of fibroblast activation. CONCLUSIONS: These results demonstrate that cardiac reprogramming could repair chronic MI by means of myocardial regeneration and reduction of fibrosis. These findings present opportunities for the development of new therapies for chronic MI and heart failure.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Camundongos , Animais , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fibrose , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Fibroblastos/metabolismo , Reprogramação Celular
6.
Biochem Biophys Res Commun ; 690: 149272, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37992523

RESUMO

Cardiomyocytes (CMs) have little regenerative capacity. After myocardial infarction (MI), scar formation and myocardial remodeling proceed in the infarct and non-infarct areas, respectively, leading to heart failure (HF). Prolonged activation of cardiac fibroblasts (CFs) and inflammatory cells may contribute to this process; however, therapies targeting these cell types remain lacking. Cardiac reprogramming converts CFs into induced CMs, reduces fibrosis, and improves cardiac function in chronic MI through the overexpression of Mef2c/Gata4/Tbx5/Hand2 (MGTH). However, whether cardiac reprogramming reduces inflammation in infarcted hearts remains unclear. Moreover, the mechanism through which MGTH overexpression in CFs affects inflammatory cells remains unknown. Here, we showed that inflammation persists in the myocardium until three months after MI, which can be reversed with cardiac reprogramming. Single-cell RNA sequencing demonstrated that CFs expressed pro-inflammatory genes and exhibited strong intercellular communication with inflammatory cells, including macrophages, in chronic MI. Cardiac reprogramming suppressed the inflammatory profiles of CFs and reduced the relative ratios and pro-inflammatory signatures of cardiac macrophages. Moreover, fluorescence-activated cell sorting analysis (FACS) revealed that cardiac reprogramming reduced the number of chemokine receptor type 2 (CCR2)-positive inflammatory macrophages in the non-infarct areas in chronic MI, thereby restoring myocardial remodeling. Thus, cardiac reprogramming reduced the number of inflammatory macrophages to exacerbate cardiac function after MI.


Assuntos
Infarto do Miocárdio , Humanos , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Macrófagos/metabolismo , Inflamação/metabolismo , Fibroblastos/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 43(9): 1668-1683, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37534464

RESUMO

BACKGROUND: The mechanisms underlying pulmonary hypertension (PH) remain largely unknown; further, why advanced vascular remodeling preferentially occurs in arterioles is yet to be answered. VEGF (vascular endothelial growth factor) regulates angiogenesis through Flk1 (fetal liver kinase 1) and Flt1 (fms-like tyrosine kinase 1) on endothelial cells (ECs), which may be related to PH pathogenesis. However, spatiotemporal expression patterns of Flk1 and Flt1 in the pulmonary vascular system and the role of endothelial Flk1 in PH development remain poorly understood. METHODS: We analyzed multiple reporter mice, including Flk1-GFP (green fluorescent protein) bacterial artificial chromosome transgenic (Tg), Flt1-DsRed bacterial artificial chromosome Tg, and Flk1-GFP/Flt1-DsRed double Tg mice, to determine the spatiotemporal expression of Flk1 and Flt1 in hypoxia-induced PH. We also used Cdh5CreERT2/Flk1f/f/Tomato (Flk1-KO [knockout]) mice to induce EC-specific Flk1 deletion and lineage tracing in chronic hypoxia. RESULTS: Flk1 was specifically expressed in the ECs of small pulmonary vessels, including arterioles. Conversely, Flt1 was more broadly expressed in the ECs of large- to small-sized vessels in adult mouse lungs. Intriguingly, Flk1+ ECs were transiently increased in hypoxia with proliferation, whereas Flt1 expression was unchanged. Flk1-KO mice did not exhibit pulmonary vascular remodeling nor PH in normoxia; however, the arteriolar ECs changed to a cuboidal shape with protrusion. In hypoxia, Flk1 deletion exacerbated EC dysfunction and reduced their number via apoptosis. Additionally, Flk1 deletion promoted medial thickening and neointimal formation in arterioles and worsened PH. Mechanistically, lineage tracing revealed that neointimal cells were derived from Flk1-KO ECs. Moreover, RNA sequencing in pulmonary ECs demonstrated that Flk1 deletion and hypoxia synergistically activated multiple pathways, including cell cycle, senescence/apoptosis, and cytokine/growth factor, concomitant with suppression of cell adhesion and angiogenesis, to promote vascular remodeling. CONCLUSIONS: Flk1 and Flt1 were differentially expressed in pulmonary ECs. Flk1 deficiency and hypoxia jointly dysregulated arteriolar ECs to promote vascular remodeling. Thus, dysfunction of Flk1+ ECs may contribute to the pathogenesis of advanced vascular remodeling in pulmonary arterioles.


Assuntos
Hipertensão Pulmonar , Remodelação Vascular , Animais , Camundongos , Células Endoteliais/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Hipóxia/complicações , Hipóxia/genética , Hipóxia/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
J Mol Cell Cardiol ; 178: 1-8, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36918145

RESUMO

The incidence of cardiovascular diseases is increasing worldwide, and cardiac regenerative therapy has great potential as a new treatment strategy, especially for ischemic heart disease. Direct cardiac reprogramming is a promising new cardiac regenerative therapy that uses defined factors to induce transdifferentiation of endogenous cardiac fibroblasts (CFs) into induced cardiomyocyte-like cells (iCMs). In vivo reprogramming is expected to restore lost cardiac function without necessitating cardiac transplantation by converting endogenous CFs that exist abundantly in cardiac tissues directly into iCMs. Indeed, we and other groups have demonstrated that in vivo cardiac reprogramming improves cardiac contractile function and reduces scar area after acute myocardial infarction (MI). Recently, we demonstrated that in vivo cardiac reprogramming is an innovative cardiac regenerative therapy that not only regenerates the myocardium, but also reverses fibrosis by inducing the quiescence of pro-fibrotic fibroblasts, thereby improving heart failure in chronic MI. In this review, we summarize the recent progresses in in vivo cardiac reprogramming, and discuss its prospects for future clinical applications and the challenges of direct human reprogramming, which has been a longstanding issue.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Humanos , Reprogramação Celular/genética , Miocárdio , Miócitos Cardíacos , Infarto do Miocárdio/terapia , Fibroblastos
9.
Ann Surg Oncol ; 30(2): 711-721, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36273057

RESUMO

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.


Assuntos
Fibrilação Atrial , Neoplasias , Humanos , Masculino , Fibrilação Atrial/complicações , Fibrilação Atrial/epidemiologia , Hospitais , Neoplasias/cirurgia , Complicações Pós-Operatórias/epidemiologia , Fatores de Risco , Acidente Vascular Cerebral/epidemiologia , Feminino
10.
Eur Radiol ; 33(5): 3020-3028, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36441216

RESUMO

OBJECTIVES: To investigate the relationship between periprocedural myocardial injury (PMI) and plaque characteristics detected by multidetector computed tomography (MDCT) and cardiac magnetic resonance imaging (CMR). MATERIALS AND METHODS: This observational retrospective study, between July 2012 and October 2019, included chronic coronary syndrome patients undergoing elective percutaneous coronary intervention (PCI) after MDCT and CMR. High-intensity plaque (HIP) on non-contrast T1-weighted imaging was defined as a coronary plaque-to-myocardium signal intensity ratio of ≥ 1.4. High-risk plaque (HRP) in MDCT displayed ≥ 2 features: positive remodeling, low-attenuation plaque, spotty calcification, and napkin-ring sign. PMI was defined as an increase in cardiac troponin T levels > 5 times the upper normal limit at 24 h after PCI. RESULTS: Ninety-five target lesions in 76 patients (mean age ± standard deviation, 67 years ± 9; 62 males [82%]) were included. Twenty-one patients (24 lesions) were assigned to the PMI group, while 55 patients (71 lesions) to the non-PMI group. Presence of HRP characteristics on MDCT and HIP on CMR was significantly higher in the PMI group. Multivariate logistic regression analysis showed that HRP in MDCT and HIP in CMR were significant independent predictors of PMI. Target lesions with HRP on MDCT and HIP on CMR were significantly more likely to develop PMI. In 141 plaques with ≥ 50% stenosis (76 patients), patients with PMI had significantly more frequent HRP in MDCT and HIP in CMR in target and non-target lesions. CONCLUSIONS: MDCT and CMR can play an important role in the detection of high-risk lesions for PMI following elective PCI. KEY POINTS: • Multivariate logistic regression analysis showed that high-risk plaque on MDCT and high-intensity plaque on MRI were significant independent predictors of periprocedural myocardial injury (PMI). • Target lesions with high-risk plaque on MDCT and high-intensity plaque on CMR were significantly more likely to develop PMI. • In 141 plaques with ≥ 50% stenosis, patients with PMI were significantly more likely to have high-risk plaques on MDCT and high-intensity plaque on CMR in target and non-target lesions.


Assuntos
Doença da Artéria Coronariana , Traumatismos Cardíacos , Intervenção Coronária Percutânea , Placa Aterosclerótica , Masculino , Humanos , Estudos Retrospectivos , Constrição Patológica , Placa Aterosclerótica/diagnóstico por imagem , Traumatismos Cardíacos/diagnóstico por imagem , Traumatismos Cardíacos/etiologia , Fatores de Risco , Angiografia Coronária/métodos
11.
Circ J ; 87(7): 973-981, 2023 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-37258220

RESUMO

BACKGROUND: An epicardial connection (EC) between the right-sided pulmonary venous (RtPV) carina and right atrium (RA) may preclude PV isolation, but its electrophysiological role during atrial fibrillation (AF) remains unknown.Methods and Results: This prospective observational study included 98 consecutive patients undergoing catheter ablation for AF, subdivided into the EC group (n=17) and non-EC group (n=80) based on observation of RA posterior wall breakthrough during RtPV pacing. Mean left atrial (LA) dominant frequency (mean DFLA) was defined as the averaged DFs at the right and left PVs and LA appendage. The regional DF was higher in the EC group vs. the non-EC group except at the left PV antrum. The DF at the RA appendage (RAA) and mean DFLAwere equivocal (6.5±0.7 vs. 6.6±0.7 Hz) in the EC group, but the mean DFLAwas significantly higher than that at the RAA (5.8±0.6 vs. 6.1±0.5 Hz, P=0.001) in the non-EC group, suggesting an LA-to-RA DF gradient. A significant correlation of DF between the RtPV antrum and RAA was observed in the EC group (P<0.001, r=0.84) but not in the non-EC group. CONCLUSIONS: An electrophysiological link via interatrial ECs might attenuate the hierarchical nature of activation frequencies of AF, leading to advanced electrical remodeling of the atria.


Assuntos
Apêndice Atrial , Fibrilação Atrial , Ablação por Cateter , Veias Pulmonares , Humanos , Átrios do Coração , Veias Pulmonares/cirurgia , Técnicas Eletrofisiológicas Cardíacas/métodos , Ablação por Cateter/métodos
12.
Circ J ; 88(1): 83-89, 2023 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-37880107

RESUMO

BACKGROUND: The prevalence of adult congenital heart disease (ACHD) is increasing rapidly and in particular, patients who underwent complicated surgeries are reaching their youth and middle age. Therefore, the need for ACHD treatment will increase, but the current medical situation is unknown. In this study we assessed trends in unplanned admissions in patients with ACHD in Japan.Methods and Results: From the Japanese Registry of All Cardiac and Vascular Diseases-Diagnosis Procedure Combination, a nationwide claim-based database, we selected patients aged >15 years with CHD defined by the International Classification of Diseases, 10th Revision codes. We identified 39,676 admissions between April 2012 and March 2018; 10,444 (26.3%) were unplanned. Main diagnoses were categorized into 3 degrees of complexity (severe, moderate, and mild) and other. Among unplanned admissions, the proportion of the severe group increased with time. Patients in the mild group were significantly older than those in the moderate and severe groups (median age: 70.0, 39.0, and 32.0 years, respectively). There were 765 deaths during hospitalization (overall mortality rate, 7.3%). The odds ratio of death during admission was significantly higher in patients aged >50 years, especially in the moderate group. CONCLUSIONS: Patients with moderate or severe ACHD tended to experience unplanned admissions at a younger age. In anticipation of greater numbers of new, severe patients, we need to prepare for their increasing medical demands.


Assuntos
Cardiopatias Congênitas , Doenças Hematológicas , Doenças Vasculares , Pessoa de Meia-Idade , Adolescente , Humanos , Adulto , Cardiopatias Congênitas/diagnóstico , Cardiopatias Congênitas/epidemiologia , Cardiopatias Congênitas/terapia , Japão/epidemiologia , Hospitalização , Sistema de Registros
13.
Circ J ; 87(12): 1800-1808, 2023 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-37394572

RESUMO

BACKGROUND: In patients with atrial fibrillation (AF) and severe blood stasis in the left atrial appendage (LAA), dense spontaneous echo contrast (SEC) disturbs the distinct visualization of the LAA interior, thus making thrombus diagnosis inconclusive. We aimed to prospectively assess the efficacy and safety of a protocol for a low-dose isoproterenol (ISP) infusion to reduce SEC to exclude an LAA thrombus.Methods and Results: We enrolled 17 patients with AF and dense SEC (Grade 4 or sludge). ISP was infused with gradually increasing doses of 0.01, 0.02, and 0.03 µg/kg/min at 3-min intervals. After increasing the dose to 0.03 µg/kg/min for 3 min, or when the LAA interior was visible, the infusion was terminated. We reassessed the SEC grade, presence of an LAA thrombus, LAA function, and left ventricular ejection fraction (LVEF) within 1 min of ISP termination. Compared with baseline, ISP significantly increased LAA flow velocity, the LAA emptying fraction, LAA wall velocities, and LVEF (all P<0.01). ISP administration significantly reduced the SEC grade (median) from 4 to 1 (P<0.001). The SEC grade decreased to ≤2 in 15 (88%) patients, and the LAA thrombus was excluded. There were no adverse events. CONCLUSIONS: Low-dose ISP infusion may be effective and safe to reduce SEC and exclude an LAA thrombus by improving LAA function and LVEF.


Assuntos
Apêndice Atrial , Fibrilação Atrial , Cardiopatias , Trombose , Humanos , Fibrilação Atrial/complicações , Fibrilação Atrial/tratamento farmacológico , Isoproterenol , Apêndice Atrial/diagnóstico por imagem , Volume Sistólico , Ecocardiografia Transesofagiana/métodos , Função Ventricular Esquerda , Cardiopatias/etiologia , Trombose/diagnóstico por imagem , Trombose/etiologia
14.
Int Heart J ; 64(1): 90-94, 2023 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-36725074

RESUMO

Although rare, long QT syndrome (LQTS) and peripartum cardiomyopathy (PPCM) are the major causes of maternal cardiovascular death. We herein present a case study of a 23-year-old woman with LQTS, pregnancy-induced hypertension, and PPCM. During the postpartum period, her left ventricular systolic function had severely decreased, requiring the administration of loop diuretics. Diuretics cause several changes in the circulating blood volume, electrolyte balance, and hormonal status during pregnancy, delivery, and the peripartum period. Extreme QTc prolongation and fatal ventricular arrhythmia require frequent defibrillation. For the patient in this study, we corrected her electrolyte abnormality, and eventually, we controlled the arrhythmia by administering a ß-blocker and Na-channel blocker. Although the arrhythmia subsided, she continued on medication after discharge to prevent the recurrence of fatal arrhythmia. In conclusion, close attention should be paid to patients with LQTS, especially when some changes that may lead to QTc prolongation could occur during the peripartum period.


Assuntos
Cardiomiopatias , Síndrome do QT Longo , Torsades de Pointes , Humanos , Gravidez , Feminino , Adulto Jovem , Adulto , Torsades de Pointes/induzido quimicamente , Período Periparto , Eletrocardiografia , Arritmias Cardíacas/complicações , Síndrome do QT Longo/complicações , Síndrome do QT Longo/diagnóstico , Cardiomiopatias/complicações , Cardiomiopatias/diagnóstico
15.
Int Heart J ; 64(6): 1071-1078, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-37967975

RESUMO

Sacubitril/valsartan improves outcomes in patients with heart failure (HF) with reduced ejection fraction. However, the relationship between longitudinal changes in natriuretic peptides and echocardiographic parameters in patients with HF treated with sacubitril/valsartan across the left ventricular ejection fraction (LVEF) range is not fully understood.In patients with HF treated with sacubitril/valsartan, comprehensive data on natriuretic peptides, including atrial natriuretic peptide (ANP), N-terminal pro-brain-type natriuretic peptide (NT-proBNP), BNP, and echocardiography, were measured after 6 months of treatment. We assessed the change in natriuretic peptides and echocardiographic parameters in LVEF classification subgroups.Among 49 patients, the median ANP concentration increased from 55 pg/mL at baseline to 78 pg/mL (P < 0.001). The NT-proBNP concentration decreased from 250 pg/mL to 146 pg/mL (P < 0.001). No significant change was observed in the BNP concentration (P = 0.640). The trajectories of each natriuretic peptide in patients with LVEF > 40% (n = 22) were similar to those in individuals with LVEF ≤ 40% (n = 27). Regardless of LVEF classification, echocardiography at 6 months showed a significant improvement in LVEF, left ventricular end-diastolic volume, and the ratio of early diastolic mitral inflow velocity to early diastolic mitral annulus velocity (E/e'). The reduction in natriuretic peptide concentration was related to LV reverse remodeling and decreased left and right atrial pressures assessed by E/e' and inferior vena cava diameter.Sacubitril/valsartan induced an increase in ANP, a reduction in NT-proBNP, and no change in plasma BNP, regardless of LVEF. It caused LV reverse remodeling, and the natriuretic peptide concentration changes were associated with structural and functional echocardiographic parameters.


Assuntos
Insuficiência Cardíaca , Tração , Humanos , Volume Sistólico , Remodelação Ventricular , Tetrazóis/uso terapêutico , Função Ventricular Esquerda , Valsartana , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/tratamento farmacológico
16.
J Phys Ther Sci ; 35(2): 114-120, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36744202

RESUMO

[Purpose] We have recently reported that using a wearable cyborg hybrid assistive limb improved the isometric knee extensor muscle strength of patients with chronic heart failure. Here, we investigated the long-term effects of a lumbar-type hybrid assistive limb for patients with chronic heart failure. [Participants and Methods] A total of 28 hospitalized patients with chronic heart failure (mean age, 73.1 ± 13.8 years) were randomly assigned to two groups: the hybrid assistive limb group or the control group, in which they performed a sit-to-stand exercise with or without the hybrid assistive limb, respectively. The cardiac rehabilitation therapy included this intervention, which was performed as many times as possible for 5-30 minutes per day for 6-10 days. Clinical assessments like lower-limb muscle strength, walking ability, etc., were measured before and after the intervention. Cardiac events were followed up for up to a year after discharge. [Results] No adverse events occurred during the study period in either group. In terms of long-term effects, the incidence of cardiac events was 23% and 45% in the hybrid assistive limb and the control groups, respectively. [Conclusion] Hybrid assistive limb-assisted exercise therapy may be a safe and feasible cardiac rehabilitation tool in patients with chronic heart failure. The lumbar-type wearable cyborg hybrid assistive limb may have a positive effect on heart failure prognosis by adding long-term exercise therapy.

17.
Clin Sci (Lond) ; 136(24): 1831-1849, 2022 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-36540030

RESUMO

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.


Assuntos
Fibrilação Atrial , Remodelamento Atrial , Masculino , Camundongos , Animais , Fibrilação Atrial/tratamento farmacológico , Fibrilação Atrial/prevenção & controle , Cálcio/metabolismo , Camundongos Endogâmicos C57BL , Átrios do Coração/patologia , Miócitos Cardíacos/metabolismo , Angiotensina II/metabolismo
18.
Europace ; 24(4): 587-597, 2022 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-34543395

RESUMO

AIMS: A high-density pace-mapping can depict an abrupt transition in paced QRS morphology from a poor to excellent match, unmasking the critical component of ventricular tachycardia (VT) isthmus from the entrance to exit. We sought to assess pace-mapping at multiple sites within the endo- and epicardial scars to identify the VT isthmus in patients with ischaemic (ICM) and non-ischaemic cardiomyopathy (NICM). METHODS AND RESULTS: Colour-coded maps correlating to the percentage matches between 12-lead electrocardiograms during VT and pace-mapping [referred to as correlation score maps (CSMs)] were analysed. We studied 115 CSMs (80 endo- and 35 epicardial CSMs) in 37 patients (17 ICM, 20 NICM). The CSM with an abrupt change (AC) in pacemap score (AC-type) on the endocardium was more frequently observed in ICM than in NICM [11/39 (28%) vs. 1/41 (2%); P = 0.001]. Among 35 CSMs that were analysed by the combined endo- and epicardial mapping, 10 (29%) CSMs exhibited non-AC-type on the endocardium; however, AC-type was present on the opposite epicardium. Although 24 (69%) CSMs did not show AC-type on both the endocardium and epicardium, 16 of them had either an excellent (>90%) or poor (<0%) correlation score on either side, associated with isthmus exit or entrance, respectively. However, the remaining eight CSMs had neither excellent nor poor scores. CONCLUSION: The CSM may provide electrophysiological information to localize the endo- and epicardial VT isthmus. The absence of AC-type CSM on the endocardium, which is frequently observed in NICM, appears to indicate the sub-epicardial or intramural course of the critical isthmus.


Assuntos
Cardiomiopatias , Ablação por Cateter , Isquemia Miocárdica , Taquicardia Ventricular , Cardiomiopatias/complicações , Cardiomiopatias/diagnóstico , Endocárdio , Mapeamento Epicárdico , Humanos , Isquemia Miocárdica/complicações , Isquemia Miocárdica/diagnóstico , Taquicardia Ventricular/diagnóstico , Taquicardia Ventricular/etiologia , Taquicardia Ventricular/cirurgia
19.
Circ J ; 86(8): 1263-1272, 2022 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-35786689

RESUMO

BACKGROUND: The left atrial appendage (LAA) is a therapeutic target for preventing cardioembolic stroke in patients with non-valvular atrial fibrillation (NVAF). A large LAA ostium limits percutaneous LAA closure. This study investigated the characteristics and factors associated with a large LAA ostium in Japanese patients with NVAF.Methods and Results: In 1,102 NVAF patients, the maximum LAA diameter was measured using transesophageal echocardiography (TEE). A large LAA ostium was defined by a maximum diameter of >30 mm. Forty-four participants underwent repeated TEEs, and changes in LAA size under lasting AF were assessed. A large LAA ostium was observed in 3.1% of all participants and 8.9% of patients with long-standing persistent AF (LSAF). The large LAA group had greater CHA2DS2-VASc (P=0.024) and HAS-BLED scores (P=0.046) and a higher prevalence of LAA thrombus (P=0.004) than did the normal LAA group. LSAF, moderate or severe mitral regurgitation, left atrial volume ≥42 mL/m2, E/E' ratio ≥9.5, and left ventricular mass ≥85 mg/m2were independently associated with a large LAA ostium (P<0.001, P<0.001, P=0.009, P=0.009, and P=0.032, respectively). In 44 patients with lasting AF, the LAA ostial diameter increased over time (P<0.001). CONCLUSIONS: NVAF patients with a large LAA ostium may have a higher risk of stroke and bleeding. LSAF and factors leading to LA overload may be closely associated with LAA ostial dilatation and can promote it.


Assuntos
Apêndice Atrial , Fibrilação Atrial , Acidente Vascular Cerebral , Trombose , Apêndice Atrial/diagnóstico por imagem , Apêndice Atrial/cirurgia , Fibrilação Atrial/complicações , Fibrilação Atrial/cirurgia , Ecocardiografia Transesofagiana , Humanos , Fatores de Risco , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/prevenção & controle
20.
Circ J ; 86(12): 2029-2039, 2022 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-35944977

RESUMO

BACKGROUND: Elevated central venous pressure (CVP) in heart failure causes renal congestion, which deteriorates prognosis. Sodium glucose co-transporter 2 inhibitor (SGLT2-i) improves kidney function and heart failure prognosis; however, it is unknown whether they affect renal congestion. This study investigated the effect of SGLT2-i on the kidney and left ventricle using model rats with hypertensive heart failure.Methods and Results: Eight rats were fed a 0.3% low-salt diet (n=7), and 24 rats were fed an 8% high-salt diet, and they were divided into 3 groups of untreated (n=6), SGLT2-i (canagliflozin; n=6), and loop diuretic (furosemide; n=5) groups after 11 weeks of age. At 18 weeks of age, CVP and renal intramedullary pressure (RMP) were monitored directly by catheterization. We performed contrast-enhanced ultrasonography to evaluate intrarenal perfusion. In all high-salt fed groups, systolic blood pressure was elevated (P=0.287). The left ventricular ejection fraction did not differ among high-salt groups. Although CVP decreased in both the furosemide (P=0.032) and the canagliflozin groups (P=0.030), RMP reduction (P=0.003) and preserved renal medulla perfusion were only observed in the canagliflozin group (P=0.001). Histological analysis showed less cast formation in the intrarenal tubule (P=0.032), left ventricle fibrosis (P<0.001), and myocyte thickness (P<0.001) in the canagliflozin group than in the control group. CONCLUSIONS: These results suggest that SGLT2-i causes renal decongestion and prevents left ventricular hypertrophy, fibrosis, and dysfunction.


Assuntos
Insuficiência Cardíaca , Hipertensão , Inibidores do Transportador 2 de Sódio-Glicose , Animais , Ratos , Canagliflozina/farmacologia , Fibrose , Furosemida/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/etiologia , Ventrículos do Coração , Hipertensão/complicações , Hipertensão/tratamento farmacológico , Rim , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Volume Sistólico , Função Ventricular Esquerda
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA