Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38902033

RESUMEN

Sympathetic hyperinnervation is the leading cause of fatal ventricular arrhythmia (VA) following 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 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 1.We confirmed the role and mechanism of oxytocin (OT) in stabilizing cardiac mast cells. 2. It is the first study to elucidate the mechanism of oxytocin (OT)-mediated sympathetic hyperinnervation post-myocardial infarction (MI).

2.
Am Heart J ; 260: 34-43, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36813122

RESUMEN

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.


Asunto(s)
Fibrilación Atrial , Ablación por Catéter , Venas Pulmonares , Humanos , Fibrilación Atrial/cirugía , Venas Pulmonares/cirugía , Estudios Prospectivos , Atrios Cardíacos/cirugía , Etanol , Ablación por Catéter/métodos , Resultado del Tratamiento , Recurrencia
3.
J Mol Cell Cardiol ; 170: 87-99, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35717715

RESUMEN

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.


Asunto(s)
Infarto del Miocardio , FN-kappa B , Animales , Ratas , Arritmias Cardíacas , Metiltransferasas/metabolismo , Infarto del Miocardio/metabolismo , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Remodelación Ventricular
4.
J Cell Mol Med ; 26(10): 2959-2971, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35393774

RESUMEN

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.


Asunto(s)
Infarto del Miocardio , Núcleo Hipotalámico Paraventricular , Animales , Arritmias Cardíacas/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Infarto del Miocardio/metabolismo , FN-kappa B/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
5.
J Cell Mol Med ; 26(4): 1264-1280, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35040253

RESUMEN

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.


Asunto(s)
Metiltransferasas , Infarto del Miocardio , Animales , Corazón , Humanos , Metilación , Metiltransferasas/metabolismo , Infarto del Miocardio/complicaciones , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas , Sistema Nervioso Simpático/metabolismo
6.
J Cell Mol Med ; 23(1): 112-125, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30353660

RESUMEN

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.


Asunto(s)
Lectinas Tipo C/metabolismo , Microglía/metabolismo , Infarto del Miocardio/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores Inmunológicos/metabolismo , Sistema Nervioso Simpático/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Corazón/fisiopatología , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lectinas Tipo C/genética , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Norepinefrina/metabolismo , Interferencia de ARN , Ratas Sprague-Dawley , Receptores Inmunológicos/genética , Sistema Nervioso Simpático/fisiopatología
7.
Lipids Health Dis ; 18(1): 150, 2019 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-31286992

RESUMEN

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.


Asunto(s)
Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/mortalidad , Lipoproteína(a)/sangre , Síndrome Coronario Agudo/sangre , Síndrome Coronario Agudo/etiología , Enfermedad de la Arteria Coronaria/complicaciones , Humanos , Pronóstico , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/etiología
8.
J Cell Mol Med ; 21(11): 2695-2710, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28470940

RESUMEN

Mounting evidence supports the hypothesis that inflammation modulates sympathetic sprouting after myocardial infarction (MI). The myeloid P2X7 signal has been shown to activate the nucleotide-binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a master regulator of inflammation. We investigated whether P2X7 signal participated in the pathogenesis of sympathetic reinnervation after MI, and whether NLRP3/interleukin-1ß (IL-1ß) axis is involved in the process. We explored the relationship between P2X7 receptor (P2X7 R) and IL-1ß in the heart tissue of lipopolysaccharide (LPS)-primed naive rats. 3'-O-(4-benzoyl) benzoyl adenosine 5'-triphosphate (BzATP), a P2X7 R agonist, induced caspase-1 activation and mature IL-1ß release, which was further neutralized by a NLRP3 inhibitor (16673-34-0). MI was induced by coronary artery ligation. Following infarction, a marked increase in P2X7 R was localized within infiltrated macrophages and observed in parallel with an up-regulation of NLRP3 inflammasome levels and the release of IL-1ß in the left ventricle. The administration of A-740003 (a P2X7 R antagonist) significantly prevented the NLRP3/IL-1ß increase. A-740003 and/or Anakinra (an IL-1 receptor antagonist) significantly reduced macrophage infiltration as well as macrophage-based IL-1ß and NGF (nerve growth factor) production and eventually blunted sympathetic hyperinnervation, as assessed by the immunofluorescence of tyrosine hydroxylase (TH) and growth-associated protein 43 (GAP 43). Moreover, the use of Anakinra partly attenuated sympathetic sprouting. This indicated that the effect of P2X7 on neural remodelling was mediated at least partially by IL-1ß. The arrhythmia score of programmed electric stimulation was in accordance with the degree of sympathetic hyperinnervation. In vitro studies showed that BzATP up-regulated secretion of nerve growth factor (NGF) in M1 macrophages via IL-1ß. Together, these data indicate that P2X7 R contributes to neural and cardiac remodelling, at least partly mediated by NLRP3/IL-1ß axis. Therapeutic interventions targeting P2X7 signal may be a novel approach to ameliorate arrhythmia following MI.


Asunto(s)
Interleucina-1beta/genética , Infarto del Miocardio/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Receptores Purinérgicos P2X7/genética , Sistema Nervioso Simpático/efectos de los fármacos , Acetamidas/farmacología , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Caspasas/genética , Caspasas/metabolismo , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Estimulación Eléctrica , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Regulación de la Expresión Génica , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Inflamación , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1beta/metabolismo , Ligadura , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factor de Crecimiento Nervioso/genética , Factor de Crecimiento Nervioso/metabolismo , Agonistas del Receptor Purinérgico P2X/farmacología , Antagonistas del Receptor Purinérgico P2X/farmacología , Quinolinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2X7/metabolismo , Transducción de Señal , Sistema Nervioso Simpático/metabolismo
9.
Respir Res ; 18(1): 127, 2017 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-28646872

RESUMEN

BACKGROUND: Pulmonary arterial hypertension (PAH) is a devastating disease that lacks sufficient treatment. Studies have shown that the Nod-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome contributes to PAH pathogenesis, but the role of the upstream molecular P2X7 receptor (P2X7R) has remained unexplored. We investigated the role of P2X7R in the pathogenesis of PAH. METHODS AND RESULTS: PH was induced by a single subcutaneous injection of monocrotaline (MCT) (60 mg/kg) on left pneumonectomised Sprague-Dawley rats, as validated by significant increases in pulmonary artery pressure and vessel wall thickness. Marked P2X7R was detected by predominant PA immunostaining in lungs from PH rats. Western blot revealed a significant increase in the protein levels of P2X7R as well as NLRP3 and caspase-1 in the diseased lung tissue compared with normal tissue. The rats received A-740003 (a selective P2X7 receptor antagonist, 30 mg/kg) daily starting from 1 week before or 2 weeks after MCT injection. Consequently, A-740003 reversed the NLRP3 inflammasome upregulation, significantly decreased the mean right ventricular (RV) pressure and RV hypertrophy, and reversed pulmonary arterial remodelling 4 weeks after MCT injection, as both a pretreatment and rescue intervention. Notably, A-740003 significantly reduced macrophage and pro-inflammatory cytokine levels, as measured via bronchoalveolar lavage. The recruitment of macrophages as well as collagen fibre deposition in the perivascular areas were also reduced, as confirmed by histological staining. CONCLUSIONS: P2X7R contributes to the pathogenesis of PH, probably in association with activation of the NLRP3 inflammasome. Blockade of P2X7R might be applied as a novel therapeutic approach for the treatment of PAH.


Asunto(s)
Progresión de la Enfermedad , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Receptores Purinérgicos P2X7/fisiología , Animales , Relación Dosis-Respuesta a Droga , Masculino , Antagonistas del Receptor Purinérgico P2X/farmacología , Ratas , Ratas Sprague-Dawley
10.
Am J Physiol Cell Physiol ; 310(1): C41-53, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26491050

RESUMEN

Inflammation-dominated sympathetic sprouting adjacent to the necrotic region following myocardial infarction (MI) has been implicated in the etiology of arrhythmias resulting in sudden cardiac death; however, the mechanisms responsible remain to be elucidated. Although being a key immune mediator, the role of Notch has yet to be explored. We investigated whether Notch regulates macrophage responses to inflammation and affects cardiac sympathetic reinnervation in rats undergoing MI. MI was induced by coronary artery ligation. A high level of Notch intracellular domain was observed in the macrophages that infiltrated the infarct area at 3 days post-MI. The administration of the Notch inhibitor N-N-(3,5-difluorophenacetyl-L-alanyl)-S-phenylglycine-t-butyl ester (DAPT) (intravenously 30 min before MI and then daily until death) decreased the number of macrophages and significantly increased the M2 macrophage activation profile in the early stages and attenuated the expression of nerve growth factor (NGF). Eventually, NGF-induced sympathetic hyperinnervation was blunted, as assessed by the immunofluorescence of tyrosine hydroxylase. At 7 days post-MI, the arrhythmia score of programmed electric stimulation in the vehicle-treated infarcted rats was higher than that in rats treated with DAPT. Further deterioration in cardiac function and decreases in the plasma levels of TNF-α and IL-1ß were also detected. In vitro studies revealed that LPS/IFN-γ upregulated the surface expression of NGF in M1 macrophages in a Notch-dependent manner. We concluded that Notch inhibition during the acute inflammatory response phase is associated with the downregulation of NGF, probably through a macrophage-dependent pathway, thus preventing the process of sympathetic hyperinnervation.


Asunto(s)
Antiinflamatorios/farmacología , Dipéptidos/farmacología , Corazón/inervación , Macrófagos/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Miocardio/metabolismo , Receptor Notch1/antagonistas & inhibidores , Sistema Nervioso Simpático/efectos de los fármacos , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/prevención & control , Estimulación Cardíaca Artificial , Células Cultivadas , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Factor de Crecimiento Nervioso/metabolismo , Fenotipo , Ratas Sprague-Dawley , Receptor Notch1/metabolismo , Transducción de Señal/efectos de los fármacos , Sistema Nervioso Simpático/metabolismo , Sistema Nervioso Simpático/fisiopatología , Factor de Necrosis Tumoral alfa/metabolismo
11.
Respir Res ; 17(1): 99, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27491388

RESUMEN

BACKGROUND: Pulmonary arterial hypertension (PAH) is a progressive and life-threatening disease associated with high morbidity and mortality rates. However, the exact regulatory mechanism of PAH is unknown. Although coupling factor 6 (CF6) is known to function as a repressor, its role in PAH has not been explored. Here, we investigated the involvement of endogenous CF6 in the development of PAH. METHODS: PAH was induced with monocrotaline (MCT), as demonstrated by significant increases in pulmonary artery pressure and vessel wall thickness. The adeno-associated virus (AAV) carrying CF6 short hairpin RNA (shRNA) or control vector (2×10(10) gp) was intratracheally transfected into the lungs of rats 2 weeks before or after MCT injection. RESULTS: A 2-6-fold increase in CF6 was observed in the lungs and circulation of the MCT-injected rats as confirmed by qRT-PCR and ELISA. Immunohistochemistry analysis revealed a small quantity of CF6 localized to endothelial cells (ECs) under physiological conditions spread to surrounding tissues in a paracrine manner in PAH lungs. Notably, CF6 shRNA effectively inhibited CF6 expression, abolished lung macrophage infiltration, reversed endothelial dysfunction and vascular remodeling, and ameliorated the severity of pulmonary hypertension and right ventricular dysfunction at 4 weeks both as a pretreatment and rescue intervention. In addition, the circulating and lung levels of 6-keto-PGF1a, a stable metabolite of prostacyclin, were reversed by CF6 inhibition, suggesting that the effect of CF6 inhibition may partly be mediated through prostacyclin. CONCLUSIONS: CF6 contributes to the pathogenesis of PAH, probably in association with downregulation of prostacyclin. The blockage of CF6 might be applied as a novel therapeutic approach for PAH and PA remodeling.


Asunto(s)
Terapia Genética/métodos , Hipertensión Pulmonar/terapia , Pulmón/metabolismo , ATPasas de Translocación de Protón Mitocondriales/antagonistas & inhibidores , ATPasas de Translocación de Protón Mitocondriales/genética , Factores de Acoplamiento de la Fosforilación Oxidativa/antagonistas & inhibidores , Factores de Acoplamiento de la Fosforilación Oxidativa/genética , Interferencia de ARN , 6-Cetoprostaglandina F1 alfa/metabolismo , Animales , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Hipertensión Pulmonar/inducido químicamente , Inyecciones Espinales , Pulmón/patología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Monocrotalina , Infiltración Neutrófila , Factores de Acoplamiento de la Fosforilación Oxidativa/metabolismo , Arteria Pulmonar/patología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Remodelación Vascular , Disfunción Ventricular Derecha/etiología , Disfunción Ventricular Derecha/genética , Disfunción Ventricular Derecha/prevención & control
12.
J Cardiovasc Pharmacol ; 67(3): 252-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26566151

RESUMEN

BACKGROUND: Myocardial infarction (MI) results in decreased inward-rectifier K⁺ current (IK1), which is mediated primarily by the Kir2.1 protein and is accompanied by upregulated T cells. Interferon γ (IFN-γ), secreted predominantly by Th1 cells, causes a decrease in IK1 in microglia. Whether Th1 cells can induce IK1/Kir2.1 remodeling following MI and whether valsartan can ameliorate this phenomenon remain unclear. METHODS: Rats experiencing MI received either valsartan or saline for 7 days. Th1-enriched lymphocytes and myocytes were cocultured with or without valsartan treatment. Th1 cells were monitored by flow cytometry. The protein levels of Kir2.1 were detected by Western blot analyses. IK1 was recorded through whole-cell patch clamping. The plasma levels of IFN-γ, interleukin 2, and tumor necrosis factor α were detected by enzyme-linked immunosorbent assay. RESULTS: Th1 cell number and cytokine expression levels were higher following MI, and the Kir2.1 protein level was decreased. In MI rats, valsartan reduced Th1 cell number and cytokine expression levels and increased the Kir2.1 expression and the IK1 current compared with the rats that received saline treatment; these results are consistent with the effect of valsartan in cocultured lymphocytes and myocytes. In vitro, IFN-γ overexpression suppressed the IK1 current, whereas interleukin 2 and tumor necrosis factor α had no significant effect on the current, establishing that Th1 cell regulation of IK1/Kir2.1 expression is mainly dependent on IFN-γ. CONCLUSIONS: Valsartan ameliorates IK1/Kir2.1 remodeling by downregulating the Th1 immune response following MI.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Infarto del Miocardio/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Células TH1/efectos de los fármacos , Valsartán/farmacología , Animales , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Hemodinámica/efectos de los fármacos , Interferón gamma/sangre , Interleucina-2/sangre , Masculino , Potenciales de la Membrana , Infarto del Miocardio/sangre , Infarto del Miocardio/inmunología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/metabolismo , Potasio/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Ratas Wistar , Células TH1/inmunología , Células TH1/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/sangre
13.
BMC Cardiovasc Disord ; 16: 16, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26787044

RESUMEN

BACKGROUND: To investigate the effects of semaphorin 3A (sema 3A) on cardiac autonomic regulation and subsequent ventricular arrhythmias (VAs) in post-infarcted hearts. METHOD AND RESULTS: In order to explore the functions of sema 3A in post-infarcted hearts, lentivirus-Sema 3A-shRNA and negative control vectors were delivered to the peri-infarcted myocardium rats respectively. Meanwhile, recombinant sema 3A and control (0.9% NaCl solution) were injected intravenously into infarcted rats to test the therapeutic potential of sema 3A. Results indicated that levels of sema 3A were higher in post-infarcted hearts compared with sham rats. However, sema 3A silencing leaded to sympathetic hyperinnervation, increased myocardial norepinephrine (NE) content and inducible VAs. Conversely, the intravenous administration of sema 3A to infarcted rats reduced sympathetic nerve sprouting, improved cardiac autonomic regulation, and decreased the incidence of inducible VAs. However, both infarct size and cardiac function were similar among infarcted hearts. CONCLUSIONS: The upregulation and administration of sema 3A exerted beneficial effects on infarction-induced cardiac autonomic disorders by increasing cardiac electrical stability and reducing VAs. Sema 3A might be a potential therapeutic agent for cardiac autonomic abnormalities induced arrhythmias.


Asunto(s)
Corazón/efectos de los fármacos , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Semaforina-3A/genética , Sistema Nervioso Simpático/metabolismo , Taquicardia Ventricular/genética , Fibrilación Ventricular/genética , Animales , Sistema Nervioso Autónomo/efectos de los fármacos , Sistema Nervioso Autónomo/metabolismo , Sistema Nervioso Autónomo/fisiopatología , Western Blotting , Cromatografía Líquida de Alta Presión , Electrocardiografía , Técnicas de Silenciamiento del Gen , Corazón/inervación , Corazón/fisiopatología , Inmunohistoquímica , Masculino , Infarto del Miocardio/metabolismo , Norepinefrina/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/farmacología , Semaforina-3A/farmacología , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiopatología , Taquicardia Ventricular/fisiopatología , Fibrilación Ventricular/fisiopatología
14.
J Appl Toxicol ; 36(2): 257-65, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26011702

RESUMEN

The kidney is the principal organ targeted by exposure to cadmium (Cd), a well-known toxic metal. Even at a low level, Cd damages glomerular filtration. However, little is known about the effects of Cd on the glomerular endothelium, which performs the filtration function and directly interacts with Cd in blood plasma. In this study, we cultured human renal glomerular endothelial cells (HRGECs) in the presence of serum with treatment of a short term (1 h) and low concentration (1 µm) of Cd, which mimics the pattern of glomerular endothelium exposure to Cd in vivo. We found that this short-term, low-dose Cd exposure does not induce cytotoxicity, but increases permeability in HRGECs monolayers and redistributes adherens junction proteins vascular endothelial-cadherin and ß-catenin. Though short-term, low-dose Cd exposure activates all three major mitogen activated protein kinases, only the inhibitor of p38 mitogen activated protein kinase partially prevents Cd-induced hyperpermeability in HRGECs. Our data indicate that the presence of Cd in blood circulation might directly disrupt the glomerular endothelial cell barrier and contribute to the development of clinical symptoms of glomerular diseases.


Asunto(s)
Cadmio/toxicidad , Células Cultivadas/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Tasa de Filtración Glomerular/efectos de los fármacos , Riñón/efectos de los fármacos , Permeabilidad/efectos de los fármacos , Cadmio/sangre , Aumento de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos
15.
Cardiovasc Drugs Ther ; 29(3): 209-18, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26095682

RESUMEN

PURPOSE: Myocardial infarction (MI) results in an increased susceptibility to ventricular arrhythmias, due in part to decreased inward-rectifier K+ current (IK1), which is mediated primarily by the Kir2.1 protein. The use of renin-angiotensin-aldosterone system antagonists is associated with a reduced incidence of ventricular arrhythmias. Casein kinase 2 (CK2) binds and phosphorylates SP1, a transcription factor of KCNJ2 that encodes Kir2.1. Whether valsartan represses CK2 activation to ameliorate IK1 remodeling following MI remains unclear. METHODS: Wistar rats suffering from MI received either valsartan or saline for 7 days. The protein levels of CK2 and Kir2.1 were each detected via a Western blot analysis. The mRNA levels of CK2 and Kir2.1 were each examined via quantitative real-time PCR. RESULTS: CK2 expression was higher at the infarct border; and was accompanied by a depressed IK1/Kir2.1 protein level. Additionally, CK2 overexpression suppressed KCNJ2/Kir2.1 expression. By contrast, CK2 inhibition enhanced KCNJ2/Kir2.1 expression, establishing that CK2 regulates KCNJ2 expression. Among the rats suffering from MI, valsartan reduced CK2 expression and increased Kir2.1 expression compared with the rats that received saline treatment. In vitro, hypoxia increased CK2 expression and valsartan inhibited CK2 expression. The over-expression of CK2 in cells treated with valsartan abrogated its beneficial effect on KCNJ2/Kir2.1. CONCLUSIONS: AT1 receptor antagonist valsartan reduces CK2 activation, increases Kir2.1 expression and thereby ameliorates IK1 remodeling after MI in the rat model.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Infarto del Miocardio/metabolismo , Canales de Potasio de Rectificación Interna/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Valsartán/farmacología , Animales , Masculino , Potenciales de la Membrana/efectos de los fármacos , Infarto del Miocardio/enzimología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Cultivo Primario de Células , Ratas
16.
Cardiology ; 127(3): 155-63, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24356397

RESUMEN

OBJECTIVES: Diabetic cardiac autonomic neuropathy can lead to an increased incidence of ventricular arrhythmias (VAs). However, few data are available regarding the pathogenesis and therapy of the VAs accompanying diabetic cardiac autonomic neuropathy. We aimed to explore whether or not exogenous nerve growth factor (NGF) can reduce the sympathetic heterogeneity and the incidence of VAs in diabetes mellitus (DM). METHODS: Male Wistar rats were randomly divided into 3 groups: controls, rats with DM with saline infused into the left stellate ganglion (LSG), i.e. the DS group and rats with DM with NGF infused into the LSG, i.e. the DN group. After 28 weeks, all rats were subjected to electrophysiological experiments. Sympathetic innervations and NGF were studied by immunostaining, RT-PCR or Western blot analysis. RESULTS: The incidence of inducible VAs was significantly higher in the DS group than in the control group, but was markedly decreased in the DN group. In the DS group, the tyrosine hydroxylase (TH) and NGF expression were significantly lower than in the other groups, and significant proximal-distal heterogeneities existed regarding the TH and NGF expression in the left ventricle, but were markedly repaired in the DN group. CONCLUSIONS: NGF intervention in the LSG can reduce the heterogeneity of cardiac sympathetic innervations and the incidence of VAs in diabetic rats.


Asunto(s)
Antiarrítmicos/farmacología , Arritmias Cardíacas/tratamiento farmacológico , Diabetes Mellitus Experimental/tratamiento farmacológico , Neuropatías Diabéticas/tratamiento farmacológico , Factor de Crecimiento Nervioso/farmacología , Animales , Biomarcadores/metabolismo , Estimulación Eléctrica , Masculino , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas Wistar
17.
Tissue Cell ; 88: 102371, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38593570

RESUMEN

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.


Asunto(s)
Acetofenonas , Hipoxia de la Célula , Regulación hacia Arriba , Acetofenonas/farmacología , Animales , Ratas , Regulación hacia Arriba/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Canales Iónicos/metabolismo , Canales Iónicos/genética , Apoptosis/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/patología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Ratas Sprague-Dawley
18.
Cardiology ; 126(1): 50-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23860330

RESUMEN

OBJECTIVES: Systemic or local inflammation causes cardiac nerve sprouting and consequent arrhythmia. Metoprolol can prevent sympathetic nerve remodeling after myocardial infarction (MI), but the underlying mechanism is unclear. In this study, we evaluated the role of metoprolol in ameliorating sympathetic sprouting. METHODS: Rabbits underwent ligation of the coronary artery for MI. MI rabbits received metoprolol or saline for 7 days. Immunohistochemistry was used to measure cardiac nerve sprouting and sympathetic innervations. Nuclear factor-κB (NF-κB) DNA binding activity was analyzed by electrophoretic mobility shift assay. The protein levels of NF-κB p65, inhibitor κBα (IκBα) and nerve growth factor (NGF) were detected by Western blot analysis. The mRNA levels of NGF, interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) were examined by quantitative real-time PCR. RESULTS: MI rabbits showed nerve sprouting and sympathetic hyperinnervation. In MI rabbits, as compared with saline treatment, metoprolol reduced NF-κB DNA binding activity and NF-κB p65 level, and increased IκBα level. Moreover, metoprolol downregulated IL-1ß, TNF-α and NGF levels, and reduced the density of sympathetic nerve fibers. CONCLUSIONS: Metoprolol ameliorates sympathetic nerve sprouting in rabbits after MI and is associated in part with inhibiting NF-κB activity.


Asunto(s)
Corazón/inervación , Metoprolol/farmacología , Infarto del Miocardio/fisiopatología , Sistema Nervioso Simpático/efectos de los fármacos , Simpaticolíticos/farmacología , Animales , Vasos Coronarios , Proteínas I-kappa B/metabolismo , Interleucina-1beta/metabolismo , Ligadura , Masculino , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Factor de Crecimiento Nervioso/metabolismo , ARN Mensajero/metabolismo , Conejos , Factor de Necrosis Tumoral alfa/metabolismo
19.
Heart Lung Circ ; 22(12): 1018-25, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23850388

RESUMEN

BACKGROUND: Diabetic cardiac autonomic neuropathy (DCAN) may cause fatal ventricular arrhythmias and increase mortality in diabetics. Mesenchymal stem cells (MSCs) can secrete various cytokines and growth factors exerting neurosupportive effects. In this study, we investigated the effect of MSC on DCAN in diabetic rats. METHODS: Forty rats were divided into normal control, diabetes mellitus (DM) control, MSC treatment (6 × 10(6) MSCs via direct myocardial injection) and MSC-conditioned medium group (100 µl via direct myocardial injection). Immunohistochemistry was used to measure choline acetyltransferase (ChAT, a marker for parasympathetic nerves) and tyrosine hydroxylase (TH, a marker for sympathetic nerves) positive nerve fibres in the ventricular myocardium. Heart rate variability and programmed electrical stimulation was used to assess the inducibility of ventricular arrhythmias in the animals. RESULTS: Two weeks after MSC treatment, the density of ChAT- and TH-positive nerve fibres in MSCs and MSC-conditioned medium group was higher than in DM control group (P < 0.05 or P < 0.01). The ChAT/TH ratio in MSC group was higher than in DM control group (0.37 ± 0.014 vs. 0.27 ± 0.020, P < 0.01). The standard deviation of normal-to-normal R-R intervals in MSCs (5.13 ± 0.69) and MSC-conditioned medium group (4.30 ± 0.56) was higher than in DM control group (3.45 ± 0.60, P < 0.05). The inducibility of VAs in the MSC group was lower than in the DM control group. CONCLUSIONS: MSC therapy may promote cardiac nerve sprouting and increase the ratio of parasympathetic to sympathetic nerve fibres. It may also suppress the inducibility of ventricular arrhythmias in the diabetic rats.


Asunto(s)
Arritmias Cardíacas , Diabetes Mellitus Experimental , Cardiomiopatías Diabéticas , Sistema de Conducción Cardíaco , Trasplante de Células Madre Mesenquimatosas , Aloinjertos , Animales , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Arritmias Cardíacas/terapia , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Experimental/terapia , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/fisiopatología , Cardiomiopatías Diabéticas/terapia , Sistema de Conducción Cardíaco/patología , Sistema de Conducción Cardíaco/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley
20.
Free Radic Biol Med ; 209(Pt 2): 342-354, 2023 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-37898386

RESUMEN

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.


Asunto(s)
Infarto del Miocardio , Factor 6 Asociado a Receptor de TNF , Animales , Ratas , Metiltransferasas/genética , Metiltransferasas/metabolismo , Mitocondrias/metabolismo , Infarto del Miocardio/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA