Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
J Biochem Mol Toxicol ; 38(8): e23780, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39056188

RESUMO

Sodium and potassium channels, especially Nav1.5 and Kir2.1, play key roles in the formation of action potentials in cardiomyocytes. These channels interact with, and are regulated by, synapse-associated protein 97 (SAP97). However, the regulatory role of SAP97 in myocyte remains incompletely understood. Here, we investigate the function of SAP97 phosphorylation in the regulation of Nav1.5 and Kir2.1 channel complexes and the upstream regulation of SAP97. We found that SAP97 is phosphorylated by casein kinase II (CK2) in vitro. In addition, transfection of casein kinase 2 interacting protein-1 (CKIP-1) into cardiomyocytes to drive CK2 from the nucleus to the cytoplasm, increased SAP97 phosphorylation and Nav1.5 and Kir2.1 current activity. These findings demonstrated that CKIP-1 modulates the subcellular translocation of CK2, which regulates Nav1.5 and Kir2.1 channel complex formation and activity in cardiomyocytes.


Assuntos
Caseína Quinase II , Miócitos Cardíacos , Canal de Sódio Disparado por Voltagem NAV1.5 , Canais de Potássio Corretores do Fluxo de Internalização , Miócitos Cardíacos/metabolismo , Caseína Quinase II/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Animais , Ratos , Fosforilação , Transporte Proteico , Humanos , Proteínas de Transporte/metabolismo , Ratos Sprague-Dawley
2.
Front Psychiatry ; 15: 1392605, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39050916

RESUMO

Background: Atrial fibrillation (AF) is one of the most common form of arrhythmia. Previous studies have shown a link between AF and mental illness. However, the causal relationship between mental illness and AF remains unclear. The purpose of this study was to investigate the bidirectional causal relationship between borderline personality disorder (BPD) and AF. Method: We used the bidirectional Two-sample Mendelian randomization (TSMR) method to evaluate the causal relationship between BPD and AF. Instrumental variables associated with BPD were derived from a genome-wide association study involving 214,816 Europeans (2,637 cases and 212,179 controls). We then obtained atrial fibrillation data from the GWAS meta-analysis (60,620 cases and 970,216 controls). The TSMR analyses were performed in five methods, namely fixed-effect inverse-variance weighted (IVW) method、random-effect IVW method, MR Egger regression method, Weighted median method and Simple mode method. Several sensitivity analyses are used to test the robustness of positive results. Results: The fixed-effect inverse-variance weighted model [Odds ratio (OR), 1.033, 95% confidence interval (CI), 1.011-1.056, P = 0.0031], random-effect inverse-variance weighted model (OR, 1.033; 95%CI, 1.005-1.062; P = 0.0191) and Weighted median (OR, 1.034; 95%CI, 1.002-1.068; P = 0.0394) all showed that genetically predicted BPD was associated with an increased risk of AF. Sensitivity analysis using other MR Methods, including the MR-Egger intercept, MR-Presso method, and leave-one-out analyses, showed that the results were robust. In reverse MR analysis, there was no causal relationship of AF on BPD. Conclusion: Our study provides a causal relationship between BPD and AF. This means that patients with BPD should be monitored for the occurrence of AF. Early screening and proper management of BPD may show anti-arrhythmic benefits.

3.
J Pharmacol Exp Ther ; 390(2): 240-249, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-38902033

RESUMO

Sympathetic hyperinnervation is the leading cause of fatal ventricular arrhythmia (VA) after myocardial infarction (MI). Cardiac mast cells cause arrhythmias directly through degranulation. However, the role and mechanism of mast cell degranulation in sympathetic remodeling remain unknown. We investigated the role of oxytocin (OT) in stabilizing cardiac mast cells and improving sympathetic innervation in rats. MI was induced by coronary artery ligation. Western blotting, immunofluorescence, and toluidine staining of mast cells were performed to determine the expression and location of target protein. Mast cells accumulated significantly in peri-infarcted tissues and were present in a degranulated state. They expressed OT receptor (OTR), and OT infusion reduced the number of degranulated cardiac mast cells post-MI. Sympathetic hyperinnervation was attenuated as assessed by immunofluorescence for tyrosine hydroxylase (TH). Seven days post-MI, the arrhythmia score of programmed electrical stimulation was higher in vehicle-treated rats with MI than in rats treated with OT. An in vitro study showed that OT stabilized mast cells via the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway. Further in vivo studies on OTR-deficient mice showed worsening mast cell degranulation and worsening sympathetic innervation. OT pretreatment inhibited cardiac mast cell degranulation post-MI and prevented sympathetic hyperinnervation, along with mast cell stabilization via the PI3K/Akt pathway. SIGNIFICANCE STATEMENT: This is the first study to elucidate the role and mechanism of oxytocin (OT) in inflammatory-sympathetic communication mediated sympathetic hyperinnervation after myocardial infarction (MI), providing new approaches to prevent fatal arrhythmias.


Assuntos
Degranulação Celular , Mastócitos , Infarto do Miocárdio , Ocitocina , Ratos Sprague-Dawley , Receptores de Ocitocina , Sistema Nervoso Simpático , Animais , Ocitocina/farmacologia , Ocitocina/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Ratos , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/metabolismo , Masculino , Degranulação Celular/efeitos dos fármacos , Receptores de Ocitocina/metabolismo , Receptores de Ocitocina/antagonistas & inibidores , Camundongos , Transdução de Sinais/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/etiologia
4.
Tissue Cell ; 88: 102371, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38593570

RESUMO

BACKGROUND: Paeonol is a representative active ingredient of the traditional Chinese medicinal herbs Cortex Moutan, which has a well-established cardioprotective effect on ischemic heart disease. However, there is little evidence of the protective effect of paeonol, and its pharmacological mechanism is also unclear. This study aims to explore the protective effect and mechanism of Paeonol on myocardial infarction rat and hypoxic H9c2 cells. METHODS: Myocardial ischemia/reperfusion (I/R) was induced by occlusion of the left anterior descending coronary artery for 1 h followed by 3 h of reperfusion, and then gavage with Paeonol for 7 days. H9c2 cells were applied for the in vitro experiments and hypoxia/reoxygenation (H/R) model was established. CKIP-1 expression was evaluated by qPCR and western blot. The expression of genes involved in apoptosis, inflammation and ion channel was measured by western blot. The currents levels of Nav1.5 and Kir2.1 were measured by whole-cell patch-clamp recording. RESULTS: CKIP-1 expression was decreased in H/R-induced H9c2 cells, which was inversely increased after Paeonol treatment. Paeonol treatment could increase the viability of H/R-induced H9c2 cells and diminish the apoptosis and inflammation of H/R-induced H9c2 cells, while si-CKIP-1 treatment inhibited the phenomena. Moreover, the currents levels of Nav1.5 and Kir2.1 were reduced in H/R-induced H9c2 cells, which were inhibited after Paeonol treatment. Intragastric Paeonol can reduce the ventricular arrhythmias in rats with myocardial infarction. CONCLUSIONS: The protective effects of Paeonol on myocardial infarction rats and hypoxic H9c2 cells were achieved by up-regulating CKIP-1.


Assuntos
Acetofenonas , Hipóxia Celular , Regulação para Cima , Acetofenonas/farmacologia , Animais , Ratos , Regulação para Cima/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Canais Iônicos/metabolismo , Canais Iônicos/genética , Apoptose/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Masculino , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/patologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Ratos Sprague-Dawley
5.
Free Radic Biol Med ; 209(Pt 2): 342-354, 2023 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-37898386

RESUMO

OBJECTIVES: N6-methyladenosine (m6A) is the most prevalent post-translational modification in eukaryotic mRNA. Recently, m6A editing modified by methyltransferase-like enzyme 3 (METTL3), the core m6A methyltransferase, has been demonstrated to be involved in cardiac sympathetic hyperactivity. This study aimed to clarify the effects and underlying mechanisms of METTL3 in the paraventricular nucleus (PVN) in mediating sympathetic activity following myocardial infarction (MI). METHODS: We established rat MI models by left anterior descending coronary artery ligation. m6A quantification was performed.The expression of METTL3 and its downstream gene, tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6), were determined. The functional role of METTL3 in sympathetic hyperactivity and electrical conduction stability were verified by assessing renal sympathetic nerve activity (RSNA), norepinephrine (NE) levels, and programmed electrical stimulation. Rescue experiments were also conducted. The mechanism by which m6A is involved in mitochondrial reactive oxygen species (mROS) production, mediated by TRAF6/ECSIT pathway, was explored in lipopolysaccharide (LPS) treated primary microglial cells. RESULTS: METTL3 was predominantly localized in the microglia and significantly increased within the PVN at 3 days post-MI. Inhibition of METTL3 decreased m6A levels, TRAF6 expression, and mROS production; downregulated sympathoexcitation, indicated by attenuated NE concentration and RSNA; decreased the incidence of ventricular tachycardia or fibrillation; and improved cardiac function. Mechanistically, downregulation of METTL3 prevented TRAF6 translocation to the mitochondria in the microglia and subsequent TRAF6/ECSIT pathway activation, resulting in decreased mROS production. CONCLUSIONS: This study demonstrates that METTL3-mediated m6A modification promotes sympathetic hyperactivity through TRAF6/ECSIT pathway and mitochondrial oxidative stress in the PVN, thereby leading to ventricular arrhythmias post-MI.


Assuntos
Infarto do Miocárdio , Fator 6 Associado a Receptor de TNF , Animais , Ratos , Metiltransferases/genética , Metiltransferases/metabolismo , Mitocôndrias/metabolismo , Infarto do Miocárdio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo
6.
Am Heart J ; 260: 34-43, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36813122

RESUMO

BACKGROUND: In randomized studies, the strategy of pulmonary vein antral isolation (PVI) plus linear ablation has failed to increase success rates for persistent atrial fibrillation (PeAF) ablation when compared with PVI alone. Peri-mitral reentry related atrial tachycardia due to incomplete linear block is an important cause of clinical failures of a first ablation procedure. Ethanol infusion (EI) into the vein of Marshall (EI-VOM) has been demonstrated to facilitate a durable mitral isthmus linear lesion. OBJECTIVE: This trial is designed to compare arrhythmia-free survival between PVI and an ablation strategy termed upgraded '2C3L' for the ablation of PeAF. STUDY DESIGN: The PROMPT-AF study (clinicaltrials.gov 04497376) is a prospective, multicenter, open-label, randomized trial using a 1:1 parallel-control approach. Patients (n = 498) undergoing their first catheter ablation of PeAF will be randomized to either the upgraded '2C3L' arm or PVI arm in a 1:1 fashion. The upgraded '2C3L' technique is a fixed ablation approach consisting of EI-VOM, bilateral circumferential PVI, and 3 linear ablation lesion sets across the mitral isthmus, left atrial roof, and cavotricuspid isthmus. The follow-up duration is 12 months. The primary end point is freedom from atrial arrhythmias of >30 seconds, without antiarrhythmic drugs, in 12 months after the index ablation procedure (excluding a blanking period of 3 months). CONCLUSIONS: The PROMPT-AF study will evaluate the efficacy of the fixed '2C3L' approach in conjunction with EI-VOM, compared with PVI alone, in patients with PeAF undergoing de novo ablation.


Assuntos
Fibrilação Atrial , Ablação por Cateter , Veias Pulmonares , Humanos , Fibrilação Atrial/cirurgia , Veias Pulmonares/cirurgia , Estudos Prospectivos , Átrios do Coração/cirurgia , Etanol , Ablação por Cateter/métodos , Resultado do Tratamento , Recidiva
7.
J Mol Cell Cardiol ; 170: 87-99, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35717715

RESUMO

OBJECTIVE: Sudden cardiac death caused by ventricular arrhythmias (VAs) is the main cause of high mortality in patients with myocardial infarction (MI). Sympathetic neural remodeling caused by inflammation after MI is closely associated with the occurrence of VAs. METTL3, the earliest identified m6A methyltransferase, is critical in mediating inflammatory responses. Our aim was to investigate whether the m6A methyltransferase METTL3 was involved in sympathetic remodeling post-MI and its specific mechanism. METHODS AND RESULTS: A rat MI model was established via left coronary artery ligation. The expression of METTL3, TRAF6, NOX2, and NF-κB increased at 3 days and remained elevated at 7 days after MI, as determined via Western blotting. METTL3 was primarily present in macrophages, as determined via immunofluorescence. Intramyocardial injection of lentivirus carrying METTL3-shRNA inhibited METTL3 expression in vivo. Methylated immunoprecipitation-qPCR determined the METTL3 knockdown inhibited the m6A level of TRAF6 mRNA 3'-UTR. The co-immunoprecipitation experiment proved that METTL3 combines with TRAF6. Western blotting showed that silencing METTL3 inhibited TRAF6 level, NF-κB activation, and ROS production; decreased cytokine release (TNF-α and IL-1ß); and downregulated nerve growth factor expression. Finally, METTL3 knockdown reduced sympathetic remodeling after MI, as determined via immunofluorescence assays of tyrosine hydroxylase and growth-associated protein 43. Programmed electrical stimulation, renal sympathetic nerve activity recording, and haemodynamic measurements showed that METTL3 inhibition decreased sympathetic activity and improved cardiac function. CONCLUSIONS: Downregulation of METTL3 expression attenuated the excessive sympathetic neural remodeling induced by MI, further reducing the incidence of VAs and improving cardiac function. This was partly associated with the inhibition of the TRAF6/NF-κB pathway and ROS production.


Assuntos
Infarto do Miocárdio , NF-kappa B , Animais , Ratos , Arritmias Cardíacas , Metiltransferases/metabolismo , Infarto do Miocárdio/metabolismo , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Remodelação Ventricular
8.
J Int Med Res ; 50(4): 3000605211070755, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35441549

RESUMO

A man in his early 40s developed palpitations brought on by swallowing and was found to have short runs of atrial tachycardia induced by swallowing solid food. Atrial tachycardia during swallowing was documented on electrocardiography and 24-hour Holter monitoring. No structural heart disease or esophageal disorders were found by echocardiography. The patient then underwent an electrophysiological study and catheter ablation. We mapped the left atrium with a multipolar mapping catheter while the patient swallowed bread and found that the earliest endocardial breakthrough was on the left anterior superior atrium, where the left superior ganglionated plexus was located. We successfully eliminated the paroxysmal atrial tachycardia at this site. Interestingly, in the process of ablation, atrioventricular node reentrant tachycardia was triggered. After the slow-pathway ablation procedure, no further tachycardia was induced.


Assuntos
Ablação por Cateter , Taquicardia por Reentrada no Nó Atrioventricular , Ablação por Cateter/métodos , Deglutição , Eletrocardiografia , Átrios do Coração/diagnóstico por imagem , Átrios do Coração/cirurgia , Humanos , Masculino , Taquicardia , Taquicardia por Reentrada no Nó Atrioventricular/cirurgia
9.
J Cell Mol Med ; 26(10): 2959-2971, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35393774

RESUMO

Sympathetic activation after myocardial infarction (MI) leads to ventricular arrhythmias (VAs), which can result in sudden cardiac death (SCD). The toll-like receptor 4 (TLR4)/myeloid differentiation primary response 88 (MyD88)/nuclear factor-kappa B (NF-kB) axis within the hypothalamic paraventricular nucleus (PVN), a cardiac-neural sympathetic nerve centre, plays an important role in causing VAs. An MI rat model and a PVN-TLR4 knockdown model were constructed. The levels of protein were detected by Western blotting and immunofluorescence, and localizations were visualized by multiple immunofluorescence staining. Central and peripheral sympathetic activation was visualized by immunohistochemistry for c-fos protein, renal sympathetic nerve activity (RSNA) measurement, heart rate variability (HRV) analysis and norepinephrine (NE) level detection in serum and myocardial tissue measured by ELISA. The arrhythmia scores were measured by programmed electrical stimulation (PES), and cardiac function was detected by the pressure-volume loop (P-V loop). The levels of TLR4 and MyD88 and the nuclear translocation of NF-kB within the PVN were increased after MI, while sympathetic activation and arrhythmia scores were increased and cardiac function was decreased. However, inhibition of TLR4 significantly reversed these conditions. PVN-mediated sympathetic activation via the TLR4/MyD88/NF-kB axis ultimately leads to the development of VAs after MI.


Assuntos
Infarto do Miocárdio , Núcleo Hipotalâmico Paraventricular , Animais , Arritmias Cardíacas/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Infarto do Miocárdio/metabolismo , NF-kappa B/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
10.
J Cell Mol Med ; 26(4): 1264-1280, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35040253

RESUMO

Ventricular arrhythmias (VAs) triggers by sympathetic nerve hyperactivity contribute to sudden cardiac death in myocardial infarction (MI) patients. Microglia-mediated inflammation in the paraventricular nucleus (PVN) is involved in sympathetic hyperactivity after MI. N6-methyladenosine (m6 A), the most prevalent mRNA and epigenetic modification, is critical for mediating cell inflammation. We aimed to explore whether METTL3-mediated m6 A modification is involved in microglia-mediated sympathetic hyperactivity after MI in the PVN. MI model was established by left coronary artery ligation. METTL3-mediated m6 A modification was markedly increased in the PVN at 3 days after MI, and METTL3 was primarily located in microglia by immunofluorescence. RNA-seq, MeRIP-seq, MeRIP-qPCR, immunohistochemistry, ELISA, heart rate variability measurements, renal sympathetic nerve activity recording and programmed electrical stimulation confirmed that the elevated toll-like receptor 4 (TLR4) expression by m6 A modification on TLR4 mRNA 3'-UTR region combined with activated NF-κB signalling led to the overwhelming production of pro-inflammatory cytokines IL-1ß and TNF-α in the PVN, thus inducing the sympathetic hyperactivity and increasing the incidence of VAs post-MI. Targeting METTL3 attenuated the inflammatory response and sympathetic hyperactivity and reduced the incidence of VAs post-MI.


Assuntos
Metiltransferases , Infarto do Miocárdio , Animais , Coração , Humanos , Metilação , Metiltransferases/metabolismo , Infarto do Miocárdio/complicações , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Sistema Nervoso Simpático/metabolismo
11.
J Cardiol ; 79(3): 423-431, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34750029

RESUMO

BACKGROUND: Cardiac sympathetic hyperinnervation after myocardial infarction (MI) is associated with a high incidence of lethal arrhythmia. Erythropoietin-producing hepatoma interactor B2 (EphrinB2), a diffusible axonal chemorepellent that can induce growth cone collapse and axon repulsion of several neuronal populations, is crucial in neurodevelopment during disease development and progression. However, whether EphrinB2 could inhibit cardiac sympathetic hyperinnervation after MI remains unclear. METHODS AND RESULTS: A rat model of MI was developed by left anterior descending coronary artery ligation. EphrinB2 expression was markedly increased in the infarcted border at 3 days after MI. Downregulation of EphrinB2 by intramyocardial injection of lentivirus carrying EphrinB2-shRNA significantly increased sympathetic hyperinnervation along with downregulated RhoA expression. In contrast, injection of EphrinB2-overexpressing lentivirus markedly upregulated EphrinB2, concomitant with inhibition of sympathetic sprouting and upregulated RhoA expression, accompanied by decreased incidence of ventricular arrhythmias (VAs). However, co-administering EphrinB2-overexpressing lentivirus and Fasudil (Rho kinase inhibitor) nearly abolished the inhibition of nerve sprouting effect. Additionally, EphrinB2 expression did not affect nerve growth factor level in the infarcted heart. CONCLUSIONS: Overexpression of EphrinB2 may ameliorate MI-induced sympathetic hyperinnervation and further reduce the incidence of VAs, at least in part by activating RhoA-mediated axonal retraction.


Assuntos
Arritmias Cardíacas , Efrina-B2/metabolismo , Infarto do Miocárdio , Animais , Arritmias Cardíacas/complicações , Arritmias Cardíacas/prevenção & controle , Coração , Incidência , Infarto do Miocárdio/complicações , Miocárdio/metabolismo , Ratos , Sistema Nervoso Simpático , Regulação para Cima , Proteínas rho de Ligação ao GTP
12.
Front Cardiovasc Med ; 9: 1019435, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36684596

RESUMO

Objective: Sympathetic remodeling after myocardial infarction (MI) is the primary cause of ventricular arrhythmias (VAs), leading to sudden cardiac death (SCD). M1-type macrophages are closely associated with inflammation and sympathetic remodeling after MI. Long noncoding RNAs (lncRNAs) are critical for the regulation of cardiovascular disease development. Therefore, this study aimed to identify the lncRNAs involved in MI and reveal a possible regulatory mechanism. Methods and results: M0- and M1-type macrophages were selected for sequencing and screened for differentially expressed lncRNAs. The data revealed that lncRNA LOC100911717 was upregulated in M1-type macrophages but not in M0-type macrophages. In addition, the lncRNA LOC100911717 was upregulated in heart tissues after MI. Furthermore, an RNA pull-down assay revealed that lncRNA LOC100911717 could interact with growth-associated protein 43 (GAP43). Essentially, immunofluorescence assays and programmed electrical stimulation demonstrated that GAP43 expression was suppressed and VA incidence was reduced after lncRNA LOC100911717 knockdown in rat hearts using an adeno-associated virus. Conclusions: We observed a novel relationship between lncRNA LOC100911717 and GAP43. After MI, lncRNA LOC100911717 was upregulated and GAP43 expression was enhanced, thus increasing the extent of sympathetic remodeling and the frequency of VA events. Consequently, silencing lncRNA LOC100911717 could reduce sympathetic remodeling and VAs.

13.
Arch Pharm Res ; 44(5): 525-535, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33942232

RESUMO

Myocardial infarction (MI) is one of the leading causes of high mortality worldwide. Long non-coding RNA myocardial infarction associated transcript (MIAT) and mitochondrial coupling factor 6 (CF6) aggravate MI. This study aimed to elucidate whether miR-203 interacted with MIAT and CF6 in MI. Results revealed that MIAT and CF6 expressions were upregulated and that miR-203 was downregulated in mouse myocardial tissues after MI, as well as in hypoxic mouse cardiomyocytes. The overexpression of MIAT in mouse cardiomyocytes raised CF6 expression, whereas the knockdown of MIAT had the opposite effect. Mechanistically, the luciferase reporter and RNA pull-down assays corroborated the binding between miR-203 and CF6 3'UTR and between miR-203 and MIAT. The simultaneous overexpression of miR-203 and MIAT restored the reduction of CF6 caused by miR-203 overexpression alone, and the overexpression of miR-203 diminished the percentage of infarct area and the apoptosis of cardiomyocytes in vivo. Our findings corroborate that overexpressing miR-203 alleviates MI via interacting with MIAT and CF6.


Assuntos
MicroRNAs/genética , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Infarto do Miocárdio/genética , Fatores Acopladores da Fosforilação Oxidativa/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , ATPases Mitocondriais Próton-Translocadoras/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Fatores Acopladores da Fosforilação Oxidativa/genética , RNA Longo não Codificante/genética
14.
Lipids Health Dis ; 18(1): 150, 2019 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-31286992

RESUMO

BACKGROUND: Elevated lipoprotein (a) is recognized as a risk factor for incident cardiovascular events in the general population and established cardiovascular disease patients. However, there are conflicting findings on the prognostic utility of elevated lipoprotein (a) level in patients with coronary artery disease (CAD).Thus, we performed a meta-analysis to evaluate the prognostic value of elevated lipoprotein (a) level in CAD patients. METHODS AND RESULTS: A systematic literature search of PubMed and Embase databases was conducted until April 16, 2019. Observational studies reporting the prognostic value of elevated lipoprotein (a) level for cardiac events (cardiac death and acute coronary syndrome), cardiovascular events (death, stroke, acute coronary syndrome or coronary revascularisation), cardiovascular death, and all-cause mortality in CAD patients were included. Pooled multivariable adjusted risk ratio (RR) and 95% confidence interval (CI) for the highest vs. the lowest lipoprotein (a) level were utilized to calculate the prognostic value. Seventeen studies enrolling 283,328 patients were identified. Meta-analysis indicated that elevated lipoprotein (a) level was independently associated with an increased risk of cardiac events (RR 1.78; 95% CI 1.31-2.42) and cardiovascular events (RR 1.29; 95% CI 1.17-1.42) in CAD patients. However, elevated lipoprotein (a) level was not significantly associated with an increased risk of cardiovascular mortality (RR 1.43; 95% CI 0.94-2.18) and all-cause mortality (RR 1.35; 95% CI 0.93-1.95). CONCLUSIONS: Elevated lipoprotein (a) level is an independent predictor of cardiac and cardiovascular events in CAD patients. Measurement of lipoprotein (a) level has potential to improve the risk stratification among patients with CAD.


Assuntos
Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/mortalidade , Lipoproteína(a)/sangue , Síndrome Coronariana Aguda/sangue , Síndrome Coronariana Aguda/etiologia , Doença da Artéria Coronariana/complicações , Humanos , Prognóstico , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/etiologia
15.
Neurosci Lett ; 707: 134319, 2019 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-31175933

RESUMO

BACKGROUND AND OBJECTIVES: Nuclear factor-κB (NF-κB) is considered to be a master inflammation regulator and involved in sympathetic neural hyperinnervation after myocardial infarction (MI). Paraventricular nucleus (PVN), acts as the sympathetic outflow tract, has been proven to play an important role during MI, but whether NF-κB pathway in the PVN participates in the pathogenesis of sympathetic sprouting and reinnervation after MI are unclear. METHODS AND RESULTS: MI was induced by coronary artery ligation, NF-κB was activated and interleukin-1ß (IL-1ß) and nerve growth factor (NGF) levels were increased in the PVN after MI. Lipopolysaccharide (LPS) stimulation also can activate NF-κB pathway, followed by increasing the expressions of IL-1ß and NGF in the PVN, once combining with gevokizumab (an IL-1ß inhibitor) significantly reduced the expressions of IL-1ß and NGF, but could not affect the NF-κB expression in the PVN. Furthermore, micro-injection of the NF-κB inhibitor pyrrolidine dithiocarbamate into PVN in MI rats, significantly inhibited the activation of NF-κB and further reduced the expression of IL-1ß and NGF in the PVN, finally blunting sympathetic hyperinnervation in the heart, moreover, the blockade of NF-κB in the PVN reduced the incidence of ventricular arrhythmias (VAs). CONCLUSIONS: Cardiac nerve sprouting after MI is associated in part with NF-κB activation in the PVN, and IL-1ß is involved in the process. Targeting blockade of NF-κB in the PVN may be a potential approach to ameliorate sympathetic hyperinnervation and reduce the incidence of VAs after MI.


Assuntos
Coração/inervação , Infarto do Miocárdio/fisiopatologia , NF-kappa B/antagonistas & inibidores , Núcleo Hipotalâmico Paraventricular/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Animais , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/fisiopatologia , Arritmias Cardíacas/prevenção & controle , Coração/fisiopatologia , Interleucina-1beta/metabolismo , Masculino , Infarto do Miocárdio/complicações , Infarto do Miocárdio/metabolismo , NF-kappa B/metabolismo , Ratos Sprague-Dawley
16.
Acta Physiol (Oxf) ; 227(2): e13315, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31116911

RESUMO

AIM: Overactivation of the sympathetic nerve may lead to severe ventricular arrhythmias (VAs) after myocardial infarction (MI). Thus, targeting sympathetic nerve activity is an effective strategy to prevent VAs clinically. The superior cervical ganglion (SCG), the extracardiac sympathetic ganglion innervating cardiac muscles, has been found to have a GABAergic signalling system, the physiological significance of which is obscure. We aimed to explore the functional significance of SCG post MI and whether the GABAergic signal system is involved in the process. METHODS: Adult male Sprague-Dawley rats were divided into seven different groups. Rats in the MI groups underwent ligation of the left anterior descending coronary artery. All animals were used for electrophysiological testing, renal sympathetic nerve activity (RSNA) testing, and ELISA. Primary SCG sympathetic neurons were used for the in vitro study. RESULTS: The GABAA receptor agonist muscimol significantly decreased the ATP-induced increase in intracellular Ca2+ (P < 0.05). GABA treatment in MI rats significantly attenuated the level of serum and cardiac norepinephrine (NE; P < 0.05). Sympathetic activity and inducible VAs were also lower in MI + GABA rats than in MI rats (P < 0.05). Knockdown of the GABAA Rs ß2 subunit (GABAA Rß2 ) in the SCG of MI rats increased the NE levels in serum and cardiac tissue, RSNA and inducible VAs compared with vehicle shRNA (P < 0.05). CONCLUSION: The GABAergic signalling system is functionally expressed in SCG sympathetic neurons, and activation of this system suppresses sympathetic activity, thereby facilitating cardiac protection and making it a potential target to alleviate VAs.


Assuntos
Arritmias Cardíacas/prevenção & controle , Neurônios GABAérgicos/fisiologia , Infarto do Miocárdio/complicações , Norepinefrina/metabolismo , Animais , Cálcio/metabolismo , Técnicas de Silenciamento de Genes , Masculino , Infarto do Miocárdio/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Transdução de Sinais
17.
Redox Biol ; 24: 101186, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30978539

RESUMO

Sympathetic nerve hyperactivity is a primary reason for fatal ventricular arrhythmias (VAs) following myocardial infarction (MI). Pro-inflammatory cytokines produced in the paraventricular nucleus (PVN) post-MI are associated with sympathetic overexcitation; however, the precise mechanism needs further investigation. Our aim was to explore the mechanism of toll-like receptor 4 (TLR4) and its downstream molecular pathway in mediating sympathetic activity post-MI within the PVN. A rat MI model was developed via left anterior descending coronary artery ligation. TLR4 was primarily localized in microglia and increased markedly within the PVN at 3 days in MI rats. Sympathoexcitation also increased, as indicated by high levels of renal sympathetic nerve activity (RSNA) and norepinephrine (NE) concentration. TLR4 knockdown via shRNA microinjection to the PVN resulted in decreased activation of Fos protein (+) neurons in the PVN and peripheral sympathetic nerve activity. TLR4 knockdown also exhibited a lower arrhythmia score following programmed electrical stimulation than those treated with MI surgery only, indicating that the knockdown of TLR4 decreased the incidence of malignant ventricular arrhythmias following MI. LPS-induced inflammatory response was analyzed to explore the underlying mechanism of TLR4 in sympathetic hyperactivity. High levels of NF-κB protein, the pro-inflammatory cytokines IL-1ß and TNF-α, and ROS production were observed in the LPS group. PVN-targeted injection of the NF-κB inhibitor PDTC attenuated NF-κB expression and sympathetic activity. Taken together, the results suggested that knockdown of microglial TLR4 within the PVN decreased sympathetic hyperactivity and subsequent VAs post-MI. The downstream NF-κB pathway and ROS production participated in the process. Interventions targeting TLR4 signaling in the PVN may be a novel approach to ameliorate the incidence of VAs post-MI.


Assuntos
NF-kappa B/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/fisiopatologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Sistema Nervoso Simpático/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Receptor 4 Toll-Like/metabolismo , Animais , Biomarcadores , Eletrocardiografia , Imunofluorescência , Imuno-Histoquímica , Masculino , Infarto do Miocárdio , Ratos
18.
J Cardiol ; 73(1): 81-88, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30487059

RESUMO

BACKGROUND: The hypothalamic paraventricular nucleus (PVN) is the center of the regulation of autonomic nervous system functions and cardiovascular activity. Phosphoinositide-3 kinase (PI3K)-AKT pathway in PVN contributes to mediate sympathetic nerve activity and is activated in spontaneously hypertensive rats. Overactivation of the sympathetic output was considered as an important mechanism of the arrhythmias. In the present study, we aimed to explore whether targeted regulation of sympathetic activity in PVN could reduce the peripheral sympathoexcitatory and attenuate the ventricular arrhythmias (VAs) in myocardial infarction (MI) rats via PI3K-AKT pathway. METHODS: A stainless steel gauge guide cannula was stereotaxically implanted into the PVN, and 7 days later, rats were randomly divided into the following 4 groups: group A, control+dimethyl sulfoxide (DMSO); group B, control+LY294002; group C, MI surgery+DMSO; and group D, MI surgery+LY294002. Studies were conducted seven days post-MI. Myocardial function, infarct size, inducible VAs by programmed electrical stimulation, renal sympathetic nerve activity (RSNA), and protein level of PI3K and AKT were measured. RESULTS: MI increased the protein ratios of p-PI3K-to-total-PI3K and p-AKT-to-total-AKT in PVN but can be reduced by LY294002 treatment. Inhibition of sympathetic nerve activity in PVN led to a reversion in plasma norepinephrine, RSNA and inducible VAs in MI rats. CONCLUSIONS: PI3K-AKT pathway in the PVN was a main mechanism in regulating sympathetic activity and arrhythmias in MI rats. Targeted inhibition of sympathetic activity in PVN may be a potential treatment for the VAs via PI3K-AKT pathway.


Assuntos
Arritmias Cardíacas/enzimologia , Infarto do Miocárdio/enzimologia , Núcleo Hipotalâmico Paraventricular/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Sistema Nervoso Simpático/enzimologia , Animais , Arritmias Cardíacas/fisiopatologia , Arritmias Cardíacas/prevenção & controle , Cromonas/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Masculino , Morfolinas/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia , Miocárdio/enzimologia , Norepinefrina/sangue , Ratos , Ratos Endogâmicos SHR , Transdução de Sinais/efeitos dos fármacos
19.
J Cell Mol Med ; 23(1): 112-125, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30353660

RESUMO

Malignant ventricular arrhythmias (VAs) following myocardial infarction (MI) is a lethal complication resulting from sympathetic nerve hyperactivity. Numerous evidence have shown that inflammation within the paraventricular nucleus (PVN) participates in sympathetic hyperactivity. Our aim was to explore the role of Macrophage-inducible C-type lectin (Mincle) within the PVN in augmenting sympathetic activity following MI,and whether NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome/IL-1ß axis is involved in this activity. MI was induced by coronary artery ligation. Mincle expression localized in microglia within the PVN was markedly increased at 24 hours post-MI together with sympathetic hyperactivity, as indicated by measurement of the renal sympathetic nerve activity (RSNA) and norepinephrine (NE) concentration. Mincle-specific siRNA was administrated locally to the PVN, which consequently decreased microglial activation and sympathetic nerve activity. The MI rats exhibited a higher arrhythmia score after programmed electric stimulation than that treated with Mincle siRNA, suggesting that the inhibition of Mincle attenuated foetal ventricular arrhythmias post-MI. The underlying mechanism of Mincle in sympathetic hyperactivity was investigated in lipopolysaccharide (LPS)-primed naïve rats. Recombinant Sin3A-associated protein 130kD (rSAP130), an endogenous ligand for Mincle, induced high levels of NLRP3 and mature IL-1ß protein. PVN-targeted injection of NLRP3 siRNA or IL-1ß antagonist gevokizumab attenuated sympathetic hyperactivity. Together, the data indicated that the knockdown of Mincle in microglia within the PVN prevents VAs by attenuating sympathetic hyperactivity and ventricular susceptibility, in part by inhibiting its downstream NLRP3/IL-1ß axis following MI. Therapeutic interventions targeting Mincle signalling pathway could constitute a novel approach for preventing infarction injury.


Assuntos
Lectinas Tipo C/metabolismo , Microglia/metabolismo , Infarto do Miocárdio/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores Imunológicos/metabolismo , Sistema Nervoso Simpático/metabolismo , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Coração/fisiopatologia , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lectinas Tipo C/genética , Masculino , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Norepinefrina/metabolismo , Interferência de RNA , Ratos Sprague-Dawley , Receptores Imunológicos/genética , Sistema Nervoso Simpático/fisiopatologia
20.
Int J Clin Exp Pathol ; 11(2): 739-747, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-31938160

RESUMO

BACKGROUND: Myocardial infarction (MI) accompanied with abnormal sympathetic innervation, meanwhile, some studies have revealed the oxytocin (OT) and its receptor (OTR) have a relationship with MI and sympathetic system. It is assumed that OT has an close relationship with superior cervical ganglion (SCG), but the existence of oxytocin receptors in SCG has not been well clarified. OBJECTIVE: Our research aims to explore the expression of OTR in SCG in the setting of MI. METHODS AND RESULTS: MI was induced by coronary artery ligation. Rats were randomly assigned to 2 groups: control, MI. The expression of OTR was measured by Western blotting. Distribution of OTR in SCG was investigated by immunofluorescence. Retrograde tracing test revealed the sprouting of tyrosine hydroxylase (TH: the markers of sensory afferent fibers) from cardiac to SCG neurons. The double-immunofluorescence evidence showed that OTR was co-localized and concomitantly changed with TH and the retrograde neuronal labeling from the cardiac afferent nerves. By Western blotting, the protein of OTR in the MI group was higher than those of the control group. CONCLUSIONS: The expression of OTR in SCG after experimental myocardial infarction group was enhanced, suggesting the involvement of OTR in SCG may play a role in the transmission of sympathetic responses after MI.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...