Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell Biol Int ; 46(3): 403-414, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34882885

RESUMEN

Sinoatrial node (SAN) pacemaker cells originate from T-box transcription factor 18 (Tbx18)-expressing progenitor cells. The present study aimed to investigate whether overexpression of human transcription factor Tbx18 could reprogram human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) into SAN-like pacemaker cells (SANLPCs) in vitro. In the study, hiPSCs were first differentiated into hiPSC-CMs through regulating the Wnt/ß-catenin pathway, then purified hiPSC-CMs were transfected by Tbx18 adenovirus (Tbx18-CMs group) or green fluorescent protein (GFP) adenovirus (GFP-CMs group). The beating frequency of the Tbx18-CMs group was significantly higher than that of the hiPSC-CMs group and GFP-CMs group. Compared with the other two groups, the expression levels of hyperpolarization-activated cyclic nucleotide-gated potassium channel isoform 4, connexin-45 in the Tbx18-CMs group were markedly upregulated, while the expressions of transcription factor NKX2.5, CX43 were significantly downregulated. Whole-cell patch-clamp results illustrated that action potential and "funny" current (If ) similar to SAN pacemaker cells could be recorded in the Tbx18-CMs group. In conclusion, this present study demonstrated that overexpression of Tbx18 promoted the conversion of hiPSC-CMs into SANLPCs.


Asunto(s)
Células Madre Pluripotentes Inducidas , Diferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Nodo Sinoatrial/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
2.
Pharmacology ; 106(7-8): 369-383, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33902056

RESUMEN

BACKGROUND/AIMS: Daphnetin (7,8-dihydroxycoumarin, DAP) exhibits various bioactivities, such as anti-inflammatory and antioxidant activities. However, the role of DAP in myocardial ischaemia/reperfusion (I/R) injury and I/R-related arrhythmia is still uncertain. This study aimed to investigate the mechanisms underlying the effects of DAP on myocardial I/R injury and electrophysiological properties in vivo and in vitro. METHODS: Myocardial infarct size was measured by triphenyltetrazolium chloride staining. Cardiac function was assessed by echocardiographic and haemodynamic analyses. The levels of creatine kinase-MB, lactate dehydrogenase, malondialdehyde, superoxide dismutase, interleukin-6 (IL-6), and tumour necrosis factor-alpha (TNF-α) were detected using commercial kits. Apoptosis was measured by terminal deoxynucleotidyl-transferase-mediated dUTP nick-end labelling staining and flow cytometry. The viability of H9c2 cells was determined by the Cell Counting Kit-8 assay. In vitro, the levels of IL-6 and TNF-α were measured by quantitative PCR. The expression levels of proteins associated with apoptosis, inflammation, and the Toll-like receptor 4/myeloid differentiation factor 88/nuclear factor kappa B (TLR4/MyD88/NF-κB) signalling pathway were detected by Western blot analysis. The RR, PR, QRS, and QTc intervals were assessed by surface ECG. The 90% action potential duration (APD90), threshold of APD alternans, and ventricular tachycardia inducibility were measured by the Langendorff perfusion technique. RESULTS: DAP preconditioning decreased myocardial I/R injury and hypoxia/reoxygenation (H/R) injury in cells. DAP preconditioning improved cardiac function after myocardial I/R injury. DAP preconditioning also suppressed apoptosis, attenuated oxidative stress, and inhibited inflammatory responses in vivo and in vitro. Furthermore, DAP preconditioning decreased the susceptibility to ventricular arrhythmia after myocardial I/R. Finally, DAP preconditioning inhibited the expression of TLR4, MyD88, and phosphorylated NF-κB (p-NF-κB)/P65 in mice subjected to I/R and cells subjected to H/R. CONCLUSIONS: DAP preconditioning protected against myocardial I/R injury and decreased susceptibility to ventricular arrhythmia by inhibiting the TLR4/MyD88/NF-κB signalling pathway.


Asunto(s)
Arritmias Cardíacas/prevención & control , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Umbeliferonas/farmacología , Animales , Apoptosis/efectos de los fármacos , Arritmias Cardíacas/etiología , Interleucina-6/metabolismo , Masculino , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , Infarto del Miocardio/etiología , Daño por Reperfusión Miocárdica/complicaciones , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Factor de Transcripción ReIA/metabolismo
3.
Med Sci Monit ; 25: 1984-1993, 2019 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-30877783

RESUMEN

BACKGROUND Augmented cardiac sympathetic afferent reflex (CSAR) plays a role in enhanced sympathetic activity. Given that a strategy for abolishing augmented CSAR-induced sympathetic activation may be beneficial for protecting against ventricular arrhythmias (VAs) triggered by acute myocardial infarction (AMI), we investigated whether cardiac sympathetic afferent denervation (CSAD) could protect against VAs by modulating cardiac sympathetic nerve activity in an AMI dog model. MATERIAL AND METHODS Twenty-two anesthetized dogs were assigned to the CSAD group (n=9) and the sham group (n=13) randomly. CSAD was produced by epicardial application of resiniferatoxin. Heart rate variability (HRV), ventricular action potential duration (APD), APD dispersion, beat-to-beat variability of repolarization (BVR), effective refractory period (ERP) of ventricles, ERP dispersion, plasma norepinephrine (NE) concentration, and left stellate ganglion (LSG) neural activity were determined at baseline and after CSAD. We designed an AMI model by occluding the left anterior coronary artery, and performed analysis of VAs for 60 minutes using electrocardiography. Then, levels of c-fos and nerve growth factor (NGF) were determined. RESULTS Relative to baseline values, CSAD prolonged ERP and APD of ventricles, increased HRV, decreased APD dispersion, BVR, ERP dispersion and serum NE concentration, and attenuated LSG activity in the CSAD group. AMI triggered a remarkable increase in LSG activity and function but decreased the HRV of the sham group animals relative to the CSAD group. Moreover, the CSAD group had higher levels of VAs relative to the sham group. This was accompanied by a corresponding decrease in proteins quantities of NGF and c-fos in the CSAD group in the LSG after AMI compared to the sham group. CONCLUSIONS CSAD can suppress LSG neural activity, hence enhance the electrophysiological stability and protect the heart from AMI-triggered VAs.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Simpatectomía/métodos , Sistema Nervioso Simpático/fisiopatología , Potenciales de Acción , Animales , Arritmias Cardíacas/prevención & control , Ablación por Catéter/métodos , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Perros , Electrocardiografía/métodos , Corazón/fisiopatología , Frecuencia Cardíaca , Ventrículos Cardíacos/fisiopatología , Infarto del Miocardio/fisiopatología , Isquemia Miocárdica/fisiopatología
4.
Biochem Biophys Res Commun ; 505(1): 134-140, 2018 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-30241939

RESUMEN

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.


Asunto(s)
Fibrilación Atrial/genética , Canales de Calcio Tipo L/genética , Regulación de la Expresión Génica , MicroARNs/genética , ARN Largo no Codificante/genética , Factor de Transcripción YY1/genética , Angiotensina II , Animales , Fibrilación Atrial/inducido químicamente , Fibrilación Atrial/fisiopatología , Secuencia de Bases , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Interferencia de ARN , ARN Largo no Codificante/metabolismo , Homología de Secuencia de Ácido Nucleico , Transducción de Señal/genética , Regulación hacia Arriba , Factor de Transcripción YY1/metabolismo
5.
Exp Physiol ; 103(9): 1221-1229, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29928790

RESUMEN

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.


Asunto(s)
Técnicas de Ablación/métodos , Corazón/inervación , Infarto del Miocardio/terapia , Neuronas , Sistema Nervioso Simpático , Disfunción Ventricular Izquierda/terapia , Remodelación Ventricular , Animales , Toxina del Cólera/administración & dosificación , Toxina del Cólera/farmacología , Perros , Ecocardiografía , Expresión Génica , Inyecciones , Masculino , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Sistemas Neurosecretores , Saporinas/administración & dosificación , Saporinas/farmacología , Ganglio Estrellado , Disfunción Ventricular Izquierda/patología
6.
Europace ; 20(12): 2036-2044, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29860489

RESUMEN

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.


Asunto(s)
Potenciales de Acción , Toxina del Cólera , Frecuencia Cardíaca , Corazón/inervación , Infarto del Miocardio/terapia , Saporinas , Ganglio Estrellado/fisiopatología , Simpatectomía Química/métodos , Función Ventricular Izquierda , Remodelación Ventricular , Animales , Biomarcadores/sangre , Modelos Animales de Enfermedad , Perros , Técnicas Electrofisiológicas Cardíacas , Infarto del Miocardio/sangre , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Péptido Natriurético Encefálico/sangre , Factor de Crecimiento Nervioso/sangre , Norepinefrina/sangre , Recuperación de la Función , Factores de Tiempo
7.
J Mol Cell Cardiol ; 109: 1-16, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28668302

RESUMEN

RATIONALE: The cellular mechanisms of obesity/hyperlipidemia-induced cardiac remodeling are many and not completely elucidated. Ca2+/calmodulin-dependent protein kinase II (CaMKII), a multifunctional serine/threonine kinase, has been reported to be involved in a variety of cardiovascular diseases. However, its role in obesity/hyperlipidemia-induced cardiac remodeling is still unknown. OBJECTIVE: The objective of this study was to demonstrate the role of CaMKII in the pathogenesis of obesity/hyperlipidemia-induced cardiac remodeling both in vitro and in vivo. METHODS AND RESULTS: In cardiac-derived H9C2 cells, palmitate treatment induced cell apoptosis coupled with activation of the mitochondrial apoptotic pathway, and cell hypertrophic and fibrotic responses. All of these alterations were inhibited by pharmacological inhibition of CaMKII with either of two specific inhibitors, Myr-AIP and KN93. In addition, an increased inflammatory response coupled with activation of the MAPKs and NF-κB signaling pathway, exaggerated oxidative stress, ER stress and autophagy were also observed in palmitate-treated H9C2 cells, while pretreatment with CaMKII inhibitors decreased these pathological signals. Furthermore, we also demonstrated that TLR4 is upstream signal of CaMKII in palmitate-treated H9C2 cells. In APOE-/- mice fed a high-fat diet (HFD) for 16weeks, serum lipid profiles (FFAs, TG, TC) and blood glucose levels were significantly increased compared with mice fed a normal diet. In addition, apparent cardiac hypertrophy, fibrosis and apoptosis associated with increased inflammation, ER stress, and autophagy were also observed in the hearts of HFD-fed mice. However, all these changes were reversed by 8-weeks of KN93 peritoneal injections. KN93 also increased antioxidant defense as evidenced by increased expression of the Nrf2 system in the hearts of HFD-fed mice. CONCLUSIONS: Taken together, our results demonstrate a critical role of CaMKII in the pathogenesis of obesity/hyperlipidemia-induced cardiac remodeling. Also, TLR4 may be an upstream signal of cardiac CaMKII under hyperlipidemia conditions. These results suggest that CaMKII has the potential to be a therapeutic target in the prevention of obesity/hyperlipidemia-induced cardiac remodeling.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Ácidos Grasos/metabolismo , Hiperlipidemias/enzimología , Mitocondrias Cardíacas/enzimología , Transducción de Señal , Remodelación Ventricular , Animales , Humanos , Hiperlipidemias/patología , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/patología
8.
J Cardiovasc Pharmacol ; 70(4): 245-254, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28662005

RESUMEN

BACKGROUND: Obesity/hyperlipidemia is closely related to both atrial and ventricular arrhythmias. CaMKII, a multifunctional serine/threonine kinase, has been involved in cardiac arrhythmias of different etiologies. However, its role in obesity/hyperlipidemia-related cardiac arrhythmia is unexplored. The aim of this was to determine the involvement of CaMKII in the process. METHODS: Adult male APOE mice were fed a high-fat diet (HFD), administrated with KN93 (10 mg·kg·2d), a specific inhibitor of CaMKII. Serum lipid and glucose profile, cardiac function, and surface electrocardiogram were determined. Electrophysiological study and epicardial activation mapping were performed in Langendorff-perfused heart. Expression of cardiac ion channels, gap junction proteins, Ca handling proteins, and CaMKII were evaluated, coupled with histological analysis. RESULTS: A hyperlipidemia condition was induced by HFD in the APOE mice, which was associated with increased expression and activity of CaMKII in the hearts. In Langendorff-perfused hearts, HFD-induced heart showed increased arrhythmia inducibility, prolonged action potential duration, and decreased action potential duration alternans thresholds, coupled with slow ventricular conduction, connexin-43 upregulation, and interstitial fibrosis. Downregulation of ion channels including Cav1.2 and Kv4.2/Kv4.3 and disturbed Ca handling proteins were also observed in HFD-induced heart. Interestingly, all these alterations were significantly inhibited by KN93 treatment. CONCLUSION: Our results demonstrated an adverse effect of metabolic components on cardiac electrophysiology and implicated an important role of CaMKII underlying this process.


Asunto(s)
Arritmias Cardíacas/enzimología , Arritmias Cardíacas/etiología , Remodelación Atrial/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Dieta Alta en Grasa/efectos adversos , Hiperlipidemias/enzimología , Animales , Activación Enzimática/fisiología , Hiperlipidemias/etiología , Preparación de Corazón Aislado/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
Biochem Biophys Res Commun ; 473(2): 551-7, 2016 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-27021681

RESUMEN

Ischemic heart disease is one of the most common diseases in modern society. Ischemic myocardium can be salvaged by vascular recanalization therapy, but its benefit is attenuated by injury that can occur during reperfusion. And apoptotic cell death plays an important part in myocardial ischemia-reperfusion (IR) injury. Regulator of G-protein signaling 5 (RGS5), highly expressed in different cell types of the human adult heart, is a guanosine triphosphatase-activating protein to inhibit many signaling pathways such as c-Jun NH2-terminal kinase 1/2 (JNK1/2) and p38 which promote cardiac IR-induced apoptosis. However the role of RGS5 in cardiac IR-induced apoptosis remains unclear. An in vitro IR model was applied to the isolated hearts of wild type mice (WT), RGS5-transgenic mice (TG), and RGS5-knockout mice (KO). Our results revealed that compared with either WT or KO mice, TG mice showed inhibition of cardiomyocyte apoptosis as indicated by a greater increase of B cell lymphoma/lewkmia-2 (Bcl-2), and an obvious reduction in the positive expression of the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL), Bcl-2 Associated X protein (Bax), and active caspase-3. Moreover, the inhibition of both JNK1/2 and p38 signaling markedly reversed IR-induced cardiomyocyte apoptosis in RGS5-KO mice. These studies show that RGS5 protects cardiomyocytes against apoptosis during IR through inhibiting both JNK1/2 and p38 signaling pathways.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Isquemia Miocárdica/metabolismo , Miocitos Cardíacos/patología , Proteínas RGS/metabolismo , Transducción de Señal , Animales , Apoptosis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Isquemia Miocárdica/genética , Isquemia Miocárdica/patología , Miocitos Cardíacos/metabolismo , Proteínas RGS/genética
11.
Sci Transl Med ; 14(626): eabk1707, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34985971

RESUMEN

Thrombosis is the leading complication of common human disorders including diabetes, coronary heart disease, and infection and remains a global health burden. Current anticoagulant therapies that target the general clotting cascade are associated with unpredictable adverse bleeding effects, because understanding of hemostasis remains incomplete. Here, using perturbational screening of patient peripheral blood samples for latent phenotypes, we identified dysregulation of the major mechanosensory ion channel Piezo1 in multiple blood lineages in patients with type 2 diabetes mellitus (T2DM). Hyperglycemia activated PIEZO1 transcription in mature blood cells and selected high Piezo1­expressing hematopoietic stem cell clones. Elevated Piezo1 activity in platelets, red blood cells, and neutrophils in T2DM triggered discrete prothrombotic cellular responses. Inhibition of Piezo1 protected against thrombosis both in human blood and in zebrafish genetic models, particularly in hyperglycemia. Our findings identify a candidate target to precisely modulate mechanically induced thrombosis in T2DM and a potential screening method to predict patient-specific risk. Ongoing remodeling of cell lineages in hematopoiesis is an integral component of thrombotic risk in T2DM, and related mechanisms may have a broader role in chronic disease.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hiperglucemia , Trombosis , Animales , Humanos , Hiperglucemia/complicaciones , Canales Iónicos/metabolismo , Mecanotransducción Celular , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
12.
Biomed Pharmacother ; 120: 109500, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31600641

RESUMEN

OBJECTIVES: Findings from prior investigations show that left stellate ganglion (LSG) inhibitory approaches protect the heart from ventricular arrhythmias (VAs) caused by acute myocardial infarction (AMI), which still remain many side effects. Targeted transient receptor potential vanilloid 1/tyrosine hydroxylase (TRPV-1/TH) expressing sympathetic neurons ablation is a novel neuro-ablative strategy. The aim of this investigation was to explore if targeted molecular neuro-ablative strategy by resiniferatoxin (RTX) stellate microinjection could protect against ischemia-induced VAs. METHODS: Twenty-four anesthetized beagles were assigned to a control group (n = 12) and RTX group (n = 12) in a random manner. Targeted molecular neuro-ablative was produced by RTX stellate microinjection and DMSO was microinjected into LSG in the same way as control. Plasma norepinephrine (NE) level, heart rate variability (HRV), Tpeak-Tend interval (Tp-Te), LSG neural activity and function, ventricular effective refractory period (ERP), beat-to-beat variability of repolarization (BVR) and ventricular action potential duration (APD) were measured at baseline and 60 min after RTX or DMSO microinjection. AMI model was established by the ligation of left anterior descending coronary artery and 60-minute electrocardiography was continuously recorded for VAs analysis. Subsequently, HRV, Tp-Te, plasma NE level from jugular vein and coronary sinus, LSG neural activity and function, ventricular ERP, ventricular APD, BVR, action potential duration alternans (APDA) cycle length and ventricular fibrillation threshold (VFT) were evaluated after AMI. Finally, tissue collection of LSG was performed for examining the TRPV-1, nerve growth factor (NGF) protein and c-fos protein. RESULTS: TRPV-1 was highly expressed in the TH-expressing neurons and RTX injection significantly ablated TRPV-1/TH-positive neurons in LSG. Compared with baseline, RTX stellate microinjection significantly reduced plasma NE level, the sympathetic component of HRV, LSG neural activity and LSG function, shortened Tp-Te, prolonged ventricular ERP and APD, but there were no remarkable differences existed for control group. AMI resulted in the significant raise in plasma NE level from jugular vein and coronary sinus, the sympathetic component of HRV, LSG neural activity and LSG function, the marked prolongation in Tp-Te and BVR, the significant decrease in ERP and APD from ischemia area, and the increase in APDA cycle length in the ischemic region of the control group, which were remarkably attenuated in the RTX group. RTX pretreatment markedly rose the VFT in the RTX group. Furthermore, the AMI-triggered VAs was significantly prevented by RTX injection in the RTX group. RTX microinjection down-regulated significantly TRPV-1, NGF and c-fos expression in the LSG compared with the control group. CONCLUSION: Targeted ablation of TRPV-1/TH positive sympathetic neurons induced by RTX stellate microinjection could suppress ischemia-induced cardiac autonomic imbalances and cardiac electrophysiology instability to protect against AMI-induced VAs.


Asunto(s)
Arritmias Cardíacas/prevención & control , Arritmias Cardíacas/veterinaria , Diterpenos/farmacología , Isquemia Miocárdica , Neuronas/fisiología , Ganglio Estrellado/citología , Canales Catiónicos TRPV/metabolismo , Técnicas de Ablación , Animales , Perros , Regulación de la Expresión Génica/efectos de los fármacos , Canales Catiónicos TRPV/genética , Tirosina 3-Monooxigenasa/metabolismo , Fibrilación Ventricular/etiología , Fibrilación Ventricular/fisiopatología
13.
Mol Med Rep ; 18(6): 5520-5526, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30365101

RESUMEN

Intracardiac injection of the growth­promoting transcription factor of the sinoatrial node T­box18 (TBX18) has been shown to reprogram cardiomyocytes into induced sinoatrial nodes that produce high­frequency and neuroregulated ectopic beats. Fibroblasts, the most important non­cardiomyocyte cell type in the heart, can affect the electrophysiological properties of cardiomyocytes by electrically coupling with them. The aim of the present study was to explore the reprogramming of cardiac fibroblasts (CFs) transfected with TBX18 in vitro and observe its effect on the pacing frequency of neonatal rat ventricular cardiomyocytes (NRVMs) in a co­culture system. CFs transfected with TBX18 could be transformed into cardiac myofibroblasts that expressed high levels of hyperpolarization­activated cyclic nucleotide­gated cation channel 4 protein and low levels of connexin 43 (COX43) and COX45 protein. In addition, TBX18­CFs could increase the beating rates of NRVMs and TBX18­NRVMs in a co­culture system. The results of the present study indicated that the TBX18 gene could induce CFs to undergo a transformation that promotes an increase of the beating rates of NRVMs and TBX18­NRVMs.


Asunto(s)
Reprogramación Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas de Dominio T Box/genética , Animales , Diferenciación Celular/genética , Técnicas de Cocultivo , Expresión Génica , Ratas , Transfección
14.
Kardiol Pol ; 75(4): 376-385, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28150281

RESUMEN

BACKGROUND: The recurrence rate of atrial fibrillation (AF) after cryoballoon ablation in drug refractory AF patients is high. Late-recurrence of AF has various predictors. AIM: The aim of the study was to explore the related risk factors that can effectively predict late AF recurrence after cryoballoon-based pulmonary vein isolation. METHODS: The PubMed and Web of Science databases were searched from 1 January 2013 to 1 August 2016, and studies were chosen that met the pre-stated inclusion criteria. The reference lists of the retrieved articles were also reviewed. Two authors independently extracted information on the designs of the studies. The strength of the relationship between different risk factors and late recurrence was assessed by the adjusted hazard ratio. RESULTS: A total of 16 papers met the inclusion criteria and were included in the meta-analysis. The hazard ratio of late atrial arrhythmia recurrence in patients with early recurrence was 4.19 compared with the reference group (95% CI 2.73-6.44, p < 0.00001); that of increased left atrial diameter was 1.25 (95% CI 1.12-1.3, p < 0.0001); that of a long duration of AF before ablation was 1.10 (95% CI 1.04-1.17, p < 0.0009); and that of persistent AF was 2.44 (95% CI 1.30-4.58, p < 0.006). However, there exists significant heterogeneity for each indicator, and a slight publication bias was observed. CONCLUSIONS: Our study suggests that early recurrence in the blanking period, increased left atrial size, a longduration of AF before ablation and persistent AF are independent predictors of late recurrence after cryoballoon ablation.


Asunto(s)
Fibrilación Atrial/cirugía , Ablación por Catéter , Criocirugía , Venas Pulmonares/cirugía , Adulto , Anciano , Fibrilación Atrial/epidemiología , Fibrilación Atrial/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Recurrencia , Factores de Riesgo
15.
Sci Rep ; 7: 41857, 2017 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-28165494

RESUMEN

Myeloid differentiation 1 (MD-1), a secreted protein interacting with radioprotective 105 (RP105), plays an important role in Toll-like receptor 4 (TLR4) signalling pathway. Previous studies showed that MD-1 may be restricted in the immune system. In this study, we demonstrated for the first time that MD-1 was highly expressed in both human and animal hearts. We also discovered that cardiac-specific overexpression of MD-1 significantly attenuated pressure overload-induced cardiac hypertrophy, fibrosis, and dysfunction, whereas loss of MD-1 had the opposite effects. Similar results were observed for in vitro angiotensin II-induced neonatal rat cardiomyocyte hypertrophy. The antihypertrophic effects of MD-1 under hypertrophic stimuli were associated with the blockage of MEK-ERK 1/2 and NF-κB signalling. Blocking MEK-ERK 1/2 signalling with a pharmacological inhibitor (U0126) greatly attenuated the detrimental effects observed in MD-1 knockout cardiomyocytes exposed to angiotensin II stimuli. Similar results were observed by blocking NF-κB signalling with a pharmacological inhibitor (BAY11-7082). Our data indicate that MD-1 inhibits cardiac hypertrophy and suppresses cardiac dysfunction during the remodelling process, which is dependent on its modulation of the MEK-ERK 1/2 and NF-κB signalling pathways. Thus, MD-1 might be a novel target for the treatment of pathological cardiac hypertrophy.


Asunto(s)
Antígenos de Superficie/genética , Cardiomegalia/metabolismo , Glicoproteínas de Membrana/genética , Remodelación Ventricular , Angiotensina II/farmacología , Animales , Antígenos de Superficie/metabolismo , Cardiomegalia/patología , Células Cultivadas , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Nitrilos/farmacología , Sulfonas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA