Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Cell Mol Med ; 28(13): e18493, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38963241

RESUMO

Interleukin-5 (IL-5) has been reported to be involved in cardiovascular diseases, such as atherosclerosis and cardiac injury. This study aimed to investigate the effects of IL-5 on cardiac remodelling. Mice were infused with angiotensin II (Ang II), and the expression and source of cardiac IL-5 were analysed. The results showed that cardiac IL-5 expression was time- and dose-dependently decreased after Ang II infusion, and was mainly derived from cardiac macrophages. Additionally, IL-5-knockout (IL-5-/-) mice were used to observe the effects of IL-5 knockout on Ang II-induced cardiac remodelling. We found knockout of IL-5 significantly increased the expression of cardiac hypertrophy markers, elevated myocardial cell cross-sectional areas and worsened cardiac dysfunction in Ang II-infused mice. IL-5 deletion also promoted M2 macrophage differentiation and exacerbated cardiac fibrosis. Furthermore, the effects of IL-5 deletion on cardiac remodelling was detected after the STAT3 pathway was inhibited by S31-201. The effects of IL-5 on cardiac remodelling and M2 macrophage differentiation were reversed by S31-201. Finally, the effects of IL-5 on macrophage differentiation and macrophage-related cardiac hypertrophy and fibrosis were analysed in vitro. IL-5 knockout significantly increased the Ang II-induced mRNA expression of cardiac hypertrophy markers in myocardial cells that were co-cultured with macrophages, and this effect was reversed by S31-201. Similar trends in the mRNA levels of fibrosis markers were observed when cardiac fibroblasts and macrophages were co-cultured. In conclusions, IL-5 deficiency promote the differentiation of M2 macrophages by activating the STAT3 pathway, thereby exacerbating cardiac remodelling in Ang II-infused mice. IL-5 may be a potential target for the clinical prevention of cardiac remodelling.


Assuntos
Angiotensina II , Cardiomegalia , Fibrose , Interleucina-5 , Macrófagos , Camundongos Knockout , Fator de Transcrição STAT3 , Transdução de Sinais , Remodelação Ventricular , Animais , Angiotensina II/farmacologia , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/genética , Remodelação Ventricular/efeitos dos fármacos , Camundongos , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Interleucina-5/metabolismo , Interleucina-5/genética , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Cardiomegalia/genética , Cardiomegalia/induzido quimicamente , Masculino , Camundongos Endogâmicos C57BL , Diferenciação Celular , Miocárdio/metabolismo , Miocárdio/patologia
2.
Europace ; 26(7)2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38801673

RESUMO

AIMS: Radiofrequency ablation is used as a first-line therapy for accessory pathways (APs). However, data regarding the effects of pulsed field ablation (PFA) on APs are limited. We sought to evaluate the acute procedural and 6-month success and safety of PFA in a cohort of patients with APs. METHODS AND RESULTS: A focal contact force-sensing PFA catheter was used for patients with APs. Pulsed field ablation generator generated a bipolar and biphasic waveform (±1000 V) with a duration of 100 ms from the tip of the PFA catheter. A 100% acute procedural success was achieved in 10 conscious patients with APs (7 left anterolateral, 2 left inferolateral, and 1 right posteroseptal APs) including 6 (60%) patients after an initial application. The average total ablation time was 6.3 ± 4.9 s for 4.7 ± 1.8 ablation sites (ASs), including 3.1 ± 2.4 s at targets and 3.2 ± 2.9 s at 3.2 ± 2 bolus ASs. The mean skin-to-skin time was 59.3 ± 15.5 min, and PFA catheter dwell time was 29.4 ± 7.8 min. One patient encountered transient sinus arrest during PFA due to parasympathetic overexcitation. Sinus rhythm was restored in all patients without any significant adverse events during the short-term follow-up. CONCLUSION: Pulsed field ablation of APs was feasible, effective, and safe. Its efficiency was remarkable for its ultrarapid termination of AP conduction. Further studies are warranted to prove whether utilization of PFA with current parameters can extend to manifold AP ablation.


Assuntos
Feixe Acessório Atrioventricular , Ablação por Cateter , Humanos , Projetos Piloto , Feminino , Masculino , Feixe Acessório Atrioventricular/cirurgia , Feixe Acessório Atrioventricular/fisiopatologia , Resultado do Tratamento , Adulto , Ablação por Cateter/métodos , Ablação por Cateter/efeitos adversos , Pessoa de Meia-Idade , Adulto Jovem , Fatores de Tempo , Frequência Cardíaca , Adolescente , Cateteres Cardíacos
3.
J Cell Mol Med ; 27(17): 2562-2571, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37584247

RESUMO

Previous studies have reported that visfatin can regulate macrophage polarisation, which has been demonstrated to participate in cardiac remodelling. The aims of this study were to investigate whether visfatin participates in transverse aortic constriction (TAC)-induced cardiac remodelling by regulating macrophage polarisation. First, TAC surgery and angiotensin II (Ang II) infusion were used to establish a mouse cardiac remodelling model, visfatin expression was measured, and the results showed that TAC surgery or Ang II infusion increased visfatin expression in the serum and heart in mice, and phenylephrine or hydrogen peroxide promoted the release of visfatin from macrophages in vitro. All these effects were dose-dependently reduced by superoxide dismutase. Second, visfatin was administered to TAC mice to observe the effects of visfatin on cardiac remodelling. We found that visfatin increased the cross-sectional area of cardiomyocytes, aggravated cardiac fibrosis, exacerbated cardiac dysfunction, further regulated macrophage polarisation and aggravated oxidative stress in TAC mice. Finally, macrophages were depleted in TAC mice to investigate whether macrophages mediate the regulatory effect of visfatin on cardiac remodelling, and the results showed that the aggravating effects of visfatin on oxidative stress and cardiac remodelling were abrogated. Our study suggests that visfatin enhances cardiac remodelling by promoting macrophage polarisation and enhancing oxidative stress. Visfatin may be a potential target for the prevention and treatment of clinical cardiac remodelling.


Assuntos
Estenose da Valva Aórtica , Remodelação Ventricular , Camundongos , Animais , Nicotinamida Fosforribosiltransferase/metabolismo , Constrição , Miócitos Cardíacos/metabolismo , Estenose da Valva Aórtica/metabolismo , Macrófagos/metabolismo , Estresse Oxidativo , Angiotensina II/metabolismo , Camundongos Endogâmicos C57BL , Fibrose , Cardiomegalia/metabolismo
4.
Artigo em Inglês | MEDLINE | ID: mdl-37256416

RESUMO

BACKGROUND: C1q/tumor necrosis factor-related protein 5 (CTRP5) has been reported to be a crucial regulator in cardiac ischemia/reperfusion (I/R) injury. Nevertheless, the potential role of CTRP5 in doxorubicin (DOX)-induced cardiotoxicity and the potential mechanisms remain largely unclear. METHODS: We overexpressed CTRP5 in the hearts using an adeno-associated virus 9 (AAV9) system through tail vein injection. C57BL/6 mice were subjected to DOX (15 mg/kg/day, i.p.) to generate DOX-induced cardiotoxicity for 4 weeks. Subsequently, cardiac staining and molecular biological analysis were performed to analyze the morphological and biochemical effects of CTRP5 on the cardiac injury. H9c2 cells were used for validation in vitro. RESULTS: CTRP5 expression was down-regulated after DOX treatment both in vivo and in vitro. CTRP5 overexpression significantly attenuated DOX-induced cardiac injury, cardiac dysfunction, inhibited oxidative stress and inflammatory response. Mechanistically, CTRP5 overexpression markedly decreased the protein expression of toll-like receptor 4 (TLR4), NLRP3, cleaved caspase-1 and caspase-1, indicating TLR/NLRP3 signaling contributes to the cardioprotective role of CTRP5 in DOX-induced cardiotoxicity. CONCLUSIONS: Together, our findings demonstrated that CTRP5 overexpression could protect the heart from oxidative stress and inflammatory injury induced by DOX through inhibiting TLR4/NLRP3 signaling, suggesting that CTRP5 might be a potential therapeutic target in the prevention of DOX-induced cardiotoxicity.

5.
Artigo em Inglês | MEDLINE | ID: mdl-38141139

RESUMO

BACKGROUND: Diabetic cardiomyopathy (DCM) pathogenesis is a common complication of diabetes, but effective treatments remain limited. Mitochondrial-derived peptide MOTS-c has shown therapeutic promise in animal models of various heart diseases, but its efficacy in DCM is unknown. This study investigates the effects of MOTS-c treatment in a mouse model of type 1 diabetes-induced DCM. METHODS: Type 1 diabetes (T1DM) was induced in mice by streptozotocin (STZ) injection. After diabetes establishment, the mice were randomly dividend into two groups treated with or without MOTS-c peptide, which was administered subcutaneously by osmotic pump for 12 weeks. At the end of the experiment, cardiac function, histology, and molecular changes were determined. RESULTS: The results showed that diabetic mice exhibited significant cardiac dysfunction, dilatation, and adverse cardiac remodeling. MOTS-c treatment markedly ameliorated these diabetes-associated myocardial function and structure abnormalities. Additionally, MOTS-c reversed AMPK signaling deactivation and inhibited inflammation in the diabetic heart. CONCLUSIONS: Our data demonstrated a protective effect of MOTS-c against diabetic cardiomyopathy potentially by activating the AMPK pathway and inhibiting inflammation. These findings demonstrate the therapeutic efficacy of MOTS-c for diabetic cardiomyopathy and warrant further investigation into its clinical potential.

6.
J Cell Mol Med ; 26(21): 5369-5378, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36156853

RESUMO

MOTS-c, a mitochondrial-derived peptide (MDP), has been shown to have multiple biological activities such as antioxidation, anti-inflammation, and anti-apoptosis properties. In the present study, we aimed at evaluating the therapeutic effect of MOTS-c peptide in an animal model of heart failure. The heart failure mouse model was made by transverse aortic constriction (TAC) operations. The MOTS-c peptide was administrated subcutaneously by using an osmotic pump. At the end of the animal experiment, cardiac function was evaluated by echocardiography, and heart tissues were subjected to histological and molecular analysis. In vitro cultured H9C2 cells were used to test the effects of MOTS-c overexpression on cell death in response to H2 O2 stimulation. Our study showed that MOTS-c peptide attenuated TAC-induced cardiac dysfunction and remodelling. In addition, the MOTS-c peptide reduced the inflammatory response and upregulated the antioxidant capacity, coupled with the activation of the AMPK pathway in the heart of the TAC mouse model. In in vitro cultured cardiac cells, overexpression of MOTS-c was shown to activate the AMPK pathway and protect cell apoptosis in response to H2 O2 stimulation. Taken together, our study suggested that MOTS-c peptides may have therapeutic potential in treating HF.


Assuntos
Proteínas Quinases Ativadas por AMP , Insuficiência Cardíaca , Camundongos , Animais , Proteínas Quinases Ativadas por AMP/metabolismo , Peptídeo C , Insuficiência Cardíaca/metabolismo , Coração , Modelos Animais de Doenças , Antioxidantes , Peptídeos/farmacologia , Camundongos Endogâmicos C57BL
7.
J Cardiovasc Pharmacol ; 80(6): 804-812, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-35856909

RESUMO

ABSTRACT: Grb2-associated binding protein 1 (Gab1), an intracellular scaffolding adaptor, was involved in several cardiovascular diseases. However, the role of Gab1 in doxorubicin (DOX)-induced cardiotoxicity remains largely unknown. The present study investigated whether Gab1 protected against DOX-induced cardiotoxicity and the underlying mechanism. We overexpressed Gab1 in the hearts using an adeno-associated virus 9 system through tail vein injection. C57BL/6 mice were subjected to DOX (15 mg/kg/d, i.p.) to generate DOX-induced cardiotoxicity. Echocardiography, histological analysis, immunofluorescence and enzyme-linked immunosorbent assay (ELISA) kits, Western blotting, and quantitative real-time polymerase chain reaction (PCR) evaluated DOX-induced cardiotoxicity and the underlying mechanisms. Myocardial Gab1 protein and messenger RNA (mRNA) levels were markedly decreased in DOX-administered mice. Overexpression of Gab1 in myocardium significantly improved cardiac function and attenuated cardiac oxidative stress, inflammatory response, and apoptosis induced by DOX. Mechanistically, we found that PI3K/Akt signaling pathway was downregulated after DOX treatment, and Gab1 overexpression activated PI3K/Akt signaling pathway, whereas PI3K/Akt signaling pathway inhibition abolished the beneficial effect of Gab1 overexpression in the heart. Collectively, our results indicated that Gab1 is essential for cardioprotection against DOX-induced oxidative stress, inflammatory response, and apoptosis by mediating PI3K/Akt signaling pathway. And cardiac gene therapy with Gab1 provides a novel therapeutic strategy against DOX-induced cardiotoxicity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Apoptose , Cardiotoxicidade , Doxorrubicina , Estresse Oxidativo , Animais , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/genética , Doxorrubicina/toxicidade , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases , Transdução de Sinais
8.
Mediators Inflamm ; 2022: 3306559, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36262545

RESUMO

Background: Visfatin is an adipocytokine that has been demonstrated to be involved in cardiovascular diseases. This study aims at determining the role of visfatin in sepsis-induced cardiac injury and identify its possible mechanisms. Methods: Dynamic changes in visfatin expression in mice with lipopolysaccharide- (LPS-) induced septicemia were measured. Additionally, mice were pretreated with visfatin and further administered LPS to observe the effects of visfatin on cardiac injury. Finally, septic mice were also pretreated with JSH-23 to investigate whether visfatin regulates cardiac injury via the NF-κB p65 pathway. Results: Visfatin expression levels in both the heart and serum were increased in LPS-treated mice and peaked at 6 hours, and visfatin was derived from cardiac macrophages. In septic mice, pretreatment with visfatin reduced the survival rate, worsened cardiac dysfunction, and increased the expression of cardiac injury markers, including creatine kinase myocardial bound (CK-MB) and lactate dehydrogenase (LDH). Treatment with visfatin also increased the infiltration of CD3+ cells and F4/80+ cells, amplified the cardiac inflammatory response, and elevated myocardial cell apoptosis. Treatment with JSH-23 reversed the effects of visfatin in septic mice. Conclusions: This study showed that visfatin amplifies the cardiac inflammatory response and aggravates cardiac injury through the p65 signaling pathway. Visfatin may be a clinical target for preventing cardiac injury in sepsis.


Assuntos
Traumatismos Cardíacos , Nicotinamida Fosforribosiltransferase , Sepse , Animais , Camundongos , Adipocinas , Creatina Quinase/metabolismo , Lactato Desidrogenases/metabolismo , Lipopolissacarídeos/toxicidade , NF-kappa B/metabolismo , Nicotinamida Fosforribosiltransferase/metabolismo , Sepse/tratamento farmacológico , Sepse/metabolismo , Transdução de Sinais , Citocinas/metabolismo
9.
Yi Chuan ; 44(3): 198-207, 2022 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-35307643

RESUMO

Hypertrophic cardiomyopathy (HCM) is an autosomal dominant genetic disease characterized by left ventricular hypertrophy with prevalence of 1/500-1/200. Up to now, 1500 mutations in more than 30 genes have been found to be related to the disease. Pathogenic gene mutations together with polymorphisms of modifying genes and environmental factors play various roles in the disease processes, resulting in phenotypic heterogeneity of the disease, ranging from no symptoms to sudden cardiac death. The pathological phenotypes of HCM mainly include cardiomyocyte hypertrophy, disordered array, fibrosis, myocardial ischemia, and others. In recent years, many research efforts have been devoted to exploring the influence of HCM genotype on phenotype, and development of treatment methods based on genetics. This article focuses on the correction between HCM genotype and phenotype and summarizes the research progresses on HCM in terms of pathogenic genes, pathogenesis, associated modification factors and treatment methods, thereby providing insights on the future research and development on the genetics of HCM.


Assuntos
Cardiomiopatia Hipertrófica , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/patologia , Genótipo , Humanos , Mutação , Fenótipo
10.
J Formos Med Assoc ; 119(11): 1586-1592, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32703696

RESUMO

BACKGROUND/PURPOSE: Left atrial appendage closure (LAAC) is conventionally guided by fluoroscopy and transesophageal echocardiography. We introduce an LAAC technique without fluoroscopy exposure using intracardiac echocardiography (ICE) and electroanatomic mapping system (EAMS) under local anesthesia. METHODS: Seven non-valvular atrial fibrillation patients who underwent LAAC with the LAmbre devices under the ICE and EAMS guidance were compared to 7 patients following the conventional approach by propensity score matching. ICE probe was advanced into the left atrium (LA) to guide sizing and device implantation following the orthogonal tri-axial technique (Axis-X: from left pulmonary veins [PVs] to LAA; Axis-Y: from right PV ostium to LAA; Axis-Z: from lower LA to LAA). RESULTS: The mean diameters of ostia and landing zone were 21.4 ± 3.9 mm and 20.4 ± 4.2 mm, respectively. LAmbre devices with a mean umbrella diameter of 23.7 ± 4.2 mm and cover disc diameter of 29.4 ± 3.6 mm were successfully implanted and acute complete LAA sealing without peri-device leak (PDL) were achieved in all cases. Neither fluoroscopy exposure nor contrast consumption was recorded. No procedure related complications were documented. The mean procedural time and PDLs at follow-ups were comparable to those in the conventional group. No stroke or thromboembolic events were documented. CONCLUSION: The fluoroscopy exposure could be minimized, even to zero, in the ICEguided LAAC procedures feasibly and safely using LAmbre devices. The orthogonal triaxial technique is considered efficacious and safe for the procedures.


Assuntos
Apêndice Atrial , Apêndice Atrial/diagnóstico por imagem , Apêndice Atrial/cirurgia , Fibrilação Atrial/diagnóstico por imagem , Fibrilação Atrial/cirurgia , Cateterismo Cardíaco , Ecocardiografia Transesofagiana , Fluoroscopia , Humanos , Resultado do Tratamento
11.
J Cell Mol Med ; 23(8): 5303-5316, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31140723

RESUMO

Myeloid differentiation 1 (MD-1) is a secreted protein that regulates the immune response of B cell through interacting with radioprotective 105 (RP105). Disrupted immune response may contribute to the development of cardiac diseases, while the roles of MD-1 remain elusive. Our studies aimed to explore the functions and molecular mechanisms of MD-1 in obesity-induced cardiomyopathy. H9C2 myocardial cells were treated with free fatty acid (FFA) containing palmitic acid and oleic acid to challenge high-fat stimulation and adenoviruses harbouring human MD-1 coding sequences or shRNA for MD-1 overexpression or knockdown in vitro. MD-1 overexpression or knockdown transgenic mice were generated to assess the effects of MD-1 on high-fat diet (HD) induced cardiomyopathy in vivo. Our results showed that MD-1 was down-regulated in H9C2 cells exposed to FFA stimulation for 48 hours and in obesity mice induced by HD for 20 weeks. Both in vivo and in vitro, silencing of MD-1 accelerated myocardial function injury induced by HD stimulation through increased cardiac hypertrophy and fibrosis, while overexpression of MD-1 alleviated the effects of HD by inhibiting the process of cardiac remodelling. Moreover, the MAPK and NF-κB pathways were overactivated in MD-1 deficient mice and H9C2 cells after high-fat treatment. Inhibition of MAPK and NF-κB pathways played a cardioprotective role against the adverse effects of MD-1 silencing on high-fat stimulation induced pathological remodelling. In conclusion, MD-1 protected myocardial function against high-fat stimulation induced cardiac pathological remodelling through negative regulation for MAPK/NF-κB signalling pathways, providing feasible strategies for obesity cardiomyopathy.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Células Mieloides/metabolismo , Miócitos Cardíacos/metabolismo , Extratos Vegetais/metabolismo , Animais , Cardiomegalia/metabolismo , Cardiomiopatias/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Fibrose/metabolismo , Camundongos , Miocárdio/metabolismo , NF-kappa B/metabolismo , Obesidade/metabolismo , Ratos , Transdução de Sinais/fisiologia
12.
Nutr Metab Cardiovasc Dis ; 29(9): 991-998, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31353205

RESUMO

BACKGROUND AND AIM: Obesity is an important risk factor for ventricular arrhythmia (VA), and myeloid differentiation protein 1 (MD1) has been reported to decrease in obese hearts. Nevertheless, underlying mechanisms linking MD1 and VA have not been fully studied. This study aims to investigate the regulatory role of MD1 in VA caused by diet-induced obesity. METHODS AND RESULTS: MD1 knock-out (KO) and wild type (WT) mice from experimental groups were fed with a high-fat diet (HFD) since the age of six weeks for 20 weeks. The body weight gain, fast glucose and serum lipid levels were measured and recorded. In addition, pathological analysis, echocardiography, electrocardiography, langendorff-perfused heart and molecular analysis were performed to detect HFD-induced vulnerability to VA and its underlying mechanisms. After a 20-week HFD feeding, the mice showed an increase in body weight, glycemic, lipid levels, QTc interval, LVEDd, LVEDs and LVFS. HFD feeding also increased vulnerability to VA, as shown by the prolonged action potential duration (APD), enhanced APD alternans threshold and greater incidence of VA. Moreover, HFD feeding caused LV hypertrophy and fibrosis, and decreased the protein expressions of Kv4.2, Kv4.3, Kv1.5, Kv2.1 and Cav1.2 channels. At last, the above-mentioned HFD-induced adverse effects were further exacerbated in KO mice compared with WT mice. Mechanistically, MD1 deletion markedly enhanced the activation of TLR4/MyD88/CaMKII signaling pathway in HFD-fed mice. CONCLUSION: MD1 deficiency increased HFD-induced vulnerability to VA. This is mainly caused by the aggravated maladaptive LV hypertrophy, fibrosis and decreased protein expressions of ion channels, which are induced by the enhanced activation of the TLR4/MyD88/CaMKII signaling pathway.


Assuntos
Arritmias Cardíacas/enzimologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Frequência Cardíaca , Ventrículos do Coração/enzimologia , Fator 88 de Diferenciação Mieloide/metabolismo , Proteína MyoD/metabolismo , Obesidade/enzimologia , Receptor 4 Toll-Like/metabolismo , Potenciais de Ação , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Dieta Hiperlipídica , Modelos Animais de Doenças , Fibrose , Ventrículos do Coração/fisiopatologia , Hipertrofia Ventricular Esquerda/enzimologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Masculino , Camundongos Knockout , Proteína MyoD/genética , Obesidade/genética , Obesidade/fisiopatologia , Transdução de Sinais , Fatores de Tempo , Função Ventricular Esquerda , Remodelação Ventricular
13.
J Stroke Cerebrovasc Dis ; 28(11): 104143, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31477449

RESUMO

BACKGROUND: The CHADS2 and CHA2DS2-VASc scoring systems have been proved efficacy to stratify stroke and thromboembolism risk in patients with atrial fibrillation (AF). Whether CHADS2 and CHA2DS2-VASc score has predictive value for the prognosis in lacunar stroke (LS) patients remains unclear. METHODS: A total of 763 consecutive patients with LS (mean age: 66 ± 12 years; 464 male) were enrolled in this study between January 2013 and December 2014. Patients were divided into LS without AF (LS; n = 679) and LS with AF (LS-AF; n = 84) groups. Measures of performance for the risk scores were evaluated at predicting mortality and restroke in LS-AF and LS without AF patients. All patients were evaluated with respect to clinical features and in-hospital clinical results. RESULTS: During the mean follow-up period of 20 ± 5.8 months, 29 patients (3.8%) experienced all-cause death, 105 patients (13.8%) experienced recurrence of ischemic stroke. Multivariate analysis revealed that CHADS2 and CHA2DS2-VASc score were independently associated with all-cause death (all P < .05). On receiver operating characteristic curve analysis, area under the curve (AUC) for CHADS2 score was .942 with a similar accuracy of the CHA2DS2-VASc score (AUC: .908) in predicting mortality in LS-AF patients. Kaplan-Meier curves were conducted according to the cut-off value of CHA2DS2-VASc score. When CHADS2 score greater than or equal to 4 point or CHA2DS2-VASc score greater than or equal to 5 point, the mortality in LS-AF patients was significantly higher compared with those CHADS2 score less than 4 point or CHA2DS2-VASc score less than 5 point. However, after adjusting for clinical covariates, CHADS2 and CHA2DS2-VASc score could not predict both mortality and restroke in LS without AF patients. CONCLUSIONS: The CHADS2 and CHA2DS2-VASc score have excellent predictive value for mortality in LS-AF patients but could not predict both mortality and restroke in LS without AF patients.


Assuntos
Fibrilação Atrial/diagnóstico , Técnicas de Apoio para a Decisão , Acidente Vascular Cerebral Lacunar/diagnóstico , Idoso , Idoso de 80 Anos ou mais , Fibrilação Atrial/complicações , Fibrilação Atrial/mortalidade , Causas de Morte , Bases de Dados Factuais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Prognóstico , Recidiva , Reprodutibilidade dos Testes , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Acidente Vascular Cerebral Lacunar/etiologia , Acidente Vascular Cerebral Lacunar/mortalidade , Fatores de Tempo
14.
Biochem Biophys Res Commun ; 505(1): 134-140, 2018 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-30241939

RESUMO

Recent years, the role of long non-coding RNAs (lncRNAs) in atrial fibrillation (AF) has been gradually elucidated. In the current study, we measured the expression of ten AF-related lncRNAs to do qRT-PCR analysis. LncRNA KCNQ1 overlapping transcript 1 (KCNQ1OT1) was found to be significantly upregulated in AF model and Ang-II-induced mice heart. CACNA1C has been reported as a biomarker in atrial fibrillation. Here, we found that the expression pattern of CACNA1C was consistent with that of KCNQ1OT1. Electrophysiological study was conducted to demonstrate the effect of KCNQ1OT1 and CACNA1C on the Effective refractory period (ERP), interatrial conduction time (IACT), incidence of AF and AF duration of Ang-II-induced mice heart. Mechanically, KCNQ1OT1 contributed to the upregulation of CACNA1C by binding with miR-384. Furthermore, YY1 could activate the transcription of KCNQ1OT1 and CACNA1C. In conclusion, the present study revealed that YY1-induced upregulation of lncRNA KCNQ1OT1 regulates angiotensin II-induced atrial fibrillation by regulating miR-384/CACNA1C axis.


Assuntos
Fibrilação Atrial/genética , Canais de Cálcio Tipo L/genética , Regulação da Expressão Gênica , MicroRNAs/genética , RNA Longo não Codificante/genética , Fator de Transcrição YY1/genética , Angiotensina II , Animais , Fibrilação Atrial/induzido quimicamente , Fibrilação Atrial/fisiopatologia , Sequência de Bases , Canais de Cálcio Tipo L/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Interferência de RNA , RNA Longo não Codificante/metabolismo , Homologia de Sequência do Ácido Nucleico , Transdução de Sinais/genética , Regulação para Cima , Fator de Transcrição YY1/metabolismo
15.
Exp Physiol ; 103(9): 1221-1229, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29928790

RESUMO

NEW FINDINGS: What is the central question of this study? Can targeted ablation of cardiac sympathetic neurons suppress myocardial infarction-induced adverse cardiac remodelling and left ventricular dysfunction? What is the main finding and its importance? Targeted ablation of cardiac sympathetic neurons significantly alleviated sympathetic remodelling and neuroendocrine activation, attenuated cardiac hypertrophy and fibrosis and improved left ventricular function. Thus, targeted ablation of cardiac sympathetic neurons might have a beneficial effect on adverse postinfarction remodelling and left ventricular dysfunction. ABSTRACT: Sympathetic overactivation is crucial in the development and progression of adverse cardiac remodelling and dysfunction. Targeted ablation of cardiac sympathetic neurons (TACSN) is an effective approach to inhibit overactivation of the sympathetic nervous system. The aim of this study was to investigate whether TACSN could suppress myocardial infarction (MI)-induced adverse cardiac remodelling and dysfunction, thereby producing protective effects. Thirty-eight dogs were randomly assigned into the sham-operated, MI or MI-TACSN group. The TACSN was induced by injecting cholera toxin B subunit-saporin compound into the stellate ganglia 1 week after MI. Five weeks after MI surgery, echocardiographic and haemodynamic parameters of cardiac function were significantly improved in the TACSN group compared with the MI group. In addition, TACSN attenuated the extent of cardiac hypertrophy and fibrosis and suppressed the increase in the plasma concentrations of noradrenaline, nerve growth factor, atrial natriuretic peptide, brain natriuretic peptide, angiotensin II and aldosterone. Furthermore, TACSN alleviated the growth associated protein-43-positive and tyrosine hydroxylase-positive nerve densities in the infarcted border zone and restored protein expression of the ß1 -adrenergic receptor in the left ventricular myocardium. These findings indicate that TACSN might have a beneficial effect on adverse postinfarction remodelling and left ventricular dysfunction, which might be attributable, at least in part, to the attenuation of both sympathetic remodelling and the cardiac neuroendocrine system.


Assuntos
Técnicas de Ablação/métodos , Coração/inervação , Infarto do Miocárdio/terapia , Neurônios , Sistema Nervoso Simpático , Disfunção Ventricular Esquerda/terapia , Remodelação Ventricular , Animais , Toxina da Cólera/administração & dosagem , Toxina da Cólera/farmacologia , Cães , Ecocardiografia , Expressão Gênica , Injeções , Masculino , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/patologia , Sistemas Neurossecretores , Saporinas/administração & dosagem , Saporinas/farmacologia , Gânglio Estrelado , Disfunção Ventricular Esquerda/patologia
16.
Europace ; 20(12): 2036-2044, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29860489

RESUMO

Aims: The purpose of this study was to evaluate the cardiac electrophysiologic effects of targeted ablation of cardiac sympathetic neurons (TACSN) in a canine model of chronic myocardial infarction (MI). Methods and results: Thirty-eight anaesthetized dogs were randomly assigned into the sham-operated, MI, and MI-TACSN groups, respectively. Myocardial infarction-targeted ablation of cardiac sympathetic neuron was induced by injecting cholera toxin B subunit-saporin compound in the left stellate ganglion (LSG). Five weeks after surgery, the cardiac function, heart rate variability (HRV), ventricular electrophysiological parameters, LSG function and neural activity, serum norepinephrine (NE), nerve growth factor (NGF), and brain natriuretic peptide (BNP) levels were measured. Cardiac sympathetic innervation was determined with immunofluorescence staining of growth associated protein-43 (GAP43) and tyrosine hydroxylase (TH). Compared with MI group, TACSN significantly improved HRV, attenuated LSG function and activity, prolonged corrected QT interval, decreased Tpeak-Tend interval, prolonged ventricular effective refractory period (ERP), and action potential duration (APD), decreased the slopes of APD restitution curves, suppressed the APD alternans, increased ventricular fibrillation threshold, and reduced serum NE, NGF, and BNP levels. Moreover, the densities of GAP43 and TH-positive nerve fibres in the infarcted border zone in the MI-TACSN group were lower than those in the MI group. Conclusion: Targeted ablation of cardiac sympathetic neuron attenuates sympathetic remodelling and improves ventricular electrical remodelling in the chronic phase of MI. These data suggest that TACSN may be a novel approach to treating ventricular arrhythmias.


Assuntos
Potenciais de Ação , Toxina da Cólera , Frequência Cardíaca , Coração/inervação , Infarto do Miocárdio/terapia , Saporinas , Gânglio Estrelado/fisiopatologia , Simpatectomia Química/métodos , Função Ventricular Esquerda , Remodelação Ventricular , Animais , Biomarcadores/sangue , Modelos Animais de Doenças , Cães , Técnicas Eletrofisiológicas Cardíacas , Infarto do Miocárdio/sangue , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Peptídeo Natriurético Encefálico/sangue , Fator de Crescimento Neural/sangue , Norepinefrina/sangue , Recuperação de Função Fisiológica , Fatores de Tempo
17.
Jpn J Clin Oncol ; 46(3): 209-21, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26732382

RESUMO

OBJECTIVE: Today, more and more evidence suggests that Foxk proteins (Foxk1 and Foxk2) work as transcriptional repressors in different kinds of cancer, but whether Foxk1 has a role in mediating tumorigenesis in breast cancer, the evidence is rare. METHODS: MCF-7 cells transfected with shFoxk1 displayed a mesenchymal morphology and reduced the expression of E-cadherin, and increased the expression of N-cadherin. Transwell invasion assay and living imaging assay show that the overexpression of Foxk1 could inhibit metastasis in vitro and in vivo. Ribonucleic acid sequencing revealed that the knockdown of Foxk1 resulted in the up-regulation of different oncogenes, which was implicated in metastasis and tumor angiopoiesis. Quantitative chromatin immunoprecipitation, chromatin immunoprecipitation and Luciferase reporter assays suggested that Foxk1 could bind to the promoter of epithelial-mesenchymal transition inducer Twist and vascular endothelial growth factor, VEGF. Mass Spectrometry, co-immunoprecipitation assays and glutathione-S-transferase pull-down assay detected that Foxk1 was physically associated with Ten-eleven translocation 1, TET1, in vivo and in vitro. RESULTS: We reported that the mean expression level of Foxk1 in breast cancer was significantly lower than the adjacent noncarcinoma tissue. The higher Foxk1 expression was associated with better prognosis. Endothelial tube formation assays indicated that Foxk1 might regulate breast cancer angiogenesis through transcriptional repression of vascular endothelial growth factor. Furthermore, in vivo magnetic resonance imaging revealed the overexpression of Foxk1 could enhance the detection of the tumors. Further, a strong negative correlation was observed between Foxk1 and Twsit or between Foxk1 and vascular endothelial growth factor, and the higher Foxk1 expression is correlated with better over all survivals and better relapse-free survivals. CONCLUSIONS: Together, our data indicated the function of Foxk1 as a tumor suppressor in facilitating angiogenesis and metastasis in breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Imageamento por Ressonância Magnética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Caderinas/metabolismo , Imunoprecipitação da Cromatina , Regulação para Baixo , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Oxigenases de Função Mista , Recidiva Local de Neoplasia , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase em Tempo Real , Proteína 1 Relacionada a Twist/metabolismo , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Mol Cell Endocrinol ; 592: 112315, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38878954

RESUMO

Diabetic cardiomyopathy (DCM) is characterized by oxidative damage and inflammatory responses. Myeloid differentiation protein 1 (MD1) exhibits antioxidant and anti-inflammatory properties. However, the specific role of MD1 in DCM has yet to be elucidated. This study aims to investigate the role of MD1 in DCM and to elucidate the underlying mechanisms. We utilized a gain-of-function approach to explore the involvement of MD1 in DCM. Diabetes was induced in MD1-transgenic (MD1-TG) mice and their wild-type (WT) counterparts via streptozotocin (STZ) injection. Additionally, a diabetes cell model was established using H9c2 cells exposed to high glucose levels. We conducted comprehensive evaluations, including pathological analyses, echocardiography, electrocardiography, and molecular assessments, to elucidate the underlying mechanisms of MD1 in DCM. Notably, MD1 expression was reduced in the hearts of STZ-induced diabetic mice. Overexpression of MD1 significantly improved cardiac function and markedly inhibited ventricular pathological hypertrophy and fibrosis in these mice. Furthermore, MD1 overexpression resulted in a substantial decrease in myocardial reactive oxygen species (ROS) accumulation, mitigating myocardial oxidative stress and reducing the levels of inflammation-related markers such as IL-1ß, IL-6, and TNF-α. Mechanistically, MD1 overexpression inhibited the activation of the TLR4/STAT3 signaling pathway, as demonstrated in both in vivo and in vitro experiments. The overexpression of MD1 significantly impeded pathological cardiac remodeling and improved cardiac function in STZ-induced diabetic mice. This effect was primarily attributed to a reduction in ROS accumulation and mitigation of myocardial oxidative stress and inflammation, facilitated by the inhibition of the TLR4/STAT3 signaling pathway.

20.
Genes Genomics ; 46(1): 149-160, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37523128

RESUMO

BACKGROUND: Bupivacaine, a common local anesthetic, can cause neurotoxicity and permanent neurological disorders. Crocin has been widely reported as a potential neuroprotective agent in neural injury models. OBJECTIVE: The aim of this study was to investigate the role and regulatory mechanism of crocin underlying bupivacaine-induced neurotoxicity. METHOD: Human neuroblastoma SH-SY5Y cells were treated with bupivacaine and/or crocin for 24 h, followed by detecting cell viability using 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay. The effect of crocin or bupivacaine on SH-SY5Y cell proliferation was measured by Ki67 immunofluorescence assay. The levels of apoptosis-related proteins and the markers in the PI3K/Akt signaling pathway were examined using western blot analysis. The activities of caspase 3, catalase (CAT), superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) were tested using respective commercial assay kits. Flow cytometry analysis was executed for detecting SH-SY5Y cell apoptosis. RESULT: Crocin attenuated bupivacaine-induced neurotoxicity in SH-SY5Y cells. Meanwhile, crocin inhibited SH-SY5Y cell apoptosis induced by bupivacaine via repressing the activity of caspase-3, reducing Bax expression, and elevating Bcl-2 expression. Moreover, crocin mitigated oxidative stress in SH-SY5Y cells by increasing the content of CAT, SOD, GSH-Px and reducing the content of MDA. Additionally, crocin protected against bupivacaine-induced dephosphorylation of Akt and GSK-3ß. The protective effects of crocin against bupivacaine-induced neurotoxicity in SH-SY5Y cells were counteracted by the Akt inhibitor. CONCLUSION: These results suggested that crocin may exert a neuroprotective function by promoting cell proliferation and suppressing apoptosis and oxidative stress in SH-SY5Y cells. Thus, crocin might become a promising drug for the treatment of bupivacaine-induced neurotoxicity.


Assuntos
Carotenoides , Neuroblastoma , Proteínas Proto-Oncogênicas c-akt , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Bupivacaína/toxicidade , Glicogênio Sintase Quinase 3 beta/metabolismo , Glicogênio Sintase Quinase 3 beta/farmacologia , Linhagem Celular Tumoral , Transdução de Sinais , Superóxido Dismutase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA