Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(17): e2218204121, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38621141

RESUMEN

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.


Asunto(s)
Taquicardia Ventricular , Ratones , Animales , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Arritmias Cardíacas/genética , Flecainida , Mutación Missense , Muerte Súbita Cardíaca , Mutación
2.
Circ J ; 88(5): 751-759, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38462534

RESUMEN

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.


Asunto(s)
Antraciclinas , Cardiotoxicidad , Péptido Natriurético Encefálico , Humanos , Antraciclinas/efectos adversos , Persona de Mediana Edad , Femenino , Masculino , Estudios Prospectivos , Anciano , Péptido Natriurético Encefálico/sangre , Biomarcadores/sangre , Troponina T/sangre , Ecocardiografía , Sistema de Registros , Diagnóstico Precoz
3.
Ann Surg Oncol ; 30(2): 711-721, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36273057

RESUMEN

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.


Asunto(s)
Fibrilación Atrial , Neoplasias , Humanos , Masculino , Fibrilación Atrial/complicaciones , Fibrilación Atrial/epidemiología , Hospitales , Neoplasias/cirugía , Complicaciones Posoperatorias/epidemiología , Factores de Riesgo , Accidente Cerebrovascular/epidemiología , Femenino
4.
Jpn J Clin Oncol ; 53(3): 187-194, 2023 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-36629281

RESUMEN

Cancer and cardiovascular disease share several risk factors. Clonal heamatopoiesis, a novel risk factor associated with both diseases, has received increasing attention in the fields of cardiology, heamatology and oncology. Clonal heamatopoiesis of indeterminate potential refers to the presence of at least one driver mutation in the heamatopoietic cells of peripheral blood without heamatological malignancy. Clonal heamatopoiesis of indeterminate potential is a common age-related condition that affects up to 60% of individuals aged > 80 years. Importantly, clonal heamatopoiesis of indeterminate potential carriers have a 2- to 4-fold higher risk of developing cardiovascular disease than non-carriers. Therefore, we performed an up-to-date review of clonal heamatopoiesis and its association with various forms of cardiovascular disease, including atherosclerotic disease, heart failure, aortic stenosis and pulmonary hypertension. In addition, we reviewed experimental studies that examined the causality and directionality between clonal heamatopoiesis and cardiovascular disease. Lastly, we discussed future research directions that will aid in the design of personalized therapies and preventive strategies for individuals with clonal heamatopoiesis. This review showed that clonal heamatopoiesis of indeterminate potential is a common condition, especially in older patients, and is associated with an increased risk of cardiovascular disease and worse prognosis. However, further research is needed to determine whether anti-inflammatory therapies or therapies that can reduce or eliminate clone size are effective in preventing cardiovascular disease in patients with clonal heamatopoiesis of indeterminate potential.


Asunto(s)
Cardiología , Enfermedades Cardiovasculares , Humanos , Anciano , Enfermedades Cardiovasculares/genética , Oncología Médica , Mutación
5.
Clin Sci (Lond) ; 136(24): 1831-1849, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36540030

RESUMEN

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.


Asunto(s)
Fibrilación Atrial , Remodelación Atrial , Masculino , Ratones , Animales , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/prevención & control , Calcio/metabolismo , Ratones Endogámicos C57BL , Atrios Cardíacos/patología , Miocitos Cardíacos/metabolismo , Angiotensina II/metabolismo
6.
Jpn J Clin Oncol ; 52(7): 659-664, 2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35348725

RESUMEN

Immune checkpoint inhibitors have demonstrated significant clinical benefits in many cancers, and the use of these drugs is rapidly expanding. Unfortunately, these agents can induce a wide range of immune-related adverse events through the activation of immune responses in non-target organs, including the cardiovascular system. Among cardiovascular immune-related adverse events, myocarditis is the most established and biologically plausible cardiac complication of immune checkpoint inhibitors therapy with immune-related pathophysiology. In contrast, atherosclerotic cardiovascular diseases, such as myocardial infarction and ischemic stroke, were not previously recognized as a part of the immune-related adverse event spectrum. However, there is now increasing preclinical and clinical evidence that suggests a possible correlation between immune checkpoint inhibitors therapy and atherosclerotic cardiovascular events, and cardiovascular disease is increasingly recognized as a toxicity of ICIs. Results from animal studies suggest that the blockade of the cytotoxic T-lymphocyte antigen 4 or programmed cell death protein 1 pathway plays a relevant role in promoting the progression of atherosclerotic lesions. Several clinical studies have reported an increased incidence of atherosclerotic vascular events after immune checkpoint inhibitor administration. Our findings suggest that clinicians should (i) recognize that immune checkpoint inhibitors can exacerbate atherosclerosis, (ii) consider the management of cardiovascular risk factors and (iii) perform periodic screening in patients receiving immune checkpoint inhibitors.


Asunto(s)
Aterosclerosis , Miocarditis , Neoplasias , Animales , Aterosclerosis/inducido químicamente , Aterosclerosis/tratamiento farmacológico , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Neoplasias/tratamiento farmacológico
7.
Clin Sci (Lond) ; 135(20): 2409-2422, 2021 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-34386810

RESUMEN

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.


Asunto(s)
Fibrilación Atrial/prevención & control , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Inhibidores Enzimáticos/farmacología , Febuxostat/farmacología , Hipertensión/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Xantina Oxidasa/antagonistas & inhibidores , Alopurinol/farmacología , Animales , Fibrilación Atrial/enzimología , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Conexinas/genética , Conexinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/enzimología , Uniones Comunicantes/patología , Hipertensión/enzimología , Hipertensión/genética , Hipertensión/fisiopatología , Masculino , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Oxidación-Reducción , Fosforilación , Ratas Endogámicas Dahl , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Cloruro de Sodio Dietético , Xantina Oxidasa/metabolismo , Proteína alfa-5 de Unión Comunicante
9.
Int J Mol Sci ; 22(2)2021 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-33466825

RESUMEN

Dilated cardiomyopathy (DCM) is a potentially lethal disorder characterized by progressive impairment of cardiac function. Chronic myocarditis has long been hypothesized to be one of the causes of DCM. However, owing to the lack of suitable animal models of chronic myocarditis, its pathophysiology remains unclear. Here, we report a novel mouse model of chronic myocarditis induced by recombinant bacille Calmette-Guérin (rBCG) expressing a CD4+ T-cell epitope of cardiac myosin heavy chain-α (rBCG-MyHCα). Mice immunized with rBCG-MyHCα developed chronic myocarditis, and echocardiography revealed dilation and impaired contraction of ventricles, similar to those observed in human DCM. In the heart, CD62L-CD4+ T cells were increased and produced significant amounts of IFN-γ and IL-17 in response to cardiac myosin. Adoptive transfer of CD62L-CD4+ T cells induced myocarditis in the recipient mice, which indicated that CD62L-CD4+ T cells were the effector cells in this model. rBCG-MyHCα-infected dendritic cells produced proinflammatory cytokines and induced MyHCα-specific T-cell proliferation and Th1 and Th17 polarization. This novel chronic myocarditis mouse model may allow the identification of the central pathophysiological and immunological processes involved in the progression to DCM.


Asunto(s)
Vacuna BCG/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Miocarditis/inmunología , Miosinas Ventriculares/inmunología , Animales , Vacuna BCG/genética , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Enfermedad Crónica , Citocinas/inmunología , Citocinas/metabolismo , Ecocardiografía , Epítopos de Linfocito T/genética , Humanos , Interleucina-17/inmunología , Interleucina-17/metabolismo , Activación de Linfocitos , Masculino , Ratones Endogámicos BALB C , Miocarditis/patología , Miocarditis/fisiopatología , Proteínas Recombinantes/inmunología , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Miosinas Ventriculares/genética
10.
Int J Mol Sci ; 22(2)2021 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-33450865

RESUMEN

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.


Asunto(s)
Elastasa de Leucocito/deficiencia , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Neutrófilos/enzimología , Animales , Apoptosis/genética , Biomarcadores , Biopsia , Modelos Animales de Enfermedad , Pruebas de Función Cardíaca , Insulinas/metabolismo , Elastasa de Leucocito/metabolismo , Ratones , Ratones Noqueados , Infarto del Miocardio/mortalidad , Infarto del Miocardio/patología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Remodelación Ventricular/genética
11.
Int J Mol Sci ; 22(3)2021 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-33572655

RESUMEN

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.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Antígeno B7-H1/inmunología , Miocarditis/inmunología , Proteína 2 Ligando de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Animales , Enfermedades Autoinmunes/patología , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Células Dendríticas/inmunología , Masculino , Ratones , Miocarditis/patología
12.
Int Heart J ; 62(5): 1171-1175, 2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34497169

RESUMEN

Erlotinib, an epidermal growth factor receptor tyrosine kinase inhibitor, is a targeted drug used for the treatment of non-small cell lung cancer (NSCLC). Erlotinib is considered relatively safe and generally well-tolerated, with rarely reported cardiac side effects. Herein, we report a case of cardiomyopathy that developed during erlotinib treatment for NSCLC. Two months after erlotinib initiation, our 70 year-old female patient complained of progressive dyspnea, and a diagnostic endomyocardial biopsy confirmed non-specific cardiomyopathy, indicating erlotinib-induced cardiomyopathy. We believed that continued administration of erlotinib would exacerbate her heart failure, while treatment of the heart failure with intensive monitoring would allow the administration of erlotinib to be continued. This case report highlights the potential cardiotoxic effects of erlotinib and suggests the need for close clinical and echocardiographic follow-up of patients receiving erlotinib.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cardiomiopatías/inducido químicamente , Clorhidrato de Erlotinib/toxicidad , Inhibidores de Proteínas Quinasas/toxicidad , Anciano , Biopsia/métodos , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Carcinoma de Pulmón de Células no Pequeñas/secundario , Cardiomiopatías/patología , Cardiotoxicidad/complicaciones , Progresión de la Enfermedad , Disnea/diagnóstico , Disnea/etiología , Clorhidrato de Erlotinib/efectos adversos , Clorhidrato de Erlotinib/uso terapéutico , Femenino , Estudios de Seguimiento , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/etiología , Humanos , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia/patología , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/uso terapéutico
13.
J Cell Mol Med ; 24(24): 14481-14490, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33140535

RESUMEN

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.


Asunto(s)
Activación de Macrófagos/genética , Macrófagos/metabolismo , Infarto del Miocardio/complicaciones , Receptores de Inmunoglobulina Polimérica/deficiencia , Receptor Toll-Like 9/metabolismo , Remodelación Ventricular/genética , Animales , Biomarcadores , Biopsia , Citocinas/metabolismo , ADN Mitocondrial , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Ecocardiografía , Mediadores de Inflamación/metabolismo , Estimación de Kaplan-Meier , Macrófagos/inmunología , Ratones , Ratones Noqueados , Infarto del Miocardio/etiología , Infarto del Miocardio/mortalidad , Miocardio/patología , Pronóstico
14.
BMC Cardiovasc Disord ; 20(1): 451, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33059582

RESUMEN

BACKGROUND: Cigarette smoking harms nearly every organ, including the heart and lungs. A comprehensive assessment of both cardiac and respiratory function is necessary for evaluating the direct effects of tobacco on the heart. However, few previous studies examining the effects of cigarette smoking on cardiac function included an assessment of lung function. This cross-sectional study investigated the influence of cigarette smoking on cardiac function, independent of respiratory function. METHODS: We retrospectively reviewed the medical records of 184 consecutive cases that underwent both spirometry and transthoracic echocardiography around the same time (within 1 month) in one hospital from April 2019 to March 2020. Participants were classified into three groups based on lifetime smoking exposure (pack-years): non-smoker (n = 49), low exposure (1-20 pack-years, n = 40), and high exposure (≥ 20 pack years, n = 95). Multiple linear regression analysis was used to assess the relationship among cigarette smoking, and cardiac and respiratory functions. The relationship between selected dependent variables and lifetime pack-years was assessed in two models with multiple linear regression analysis. Model 1 was adjusted for age and male sex; and Model 2 was adjusted for Model 1 plus forced expiratory volume percentage in 1 s and forced vital capacity percentage. RESULTS: Compared with the non-smokers, the participants with high smoking exposure had lower left ventricular (LV) systolic function and larger LV size. Multiple linear regression analysis revealed a negative association of cumulative lifetime pack-years with LV and right ventricular (RV) systolic functions, even after adjustment for age, sex, and spirometric parameters (forced expiratory volume percentage in 1 s and forced vital capacity percentage). Meanwhile, there was no significant association of smoking exposure with LV diastolic function (E/e' and E/A) and RV diastolic function (e't and e't/a't). CONCLUSIONS: Cumulative smoking exposure was associated with a negative effect on biventricular systolic function in patients with relatively preserved cardiac function, independent of respiratory function.


Asunto(s)
Hipertrofia Ventricular Izquierda/etiología , Fumar/efectos adversos , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Derecha/etiología , Función Ventricular Izquierda , Función Ventricular Derecha , Anciano , Anciano de 80 o más Años , Estudios Transversales , Ecocardiografía Doppler , Femenino , Volumen Espiratorio Forzado , Factores de Riesgo de Enfermedad Cardiaca , Humanos , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/fisiopatología , Pulmón/fisiopatología , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Medición de Riesgo , Fumar/fisiopatología , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/fisiopatología , Disfunción Ventricular Derecha/diagnóstico por imagen , Disfunción Ventricular Derecha/fisiopatología , Remodelación Ventricular , Capacidad Vital
15.
Jpn J Clin Oncol ; 50(12): 1419-1425, 2020 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-32676637

RESUMEN

OBJECTIVE: Onco-cardiology services are expanding rapidly in Japan. To provide a better service, it is important to consider the needs of oncologists. However, little is known regarding specific needs for which oncologists should consult cardiologists to manage cardiovascular problems of their patients. We analysed cardiology consultations sought by oncologists to evaluate the role of cardiologists in cancer treatment. METHOD: We retrospectively investigated consecutive 2064 cardiology consultations of cancer patients in the University of Tsukuba Hospital, Tsukuba, Japan, between January 2014 and December 2018. RESULTS: The most common timing of cardiology consultation was before the commencement of cancer treatment (n = 1355; 65.7%), followed by after the commencement of cancer treatment (n = 686; 33.2%). Among the 361 consultations before the administration of anticancer drugs, 235 (65.1%) were for anthracycline-based regimens. There were 506 (24.5%) consultations for the management of cardiovascular emergencies developing after the commencement of cancer treatment; venous thromboembolism was the most frequent (n = 125; 24.7%), followed by atrial fibrillation (n = 110; 21.7%) and heart failure (n = 74; 14.6%). There were marked differences in the types of cardiovascular emergencies depending on the type of cancer. CONCLUSIONS: This survey revealed the various cardiovascular problems for which oncologists sought interventions by cardiologists. A multidisciplinary approach in an onco-cardiology service is essential to achieve optimal long-term outcomes.


Asunto(s)
Cardiología , Oncología Médica/estadística & datos numéricos , Derivación y Consulta , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/terapia , Niño , Femenino , Humanos , Japón , Masculino , Oncología Médica/tendencias , Persona de Mediana Edad , Neoplasias/complicaciones , Neoplasias/terapia , Estudios Retrospectivos , Encuestas y Cuestionarios , Adulto Joven
16.
Int J Mol Sci ; 21(13)2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32629939

RESUMEN

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.


Asunto(s)
Fibrilación Atrial/enzimología , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Animales , Fibrilación Atrial/etiología , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Dieta Alta en Grasa/efectos adversos , Atrios Cardíacos/enzimología , Masculino , Ratones Noqueados , Obesidad/complicaciones , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
17.
Gan To Kagaku Ryoho ; 47(6): 881-884, 2020 Jun.
Artículo en Japonés | MEDLINE | ID: mdl-32541160

RESUMEN

The causes of cardiac tamponade include neoplastic pericarditis as well as radiation-induced, drug-induced, purulent and iatrogenic pericarditis. Since we can get access to the advanced cancer therapy these days, some of the cardiac tamponade patients can survive longer if we can manage their pericardial effusion well. Here, we will summarize the clinical evidence in cardiac tamponade in patients with malignant tumor, and will discuss about its presentation, diagnosis and management.


Asunto(s)
Taponamiento Cardíaco , Neoplasias , Taponamiento Cardíaco/etiología , Humanos , Neoplasias/complicaciones , Derrame Pericárdico , Pericarditis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA