Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
BMC Med Inform Decis Mak ; 24(1): 17, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38225576

RESUMO

BACKGROUND: Heart failure is a syndrome with complex clinical manifestations. Due to increasing population aging, heart failure has become a major medical problem worldwide. In this study, we used the MIMIC-III public database to extract the temporal and spatial characteristics of electrocardiogram (ECG) signals from patients with heart failure. METHODS: We developed a NYHA functional classification model for heart failure based on a deep learning method. We introduced an integrating attention mechanism based on the CNN-LSTM-SE model, segmenting the ECG signal into 2 to 20 s long segments. Ablation experiments showed that the 12 s ECG signal segments could be used with the proposed deep learning model for superior classification of heart failure. RESULTS: The accuracy, positive predictive value, sensitivity, and specificity of the NYHA functional classification method were 99.09, 98.9855, 99.033, and 99.649%, respectively. CONCLUSIONS: The comprehensive performance of this model exceeds similar methods and can be used to assist in clinical medical diagnoses.


Assuntos
Aprendizado Profundo , Insuficiência Cardíaca , Humanos , Arritmias Cardíacas/diagnóstico , Eletrocardiografia/métodos , Insuficiência Cardíaca/diagnóstico , Bases de Dados Factuais , Algoritmos
2.
J Physiol Biochem ; 76(3): 445-456, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32592088

RESUMO

Increased evidence indicates that adenosine monophosphate-activated protein kinase (AMPK) plays a vital role in vascular homeostasis, especially under hypoxia, and protects against the progression of pulmonary hypertension (PH). However, the role of AMPK in the pathogenesis of PH remains to be clarified. In the present study, we confirmed that a loss of AMPKα2 exacerbated the development of PH by using hypoxia-induced PH model in AMPKα2 -/- mice. After a 4-week period of hypoxic exposure, AMPKα2 -/- mice exhibited more severe pulmonary vascular remodeling and pulmonary vascular smooth muscle cell (SMC) proliferation when compared with wild type (WT) mice. In vitro, AMPKα2 knockdown promoted the proliferation of pulmonary arterial smooth muscle cells (PASMCs) under hypoxia. This phenomenon was accompanied by upregulated Skp2 and downregulated p27kip1 expression and was abolished by rapamycin, an inhibitor of mTOR. These results indicate that AMPKα2 deficiency exacerbates hypoxia-induced PH by promoting PASMC proliferation via the mTOR/Skp2/p27kip1 signaling axis. Therefore, enhanced AMPKα2 activity might underlie a novel therapeutic strategy for the management of PH.


Assuntos
Proteínas Quinases Ativadas por AMP/deficiência , Hipóxia/patologia , Miócitos de Músculo Liso/citologia , Hipertensão Arterial Pulmonar/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Hipóxia Celular , Linhagem Celular , Proliferação de Células , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/citologia , Artéria Pulmonar/citologia , Ratos
3.
Mol Med Rep ; 21(3): 1336-1345, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31922214

RESUMO

The aim of the present study was to investigate the protective effect of Kruppel­like factor 15 (KLF15) overexpression on heart failure (HF) induced by left ventricular (LV) pressure overload in mice. Wild­type (WT) mice and cardiac­specific KLF15­overexpressed transgenic (TG) mice were selected as research subjects, and an LV pressure overload model was constructed by ascending aortic constriction surgery. Changes in cardiac morphology and function, and ultrastructure and molecular expression were observed via M­mode echocardiography, histological and immunohistochemical staining, ELISA and western blotting at 2 and 6 weeks of LV overload. WT and TG mice subjected to 2 weeks of overload displayed adaptive LV hypertrophy characterized by ventricular thickness, cardiomyocyte size, ejection fraction and fractional shortening of heart­lung weight ratio and KLF15, and increases in vascular endothelial growth factor (VEGF) expression without other pathological changes. WT mice subjected to 6 weeks of overload displayed enlargement of the LV chamber, severe interstitial remodeling, and HW/LW, cardiac capillary and heart function decline, accompanied by downregulated expression of KLF15 and VEGF, and upregulated expression of connective tissue growth factor, phosphorylated p38 (p­p38) and phosphorylated Smad3 (p­Smad3). In contrast, TG mice exhibited improved resistance to 6 weeks of overload and a slighter molecular expression response compared with WT mice. KLF15 was revealed to be a critical factor regulating the expression of CTGF, VEGF, p­p38 and p­Smad3, and could alleviate the progression from adaptive LV hypertrophy to decompensatory cardiac insufficiency.


Assuntos
Pressão Sanguínea , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Fatores de Transcrição Kruppel-Like/metabolismo , Animais , Modelos Animais de Doenças , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/prevenção & controle , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Transgênicos
4.
J Mol Cell Cardiol ; 138: 115-135, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31783035

RESUMO

AIMS: There is a large subpopulation of multinucleated polyploid cardiomyocytes (M*Pc CMs) in the adult mammalian heart. However, the pathophysiological significance of increased M*Pc CMs in heart disease is poorly understood. We sought to determine the pathophysiological significance of increased M*Pc CMs during hypoxia adaptation. METHODS AND RESULTS: A model of hypoxia-induced cardiomyocyte (CM) multinucleation and polyploidization was established and found to be associated with less apoptosis and less reactive oxygen species (ROS) production. Compared to mononucleated diploid CMs (1*2c CMs), tetraploid CMs (4c CMs) exhibited better mitochondria quality control via increased mitochondrial autophagy (mitophagy). RNA-seq revealed Prkaa2, the gene for AMPKα2, was the most obviously up-regulated autophagy-related gene. Knockdown of AMPKα2 increased apoptosis and ROS production and suppressed mitophagy in 4c CMs compared to 1*2c CMs. Rapamycin, an autophagy activator, alleviated the adverse effect of AMPKα2 knockdown. Furthermore, silencing PINK1 also increased apoptosis and ROS in 4c CMs and weakened the adaptive superiority of 4c CMs. Finally, AMPKα2-/- mutant mice exhibited exacerbation of apoptosis and ROS production via decreases in AMPKα2-mediated mitophagy in 4c CMs compared to 1*2c CMs during hypoxia. CONCLUSIONS: Compared to 1*2c CMs, hypoxia-induced 4c CMs exhibited enhanced mitochondria quality control and less apoptosis via AMPKα2-mediated mitophagy. These results suggest that multinucleation and polyploidization allow CM to better adapt to stress via enhanced mitophagy. In addition, activation of AMPKα2 may be a promising target for myocardial hypoxia-related diseases.


Assuntos
Adaptação Fisiológica , Células Gigantes/patologia , Mitofagia , Miócitos Cardíacos/patologia , Poliploidia , Adenilato Quinase/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Hipóxia Celular , Inativação Gênica , Masculino , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Quinases/metabolismo , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
5.
Sci Rep ; 9(1): 17865, 2019 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-31780774

RESUMO

Cardiomyocyte (CM) loss is a characteristic of various heart diseases, including ischaemic heart disease. Cardiac regeneration has been suggested as a promising strategy to address CM loss. Although many studies of regeneration have focused mainly on mononucleated or diploid CM, the limitations associated with the cytokinesis of polyploid and multinucleated CMs remain less well known. Here, we show that ß-catenin, a key regulator in heart development, can increase cytokinesis in polyploid multinucleated CMs. The activation of ß-catenin increases the expression of the cytokinesis-related factor epithelial cell transforming 2 (ECT2), which regulates the actomyosin ring and thus leads to the completion of cytokinesis in polyploid CMs. In addition, hypoxia can induce polyploid and multinucleated CMs by increasing factors related to the G1-S-anaphase of the cell cycle, but not those related to cytokinesis. Our study therefore reveals that the ß-catenin can promote the cytokinesis of polyploid multinucleated CMs via upregulation of ECT2. These findings suggest a potential field of polyploid CM research that may be exploitable for cardiac regeneration therapy.


Assuntos
Cardiomegalia/metabolismo , Citocinese , Miócitos Cardíacos/metabolismo , Poliploidia , beta Catenina/metabolismo , Actomiosina/metabolismo , Animais , Hipóxia Celular , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/metabolismo , Proteínas Proto-Oncogênicas/metabolismo
6.
Int J Clin Exp Pathol ; 11(12): 5840-5849, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-31949670

RESUMO

Hypoxia-induced apoptosis is an inevitable problem in cyanotic congenital heart disease. In the present study, we investigated effects of melatonin on hypoxic cardiomyocytes in vitro and in vivo, and explored its underlying mechanism. H9C2 cells were subjected to hypoxia for 48 hours. Mice were subjected to hypoxia treatment (10% O2) for 4 weeks. Cell viability was detected by the cell counting kit-8 assay. Cellular apoptosis was assessed by Annexin V/7 AAD assay. Western blotting was employed to determine the expression of Bcl-2, Bax, cleaved caspase 3, phosphorylation of PI3K, and AKT. Melatonin increased cell viability and alleviated apoptosis in hypoxic H9C2 cells and cardiomyocytes of hypoxia-treated mice. Melatonin pretreatment increased Bcl-2 and decreased cleaved caspase 3 and Bax levels. Moreover, melatonin activated the PI3K/Akt pathway. The protective effects of melatonin were abolished by a PI3K/Akt-inhibitor, LY294002. Our results demonstrated that melatonin confers cardioprotection by inhibiting apoptosis through the activation of PI3K/Akt signaling pathway in hypoxic cardiomyocytes.

7.
Mol Med Rep ; 15(6): 3541-3548, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28393210

RESUMO

Induction of cardiomyocyte proliferation, the most promising approach to reverse myocardial attrition, has been gaining importance as a therapy for cardiovascular disease. Hypoxia and macrophages were previously independently reported to promote cardiomyocyte proliferation in mice. However, whether hypoxia promotes cardiomyocyte proliferation in humans, and the association between hypoxia and macrophages in cardiomyocyte proliferation, have not to the best of our knowledge been previously investigated. The present study investigated the cardiomyocyte proliferation in 22 acyanotic and 29 cyanotic patients. Cardiomyocyte proliferation in a hypoxic mouse model (15% O2) was subsequently performed and the macrophage subsets were analyzed. A C­C chemokine receptor type 2 (CCR2) inhibitor was used to increase the number of resident macrophages in order to investigate the effect of macrophages on cardiomyocyte proliferation. The results demonstrated that cardiomyocyte proliferation in the cyanotic infant group was significantly increased compared with the acyanotic infant group and the hypoxia­treated C57BL/6J neonates confirmed the hypoxia­induced cardiomyocyte proliferation. However, hypoxia did not induce the proliferation of isolated cardiomyocytes. Notably, hypoxia treatment increased the number of cardiac resident macrophages in neonate hearts. Furthermore, increasing the number of resident macrophages significantly enhanced cardiomyocyte proliferation. In conclusion, postnatal hypoxia promoted cardiomyocyte proliferation in humans and animals, and cardiac resident macrophages may be involved in this process. Therefore, this novel mechanism may provide a promising strategy for cardiovascular disease treatment.


Assuntos
Hipóxia/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Adolescente , Adulto , Animais , Animais Recém-Nascidos , Biomarcadores , Proliferação de Células , Células Cultivadas , Criança , Pré-Escolar , Modelos Animais de Doenças , Feminino , Humanos , Lactente , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Receptores CCR2/antagonistas & inibidores , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA