Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Pineal Res ; 74(3): e12851, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36639364

RESUMEN

Melatonin has been proven to have antiarrhythmic potential; however, several studies have recently challenged this view. Herein, using a mouse model of obesity-induced atrial fibrillation (AF), we tentatively explored whether exogenous melatonin supplementation could increase AF susceptibility in the context of obesity. We observed that an 8-week drinking administration of melatonin (60 µg/ml in water) induced a greater susceptibility to AF in obese mice, although obesity-induced structural remodeling was alleviated. An investigation of systemic insulin sensitivity showed that melatonin treatment improved insulin sensitivity in obese mice, whereas it inhibited glucose-stimulated insulin secretion. Notably, melatonin treatment inhibited protein kinase B (Akt) signaling in the atria of obese mice and palmitate-treated neonatal rat cardiomyocytes, thereby providing an AF substrate. Melatonin increased lipid stress in obesity, as evidenced by elevated lipid accumulation and lipolysis-related gene expression, thus contributing to the impairment in atrial Akt signaling. Taken together, our results demonstrated that melatonin could increase AF susceptibility in obesity, probably due to increased lipid stress and resultant impairment of atrial Akt signaling. Our findings suggest that special precautions should be taken when administering melatonin to obese subjects.


Asunto(s)
Fibrilación Atrial , Resistencia a la Insulina , Melatonina , Ratones , Ratas , Animales , Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Melatonina/farmacología , Proteínas Proto-Oncogénicas c-akt , Ratones Obesos , Obesidad/metabolismo , Lípidos
2.
J Cell Mol Med ; 25(17): 8363-8375, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34288408

RESUMEN

Necroptosis, a novel programmed cell death, plays a critical role in the development of fibrosis, yet its role in atrial fibrillation (AF) remains elusive. Mounting evidence demonstrates that aerobic exercise improves AF-related symptoms and quality of life. Therefore, we explored the role of necroptosis in AF pathogenesis and exercise-conferred cardioprotection. A mouse AF model was established either by calcium chloride and acetylcholine (CaCl2 -Ach) administration for 3 weeks or high-fat diet (HFD) feeding for 12 weeks, whereas swim training was conducted 60 min/day, for 3-week duration. AF susceptibility, heart morphology and function and atrial fibrosis were assessed by electrophysiological examinations, echocardiography and Masson's trichrome staining, respectively. Both CaCl2 -Ach administration and HFD feeding significantly enhanced AF susceptibility (including frequency and duration of episodes), left atrial enlargement and fibrosis. Moreover, protein levels of necroptotic signaling (receptor-interacting protein kinase 1, receptor-interacting protein kinase 3, mixed lineage kinase domain-like protein and calcium/calmodulin-dependent protein kinase II or their phosphorylated forms) were markedly elevated in the atria of AF mice. However, inhibiting necroptosis with necrostatin-1 partly attenuated CaCl2 -Ach (or HFD)-induced fibrosis and AF susceptibility, implicating necroptosis as contributing to AF pathogenesis. Finally, we found 3-week swim training inhibited necroptotic signaling, consequently decreasing CaCl2 -Ach-induced AF susceptibility and atrial structural remodeling. Our findings identify necroptosis as a novel mechanism in AF pathogenesis and highlight that aerobic exercise may confer benefits on AF via inhibiting cardiac necroptosis.


Asunto(s)
Fibrilación Atrial/fisiopatología , Remodelación Atrial , Necroptosis , Condicionamiento Físico Animal , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
3.
Biochem Biophys Res Commun ; 558: 126-133, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33915326

RESUMEN

Myocardial ischemia/reperfusion (I/R) injury is a major determinant of morbidity and mortality in patients undergoing treatment for cardiac disease. A variety of treatments are reported to have benefits against reperfusion injury, yet their cardioprotective effects seem to be diminished in obesity, and the underlying mechanism remains elusive. In this study, we found that db/db mice exhibit cardiac hyper-O-GlcNAcylation. In parallel, palmitate treatment (200 mM; 12 h) in H9c2 cells showed an increase in global protein O-GlcNAcylation, along with an impaired insulin response against reperfusion injury. To investigate whether O-GlcNAcylation underlies this phenomenon, glucosamine was used to increase global protein O-GlcNAc levels. Interestingly, histological staining, electrophysiological studies, serum cardiac markers and oxidative stress biomarker assays showed that preischemic treatment with glucosamine attenuated insulin cardioprotection against myocardial infarction, arrhythmia and oxidative stress. Mechanistically, glucosamine treatment decreased insulin-stimulated Akt phosphorylation, a key modulator of cell survival. Furthermore, inhibition of O-GlcNAcylation via 6-diazo-5-oxo-l-norleucine (DON) apparently increased insulin-induced Akt phosphorylation and restored its cardioprotective response against reperfusion injury in palmitate-induced insulin-resistant H9c2 cells. Our findings demonstrated that obesity-induced hyper-O-GlcNAcylation might contribute to the attenuation of insulin cardioprotection against I/R injury.


Asunto(s)
Acetilglucosamina/metabolismo , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/metabolismo , Insulina/metabolismo , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/metabolismo , Obesidad/complicaciones , Obesidad/metabolismo , Animales , Cardiotónicos/metabolismo , Hipoxia de la Célula , Línea Celular , Diazooxonorleucina/farmacología , Modelos Animales de Enfermedad , Glicosilación/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas
4.
J Mol Cell Cardiol ; 75: 131-40, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25068621

RESUMEN

Hearts of diabetic individuals are susceptible to ischemia/reperfusion (I/R) injury. The RNA-binding protein Quaking (QKI) is known to link intracellular signaling to cellular survival and QKI dysregulation may contribute to human diseases. However, the function of QKI in diabetic hearts remains unknown. The current study attempted to identify new molecular mechanisms that potentially contribute to the susceptibility to ischemic injury in diabetic myocardium. Diabetic ob/ob mice or wild-type C57BL/6J mice were subjected to in vivo myocardial I/R. Myocardial infarct size and apoptosis, QKI5 and FoxO1 expression, nitrosative stress (NS) and ER stress were compared. Knockdown of FoxO1 was obtained by intramyocardial injection of FoxO1 specific small interfering RNA (siRNA, 20µg), and upregulation of QKI5 was acquired by injecting adenovirus encoding-QKI5. Obvious NS stress was observed in the myocardium of ob/ob mice represented by elevated iNOS expression, total NO content and nitrotyrosine content. Administration of 1400W or M40401 partly reduced the caspase-3 activity in ob/ob myocardium encountering I/R (P<0.05). Higher ER stress was also observed represented by increased p-PERK, p-eIF2α and expression of CHOP in ob/ob myocardium. ER stress inhibitor did not affect the excessive NS stress, but partially reduced I/R-induced caspase-3 activity in ob/ob hearts (P<0.05). FoxO1 was overactivated in ob/ob myocardium, and knockdown of FoxO1 attenuated both levels of NS stress and ER stress (P<0.05). Furthermore, QKI5 expression was deficient in ob/ob myocardium. Upregulation of QKI5 diminished FoxO1 expression together with NS and ER stress in ob/ob myocardium, further reducing MI/R injury. Finally, QKI5 overexpression destabilized FoxO1 mRNA in cardiomyocytes. These results suggested that QKI5 deficiency contributed to the overactivation of FoxO1 in ob/ob animals and subsequently magnified nitrosative stress and ER stress, which enhances the ischemic intolerance of diabetic hearts.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Estrés del Retículo Endoplásmico , Factores de Transcripción Forkhead/metabolismo , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Proteínas de Unión al ARN/metabolismo , Animales , Apoptosis , Diabetes Mellitus Experimental/metabolismo , Regulación hacia Abajo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Isquemia Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/patología , Nitrosación , Estabilidad Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba
5.
Front Pharmacol ; 12: 771940, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899326

RESUMEN

Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia in clinical setting. Its pathogenesis was associated with metabolic disorder, especially defective fatty acids oxidation (FAO). However, whether promoting FAO could prevent AF occurrence and development remains elusive. In this study, we established a mouse model of obesity-related AF through high-fat diet (HFD) feeding, and used l-carnitine (LCA, 150 mg/kg⋅BW/d), an endogenous cofactor of carnitine palmitoyl-transferase-1B (CPT1B; the rate-limiting enzyme of FAO) to investigate whether FAO promotion can attenuate the AF susceptibility in obesity. All mice underwent electrophysiological assessment for atrial vulnerability, and echocardiography, histology and molecular evaluation for AF substrates and underlying mechanisms, which were further validated by pharmacological experiments in vitro. HFD-induced obese mice increased AF vulnerability and exhibited apparent atrial structural remodeling, including left atrial dilation, cardiomyocyte hypertrophy, connexin-43 remodeling and fibrosis. Pathologically, HFD apparently leads to defective cardiac FAO and subsequent lipotoxicity, thereby evoking a set of pathological reactions including oxidative stress, DNA damage, inflammation, and insulin resistance. Enhancing FAO via LCA attenuated lipotoxicity and lipotoxicity-induced pathological changes in the atria of obese mice, resulting in restored structural remodeling and ameliorated AF susceptibility. Mechanistically, LCA activated AMPK/PGC1α signaling both in vivo and in vitro, and pharmacological inhibition of AMPK via Compound C attenuated LCA-induced cardio-protection in palmitate-treated primary atrial cardiomyocytes. Taken together, our results demonstrated that FAO promotion via LCA attenuated obesity-mediated AF and structural remodeling by activating AMPK signaling and alleviating atrial lipotoxicity. Thus, enhancing FAO may be a potential therapeutic target for AF.

6.
Cell Death Dis ; 12(9): 813, 2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34453039

RESUMEN

Atrial fibrillation (AF) is an increasingly prevalent arrhythmia with significant health and socioeconomic impact. The underlying mechanism of AF is still not well understood. In this study, we sought to identify hub genes involved in AF, and explored their functions and underlying mechanisms based on bioinformatics analysis. Five microarray datasets in GEO were used to identify the differentially expressed genes (DEGs) by Robust Rank Aggregation (RRA), and hub genes were screened out using protein-protein interaction (PPI) network. AF model was established using a mixture of acetylcholine and calcium chloride (Ach-CaCl2) by tail vein injection. We totally got 35 robust DEGs that mainly involve in extracellular matrix formation, leukocyte transendothelial migration, and chemokine signaling pathway. Among these DEGs, we identified three hub genes involved in AF, of which CXCL12/CXCR4 axis significantly upregulated in AF patients stands out as one of the most potent targets for AF prevention, and its effect on AF pathogenesis and underlying mechanisms were investigated in vivo subsequently with the specific CXCR4 antagonist AMD3100 (6 mg/kg). Our results demonstrated an elevated transcription and translation of CXCL12/CXCR4 axis in AF patients and mice, accompanied with the anabatic atrial inflammation and fibrosis, thereby providing the substrate for AF maintenance. Blocking its signaling via AMD3100 administration in AF model mice reduced AF inducibility and duration, partly ascribed to decreased atrial inflammation and structural remodeling. Mechanistically, these effects were achieved by reducing the recruitment of CD3+ T lymphocytes and F4/80+ macrophages, and suppressing the hyperactivation of ERK1/2 and AKT/mTOR signaling in atria of AF model mice. In conclusion, this study provides new evidence that antagonizing CXCR4 prevents the development of AF, and suggests that CXCL12/CXCR4 axis may be a potential therapeutic target for AF.


Asunto(s)
Fibrilación Atrial/metabolismo , Quimiocina CXCL12/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Animales , Fibrilación Atrial/diagnóstico por imagen , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Bencilaminas/administración & dosificación , Bencilaminas/farmacología , Estudios de Casos y Controles , Biología Computacional , Ciclamas/administración & dosificación , Ciclamas/farmacología , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Electrocardiografía , Fibrosis , Perfilación de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/patología , Atrios Cardíacos/fisiopatología , Humanos , Inflamación/patología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Remodelación Vascular/efectos de los fármacos
7.
Chin Med J (Engl) ; 132(9): 1015-1022, 2019 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-30925549

RESUMEN

BACKGROUND: Large-nerve fiber dysfunction, as assessed by vibration perception threshold (VPT) predicts risks of ulceration, amputation, and mortality in diabetes. Serum uric acid (UA) is closely associated with various metabolic disorders, especially diabetes. Thus, we sought to investigate the clinical relevance of UA to large-nerve fiber dysfunction, among patients with type 2 diabetes (T2D). METHODS: Medical records of consecutive patients with T2D who were admitted to Beijing Friendship Hospital Pinggu Campus between May 2014 and December 2016 were collected. Data for the 824 eligible patients included in the final analysis were extracted using a structured form. A VPT value ≥15 in either foot was defined as abnormal. We compared the clinical characteristics between patients with abnormal VPT and those with normal VPT (VPT value <15 in both feet) in the overall population and in gender subgroups. Logistic regression analysis was performed to explore the association of abnormal VPT with UA level. One-way analysis of variance was used to compare VPT values across four UA quartiles. RESULTS: UA levels were significantly lower in T2D patients with abnormal VPT than in those with normal VPT (294.5 ±â€Š84.0 vs. 314.9 ±â€Š92.8 µmol/L, P < 0.01), especially among male patients (311.7 ±â€Š85.2 vs. 336.9 ±â€Š89.6 µmol/L, P < 0.01). From the logistic regression analysis, hyperuricemia (males >420 µmol/L; females >360 µmol/L) was associated with a reduced risk of abnormal VPT (odds ratio [OR], 0.60; 95% confidence interval [CI], 0.39-0.91; P < 0.05). This association was robust in male patients (OR, 0.43; 95% CI, 0.24-0.76; P < 0.01) but not in female patients (OR, 0.92; 95% CI, 0.47-1.82; P = 0.816), even after adjustment for confounding factors. For the younger male subgroup (age <65 years), VPT values decreased as the UA level increased (P for trend = 0.002), but this trend was not significant in older male subgroup (age ≥65 years; P for trend = 0.400). CONCLUSIONS: Low serum UA levels showed a significant association with an increased risk of large-nerve fiber dysfunction in male patients with T2D, but not in female patients with T2D. In addition, in only the younger subgroup of male patients (<65 years), lower levels of UA also correlated with higher VPT values.


Asunto(s)
Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/patología , Fibras Nerviosas/patología , Ácido Úrico/sangre , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedades del Sistema Nervioso Periférico/sangre , Enfermedades del Sistema Nervioso Periférico/patología , Adulto Joven
8.
Protein Pept Lett ; 18(4): 403-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21121889

RESUMEN

Neuropeptide FF (NPFF) belongs to an opioid-modulating peptide family. NPFF has been reported to play important roles in the control of pain and analgesia through interactions with the opioid system. However, very few studies examined the effect of supraspinal NPFF system on analgesia induced by opiates administered at the peripheral level. In the present study, intracerebroventricular (i.c.v.) injection of NPFF (1, 3 and 10 nmol) dose-dependently inhibited systemic morphine (0.12 mg, i.p.) analgesia in the mouse tail flick test. Similarly, i.c.v. administration of dNPA and NPVF, two agonists highly selective for NPFF(2) and NPFF(1) receptors, respectively, decreased analgesia induced by i.p. morphine in mice. Furthermore, these anti-opioid activities of NPFF and related peptides were blocked by pretreatment with the NPFF receptors selective antagonist RF9 (10 nmol, i.c.v.). These results demonstrate that activation of central NPFF(1) and NPFF(2) receptors has the similar anti-opioid actions on the antinociceptive effect of systemic morphine.


Asunto(s)
Analgésicos Opioides/antagonistas & inhibidores , Morfina/antagonistas & inhibidores , Antagonistas de Narcóticos/administración & dosificación , Oligopéptidos/administración & dosificación , Analgesia , Animales , Relación Dosis-Respuesta a Droga , Antagonismo de Drogas , Masculino , Ratones , Ratones Endogámicos , Morfina/administración & dosificación , Neuropéptidos/administración & dosificación , Dolor/tratamiento farmacológico , Receptores de Neuropéptido/agonistas , Receptores de Neuropéptido/antagonistas & inhibidores
9.
Peptides ; 31(4): 683-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19922753

RESUMEN

Neuropeptide FF (NPFF) belongs to a neuropeptide family including two precursors (pro-NPFF(A) and pro-NPFF(B)) and two receptors (NPFF(1) and NPFF(2)). NPFF and NPFF receptor mRNAs have been reported to be highly expressed and localized in the rat and human spinal cord. In the present study, the i.t. action of NPFF system on blood pressure and heart rate were examined using NPFF and two related agonists, NPVF and dNPA, which exhibit highest selectivities for NPFF(1) and NPFF(2) receptors, respectively. In urethane-anesthetized rats, NPFF and related peptides (5-40 nmol, i.t.) produced significant pressor and tachycardic responses at the spinal cord level. These effects were dose-dependent and similar with respect to time-course for the three peptides. Furthermore, i.t. injection of RF9 (20 nmol), a selective NPFF antagonist, significantly antagonized the cardiovascular responses to 20 nmol NPFF and related peptides (i.t.). Moreover, pretreatment of the rats with alpha-adrenoceptor antagonist phentolamine (1mg/kg, i.v.) significantly reduced the pressor effects of NPFF. Nevertheless, pretreatment with muscarinic receptor and adrenoceptor antagonists (i.v.) could block the tachycardic effects induced by NPFF. Collectively, our results suggested that i.t. administration of NPFF and related peptides increased MAP and HR which were possibly mediated by the activation of both NPFF(1) and NPFF(2) receptors in the rat spinal cord. In addition, our results showed that the muscarinic receptor and adrenoceptor participated in the tachycardic response to i.t. NPFF, while alpha-adrenoceptor played an important role in the regulation of pressor effect of NPFF.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Antagonistas de Narcóticos , Oligopéptidos , Antagonistas Adrenérgicos alfa/farmacología , Animales , Antiarrítmicos , Antihipertensivos/farmacología , Atropina/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Inyecciones Espinales , Masculino , Antagonistas Muscarínicos/farmacología , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/farmacología , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacología , Fentolamina/farmacología , Propranolol/farmacología , Ratas , Ratas Wistar
10.
Eur J Pharmacol ; 621(1-3): 61-6, 2009 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-19747477

RESUMEN

26RFa, a novel RFamide-related peptide, has been identified as the endogenous ligand for GPR103, and the biological functions of this neuropeptide have not been well investigated. In the present study, the cardiovascular effects of intravenous administration of rat/mouse 26RFa were tested in rats. Intravenous administered 26RFa (100-800 nmol/kg, i.v.) caused a biphasic change in blood pressure, and an increase in heart rate in urethane-anaesthetised rats. The pressor effects induced by 26RFa were significantly inhibited by pretreatment with the alpha- and beta-adrenoreceptor antagonists. In contrast, the tachycardiac responses to 26RFa were significantly attenuated by bilateral cervical vagotomy and beta-adrenoreceptor antagonist propranolol. These data imply that the peripheral cardiovascular regulation of 26RFa might be involved in vagal components and catecholaminergic pathway. Furthermore, in order to evaluate the importance of the regions of 26RFa molecule in its cardiovascular regulation, the two C-terminal fragments of rat/mouse 26RFa, 26RFa(8-26) and 26RFa(19-26), were synthesized and investigated to address their peripheral cardiovascular responses in rats. Surprisingly, intravenous injections of 26RFa(8-26) and 26RFa(19-26) (50-300 nmol/kg, i.v.) produced dose-dependent increases in blood pressure and heart rate, which exerted different sensitivities to bilateral vagotomy and beta-adrenergic receptor antagonist. The results indicate that intravenous administrations of 26RFa and its fragments induced their cardiovascular effects via different pathways, which further suggest that the N-terminal residues of 26RFa are required for its cardiovascular activities.


Asunto(s)
Sistema Cardiovascular/efectos de los fármacos , Neuropéptidos/administración & dosificación , Neuropéptidos/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Antagonistas Adrenérgicos/farmacología , Anestesia , Animales , Presión Sanguínea/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Inyecciones Intravenosas , Ligandos , Masculino , Neuropéptidos/química , Neuropéptidos/metabolismo , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Ratas , Ratas Wistar , Uretano , Vagotomía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA