Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Redox Biol ; 73: 103184, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38718533

RESUMEN

RATIONALE: The disruption of the balance between fatty acid (FA) uptake and oxidation (FAO) leads to cardiac lipotoxicity, serving as the driving force behind diabetic cardiomyopathy (DbCM). Sirtuin 5 (Sirt5), a lysine de-succinylase, could impact diverse metabolic pathways, including FA metabolism. Nevertheless, the precise roles of Sirt5 in cardiac lipotoxicity and DbCM remain unknown. OBJECTIVE: This study aims to elucidate the role and underlying mechanism of Sirt5 in the context of cardiac lipotoxicity and DbCM. METHODS AND RESULTS: The expression of myocardial Sirt5 was found to be modestly elevated in diabetic heart failure patients and mice. Cardiac dysfunction, hypertrophy and lipotoxicity were exacerbated by ablation of Sirt5 but improved by forced expression of Sirt5 in diabetic mice. Notably, Sirt5 deficiency impaired FAO without affecting the capacity of FA uptake in the diabetic heart, leading to accumulation of FA intermediate metabolites, which mainly included medium- and long-chain fatty acyl-carnitines. Mechanistically, succinylomics analyses identified carnitine palmitoyltransferase 2 (CPT2), a crucial enzyme involved in the reconversion of fatty acyl-carnitines to fatty acyl-CoA and facilitating FAO, as the functional succinylated substrate mediator of Sirt5. Succinylation of Lys424 in CPT2 was significantly increased by Sirt5 deficiency, leading to the inactivation of its enzymatic activity and the subsequent accumulation of fatty acyl-carnitines. CPT2 K424R mutation, which mitigated succinylation modification, counteracted the reduction of enzymatic activity in CPT2 mediated by Sirt5 deficiency, thereby attenuating Sirt5 knockout-induced FAO impairment and lipid deposition. CONCLUSIONS: Sirt5 deficiency impairs FAO, leading to cardiac lipotoxicity in the diabetic heart through the succinylation of Lys424 in CPT2. This underscores the potential roles of Sirt5 and CPT2 as therapeutic targets for addressing DbCM.


Asunto(s)
Carnitina O-Palmitoiltransferasa , Cardiomiopatías Diabéticas , Ácidos Grasos , Metabolismo de los Lípidos , Miocitos Cardíacos , Sirtuinas , Animales , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/patología , Carnitina O-Palmitoiltransferasa/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Sirtuinas/metabolismo , Sirtuinas/genética , Ratones , Ácidos Grasos/metabolismo , Miocitos Cardíacos/metabolismo , Humanos , Masculino , Oxidación-Reducción , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/complicaciones
2.
Int J Med Sci ; 21(4): 714-724, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38464839

RESUMEN

Sepsis-induced cardiomyopathy (SIC) represents a severe complication of systemic infection, characterized by significant cardiac dysfunction. This study examines the role of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) and Inverted Formin 2 (INF2) in the pathogenesis of SIC, focusing on their impact on mitochondrial homeostasis and dynamics. Our research demonstrates that silencing DNA-PKcs alleviates lipopolysaccharide (LPS)-induced cardiomyocyte death and dysfunction. Using HL-1 cardiomyocytes treated with LPS, we observed that DNA-PKcs knockdown notably reverses LPS-induced cytotoxicity, indicating a protective role against cellular damage. This effect is further substantiated by the reduction in caspase-3 and caspase-9 activation, key markers of apoptosis, upon DNA-PKcs knockdown. Besides, our data further reveal that DNA-PKcs knockdown attenuates LPS-induced mitochondrial dysfunction, evidenced by improved ATP production, enhanced activities of mitochondrial respiratory complexes, and preserved mitochondrial membrane potential. Moreover, DNA-PKcs deletion counteracts LPS-induced shifts towards mitochondrial fission, indicating its regulatory influence on mitochondrial dynamics. Conclusively, our research elucidates the intricate interplay between DNA-PKcs and INF2 in the modulation of mitochondrial function and dynamics during sepsis-induced cardiomyopathy. These findings offer new insights into the molecular mechanisms underpinning SIC and suggest potential therapeutic targets for mitigating mitochondrial dysfunction in this critical condition.


Asunto(s)
Cardiomiopatías , Enfermedades Mitocondriales , Sepsis , Humanos , Proteína Quinasa Activada por ADN/metabolismo , Dinámicas Mitocondriales , Lipopolisacáridos/toxicidad , Lipopolisacáridos/metabolismo , Dominio Catalítico , Cardiomiopatías/genética , Miocitos Cardíacos , Sepsis/complicaciones , Sepsis/genética , Enfermedades Mitocondriales/patología , ADN/efectos adversos , ADN/metabolismo
3.
Exp Ther Med ; 22(2): 815, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34131438

RESUMEN

Numerous previous studies have found that C-reactive protein (CRP) is associated with cardiac arrhythmia and cardiac remodeling. However, the underlying mechanisms of this association remain unclear. Sodium-calcium exchanger 1 (NCX1) serves an important role in the regulation of intracellular calcium concentration, which is closely related with cardiac arrhythmia and cardiac remodeling. The present study aimed to evaluate the effects of CRP on NCX1 and intracellular calcium concentration in cardiomyocytes. Primary neonatal mouse ventricular cardiomyocytes were cultured and treated with varying concentrations of CRP (0, 5, 10, 20 and 40 µg/ml). The cardiomyocytes were also treated with NF-κB-specific inhibitor PTDC and a specific inhibitor of the reverse NCX1 KB-R7943 before their intracellular calcium concentrations were measured. mRNA and protein expression levels of NCX1 were detected by reverse transcription-quantitative PCR and western blotting, respectively and intracellular calcium concentration was evaluated by flow cytometry. CRP treatment significantly increased mRNA and protein expression levels of NCX1 in myocytes (P=0.024), as well as intracellular calcium concentration (P=0.01). These results were significantly attenuated by the NF-κB-specific inhibitor PDTC and a specific inhibitor of the reverse NCX1, KB-R7943. CRP significantly upregulated NCX1 expression and increased intracellular calcium concentration in cardiomyocytes via the NF-κB pathway, suggesting that CRP may serve a pro-arrhythmia role via direct influence on the calcium homeostasis of cardiomyocytes.

4.
J Cell Physiol ; 236(6): 4403-4419, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33184863

RESUMEN

Prolonged endoplasmic reticulum (ER) stress is the key driving force behind diabetic cardiomyopathy (DCM). Autophagy is extensively implicated in adaptive mechanisms for cell survival. Interleukin-33 (IL-33) is known to be a potent cardiac protector, but its roles in DCM, ER stress, and autophagy are currently unknown. We aimed to explore the effects of IL-33 on DCM and characterize the roles that ER stress and autophagy play in DCM. The effects of IL-33 on DCM, ER stress, and autophagy were characterized both in db/db mice and in palmitic acid (PA)-treated cardiomyocytes. The manipulators of ER stress and autophagy were used to clarify their roles in DCM remittance conferred by IL-33. Gene expression analysis was used to identify IL-33-dependent regulators of ER stress and autophagy. Both db/db mice and PA-treated cells presented with enhanced levels of ER stress, apoptosis, and lipid deposition, as well as impaired autophagy, all of which could be reversed by IL-33. Treatment with IL-33 improved the cardiac diastolic function of diabetic mice. Nonselective autophagy inhibitors, such as 3-methyladenine (3-MA) or wortmannin, abolished the protective effects of IL-33, resulting in an increase in both ER stress and apoptosis. Strikingly, insulin-like growth factor-binding protein 3 (IGFBP3) was identified as the gene most significantly differentially expressed between IL-33 and control groups. Knockdown of IGFBP3 expression, similar to the effect of nonselective autophagy inhibitors, resulted in high levels of ER stress, impaired autophagy, and apoptosis that were not rescued upon treatment with IL-33. IL-33 abates DCM by alleviating ER stress and promoting autophagy. IGFBP3 is essential for IL-33-induced ER stress resolution and autophagic enhancement during DCM.


Asunto(s)
Autofagia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Cardiomiopatías Diabéticas/prevención & control , Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Interleucina-33/farmacología , Miocitos Cardíacos/efectos de los fármacos , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Modelos Animales de Enfermedad , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Masculino , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ácido Palmítico/toxicidad , Ratas Sprague-Dawley , Transducción de Señal
5.
J Cell Mol Med ; 23(10): 6919-6929, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31441592

RESUMEN

The present study investigated the role of long non-coding RNA (lncRNA) small nucleolar RNA host gene 16 (SNHG16) in the human aortic smooth muscle cell (HASMC) proliferation and migration and explored the potential link between SNHG16 and atherosclerosis. Our results showed that platelet-derived growth factor (PDGF)-bb treatment promoted cell proliferation and migration with concurrent up-regulation of SNHG16 in HASMCs. Small nucleolar RNA host gene 16 overexpression promoted HASMC proliferation and migration, while SNHG16 knockdown suppressed cell proliferation and migration in PDGF-bb-stimulated HASMCs. The bioinformatic analyses showed that SNHG16 possessed the complementary binding sequence with miR-205, where the interaction was confirmed by luciferase reporter assay and RNA pull-down assay in HASMCs, and SNHG16 inversely regulated miR-205 expression. MiR-205 overexpression attenuated the enhanced effects of PDGF-bb treatment on HASMC proliferation and migration. Moreover, Smad2 was targeted and inversely regulated by miR-205, while being positively regulated by SNHG16 in HASMCs. Smad2 knockdown attenuated PDGF-bb-mediated actions on HASMC proliferation and migration. Both miR-205 overexpression and Smad2 knockdown partially reversed the effects of SNHG16 overexpression on HASMC proliferation and migration. Moreover, SNHG16 and Smad2 mRNA were up-regulated, while miR-205 was down-regulated in the plasma from patients with atherosclerosis. Small nucleolar RNA host gene 16 expression was inversely correlated with miR-205 expression and positively correlated with Smad2 expression in the plasma from atherosclerotic patients. In conclusion, our data showed the up-regulation of SNHG16 in pathogenic-stimulated HASMCs and clinical samples from atherosclerotic patients. Small nucleolar RNA host gene 16 regulated HASMC proliferation and migration possibly via regulating Smad2 expression by acting as a competing endogenous RNA for miR-205.


Asunto(s)
Aorta/citología , Movimiento Celular , MicroARNs/genética , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , ARN Largo no Codificante/metabolismo , Proteína Smad2/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Secuencia de Bases , Becaplermina/farmacología , Línea Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , MicroARNs/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , ARN Largo no Codificante/genética , Regulación hacia Arriba/efectos de los fármacos
6.
Cell Prolif ; 51(6): e12517, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30156340

RESUMEN

OBJECTIVES: Yes-associated protein (YAP) has been reported to regulate cell proliferation and differentiation. We aimed to characterize the role of YAP in angiotensin II (Ang II)-induced hypertensive vascular remodelling (HVR) and vascular smooth muscle cells (VSMCs) phenotypic modulation and to explore the underlying mechanisms. MATERIALS AND METHODS: An HVR rat model was established by continuous Ang II infusion for 2 weeks. Western blotting, qRT-PCR, and confocal microscopy were conducted to assess YAP expression. YAP-shRNA interfering plasmid and adenovirus were constructed to knock down YAP. We used cell proliferation and migration assays, accompanied by pathway inhibitors, to evaluate the biological function and underlying mechanisms. RESULTS: Ang II upregulated YAP expression in the media of carotid artery; however, in vivo YAP silencing significantly mitigated HVR, independent of the blood pressure level. Ang II upregulated YAP expression and promoted YAP nuclear accumulation in a dose- and time-dependent manner in rat VSMCs. YAP knockdown ameliorated Ang II-induced VSMCs phenotypic modulation. The regulation of YAP by Ang II could be blocked by pretreatment with angiotensin receptor type 1 antagonist losartan or F-actin depolymerizing agent latrunculin B but not the AT2R antagonist PD 123319. Disrupting the YAP-TEA domain (TEAD) interaction with verteporfin inhibited Ang II-induced VSMCs phenotypic modulation. CONCLUSIONS: Yes-associated protein mediated angiotensin II-induced VSMCs phenotypic modulation and vascular remodelling. YAP is a potential therapeutic target for HVR beyond blood pressure control.


Asunto(s)
Angiotensina II/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Imidazoles/farmacología , Músculo Liso Vascular/efectos de los fármacos , Piridinas/farmacología , Remodelación Vascular/efectos de los fármacos , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Masculino , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteínas Señalizadoras YAP
7.
Int Heart J ; 58(6): 908-914, 2017 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-29151481

RESUMEN

Thyroid dysfunction is prevalent in patients with heart failure (HF) and hypothyroidism is related to the adverse prognosis of HF subjects receiving cardiac resynchronization therapy (CRT). We aim to investigate whether low-normal free triiodothyronine (fT3) level is related to CRT response and the prognosis of euthyroid patients with HF after CRT implantation.One hundred and thirteen euthyroid patients who received CRT therapy without previous thyroid disease and any treatment affecting thyroid hormones were enrolled. All of patients were evaluated for cardiac function and thyroid hormones (serum levels of fT3, free thyroxine [fT4] and thyroid-stimulating hormone [TSH]). The end points were overall mortality and hospitalization for HF worsening. During a follow-up period of 39 ± 3 weeks, 36 patients (31.9%) died and 45 patients (39.8%) had hospitalization for HF exacerbation. A higher rate of NYHA III/IV class and a lower fT3 level were both observed in death group and HF event group. Multivariate Cox regression analyses disclosed that a lower-normal fT3 level (HR = 0.648, P = 0.009) and CRT response (HR = 0.441, P = 0.001) were both independent predictors of overall mortality. In addition, they were also both related to HF re-hospitalization event (P < 0.01 for both). Patients with fT3 < 3.00 pmol/L had a significantly higher overall mortality than those with fT3 ≥ 3.00 pmol/L (P = 0.027). Meanwhile, a higher HF hospitalization event rate was also found in patients with fT3 < 3.00 pmol/L (P < 0.001).A lower-normal fT3 level is correlated with a worse cardiac function an adverse prognosis in euthyroid patients with HF after CRT implantation.


Asunto(s)
Terapia de Resincronización Cardíaca , Insuficiencia Cardíaca/terapia , Triyodotironina/sangre , Anciano , Femenino , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/diagnóstico , Humanos , Masculino , Persona de Mediana Edad , Pronóstico
8.
J Investig Med ; 65(5): 899-911, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28330835

RESUMEN

Telemedicine interventions may be associated with reductions in hospital admission rate and mortality in patients with heart failure (HF). The present study is an updated analysis (as of June 30, 2016) of randomized controlled trials, where patients with HF underwent telemedicine care or the usual standard care. Data were extracted from 39 eligible studies for all-cause and HF-related hospital admission rate, length of stay, and mortality. The overall all-cause mortality (pooled OR=0.80, 95% CI 0.71 to 0.91, p<0.001), HF-related admission rate (pooled OR=0.63, 95% CI 0.53 to 0.76, p<0.001), and HF-related length of stay (pooled standardized difference in means=-0.37, 95% CI -0.72 to -0.02, p=0.041) were significantly lower in the telemedicine group (teletransmission and telephone-supported care), as compared with the control group. In subgroup analysis, all-cause mortality (pooled OR=0.69, 95% CI 0.56 to 0.86, p=0.001), HF-related admission rate (OR=0.61, 95% CI 0.42 to 0.88, p=0.008), HF-related length of stay (pooled standardized difference in means=-0.96, 95% CI -1.88 to -0.05, p=0.039) and HF-related mortality (OR=0.68, 95% CI 0.54 to 0.85, p=0.001) were significantly lower in the teletransmission group, as opposed to the standard care group, whereas only HF-related admission rate (OR=0.64, 95% CI 0.52 to 0.79, p<0.001) was lower in the telephone-supported care group. Overall, telemedicine was shown to be beneficial, with home-based teletransmission effectively reducing all-cause mortality and HF-related hospital admission, length of stay and mortality in patients with HF.


Asunto(s)
Insuficiencia Cardíaca/mortalidad , Insuficiencia Cardíaca/terapia , Telemedicina/métodos , Estudios de Casos y Controles , Enfermedad Crónica , Hospitalización , Humanos , Tiempo de Internación , Evaluación de Resultado en la Atención de Salud , Admisión del Paciente , Readmisión del Paciente , Calidad de Vida , Resultado del Tratamiento
9.
Life Sci ; 167: 12-21, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27746188

RESUMEN

AIMS: Adverse cardiovascular effects induced by peroxisome proliferator activator receptor-γ (PPAR-γ) activation were observed in clinical setting. But the underlying mechanism is unclear. Now, transgenic mice with cardiac specific peroxisome proliferator activator receptor-γ overexpression (TG-PPAR-γ) were used to explore the possible mechanisms. MATERIALS AND METHODS: Cardiac tissues from TG-PPAR-γ mice, a PPAR-γ over-expressing human cardiomyocyte line AC16 cell, and PPAR-γ agonist-treated primary cardiomyocytes were used to evaluate the expression of cardiac calcium regulatory proteins as sarcoplasmic reticulum Ca2+ ATPase, Na+/Ca2+ exchanger 1, ryanodine receptor 2 and phospholamban. Intracellular Ca2+ levels were also examined by flow cytometry and confocal microscopy with Fluo-4/AM in these cells. KEY FINDINGS: In this study, frequent ventricular premature contraction and polymorphic ventricular tachycardia were observed in TG-PPAR-γ but not in wild-type mice. Besides, we found the calcium regulatory proteins expression were higher in the TG-PPAR-γ mice, PPAR-γ overexpressing human cardiomyocyte line AC16 cell and PPAR-γ agonist-treated primary cardiomyocytes than the control group respectively. In addition, an increase of intracellular calcium levels and CaMKII δ expression in PPAR-γ overexpression and PPAR-γ activation group. Moreover, Inhibition of CaMKII δ could improve the intracellular calcium levels and reduce the occurrence of ventricular arrhythmia. SIGNIFICANCE: PPAR-γ over-expression perturbs the intracellular calcium homeostasis in cardiomyocytes which contribute to the ventricular arrhythmias and cardiac sudden death in TG-PPAR-γ mice.


Asunto(s)
Arritmias Cardíacas/genética , Calcio/metabolismo , Ventrículos Cardíacos/patología , Miocitos Cardíacos/patología , PPAR gamma/genética , Regulación hacia Arriba , Animales , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , ATPasas Transportadoras de Calcio/genética , Línea Celular , Células Cultivadas , Regulación de la Expresión Génica , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Homeostasis , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/metabolismo
10.
Medicine (Baltimore) ; 95(6): e2602, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26871780

RESUMEN

The interactions of sodium balance and response of renin-angiotensin-aldosterone system are important for maintaining the hemodynamic stability in physiological conditions. However, the influence of short-term sodium intake intervention in the response of renin-angiotensin system (RAS) on hypertensive patients is still unclear. Thus, we conducted a clinical trial to investigate the effects of short-term sodium intake intervention on the response of RAS in hypertensive patients.One hundred twenty-five primary Chinese hypertensive patients were divided into high, moderate, and low sodium groups by 24-hour urinary sodium excretion (UNa). All the patients received a 10-day dietary sodium intake intervention with standardized sodium (173.91mmol/day) and potassium (61.53mmol/day). Blood pressure, urinary sodium, urinary potassium, plasma sodium, potassium, creatinine, the levels of plasma renin activity, plasma angiotensin II concentrations (AT-II), and plasma aldosterone concentrations were detected before and after the intervention.Before the intervention, no differences were found in blood pressure and RAS among 3 groups. After standardized dietary sodium intake intervention, both UNa excretion and systolic pressure decreased in high-sodium group, while they increased in moderate and low-sodium groups. Intriguingly, there were no changes in the levels of plasma renin activity, AT-II, and plasma aldosterone concentrations among 3 groups during the intervention.The present study demonstrated that the influenced sodium excretion and blood pressure by short-term sodium intake intervention were independent of RAS quick response in Chinese hypertensive patients.


Asunto(s)
Dieta Hiposódica , Hipertensión/dietoterapia , Sistema Renina-Angiotensina/fisiología , Adulto , Anciano , Aldosterona/sangre , Angiotensina II/sangre , Biomarcadores/sangre , Determinación de la Presión Sanguínea , China , Femenino , Humanos , Hipertensión/sangre , Hipertensión/diagnóstico , Hipertensión/fisiopatología , Masculino , Persona de Mediana Edad , Renina/sangre , Resultado del Tratamiento
11.
Heart Vessels ; 31(2): 230-40, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25491934

RESUMEN

Neovascularization plays pivotal role in ischemic heart failure; however, it is unclear in non-ischemic heart failure. Non-ischemic heart failure was induced by chronic rapid right ventricular pacing at 200 beats/min, respectively, for 3 and 6 weeks in 12 dogs. Sham-operation was performed in another 6 dogs as control. Three-week tachycardia pacing could induce mild/moderate heart failure and 6-week pacing could induce severe heart failure. Pan-microvessel density (MVD) was assessed by CD31 and neovascularization density was assessed by CD105. Mean CD31-MVD and CD105-MVD were significantly increased after 3-week pacing. However, CD105-MVD was significantly decreased by 80 % in 6-week pacing group compared with 3-week pacing group, whereas CD31-MVD was only decreased slightly (15 %; P < 0.05). Myocardial proangiogenic factor stromal cell-derived factor 1 (SDF-1), hypoxia-inducible factors 1α (HIF-1α, a transcription factor which could regulate SDF-1 expression), serum SDF-1 levels and circulating EPC mobilization were greatly elevated after 3-week pacing but nearly returned to baseline level after 6-week pacing, which were in accordance with the changes of neovascularization levels assessed by CD105. Angiogenesis and migrating ability of EPCs were enhanced after stimulation of SDF-1, which could be abolished by pretreatment with SDF-1 receptor antagonist AMD3100. In addition, angiogenesis and migrating functions of EPCs were significantly enhanced by the serum from 3-week pacing dogs, but had much weaker response to the serum from 6-week pacing dogs. In conclusion, tachycardia pacing-induced non-ischemic heart failure, promoted myocardial neovascularization and mobilized circulating EPCs, which might be mediated partly through SDF-1 pathway.


Asunto(s)
Estimulación Cardíaca Artificial , Movimiento Celular , Quimiocina CXCL12/metabolismo , Vasos Coronarios/metabolismo , Células Progenitoras Endoteliales/metabolismo , Insuficiencia Cardíaca/etiología , Neovascularización Fisiológica , Transducción de Señal , Taquicardia Ventricular/complicaciones , Animales , Células Cultivadas , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Perros , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Factores de Tiempo
12.
Am J Transl Res ; 7(5): 922-31, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26175853

RESUMEN

Several studies have found that C-reactive protein (CRP) was associated with QTc interval prolongation and ventricular arrhythmia. However, little is known about the mechanisms involved. K(+) channel interaction protein 2 (KChIP2) is a necessary subunit for the formation of transient outward potassium current (Ito.f) which plays a critical role in early repolarization and QTc interval of heart. In this study, we aimed to evaluate the effects of CRP on KChIP2 and Ito.f in cardiomyocytes and to explore the potential mechanism. The neonatal mice ventricular cardiomyocytes were cultured and treated with CRP at different concentrations. The expression of KChIP2 was detected by real time quantitative PCR and Western blot. In addition, Ito.f current density was evaluated by whole cell patch clamp techniques. Our results showed that CRP significantly decreased the mRNA and protein expression of KChIP2 in time and doses dependent manners (P < 0.05), and also reduced the current density of Ito.f (P < 0.05). In addition, CRP increased the expression of NF-κB and decreased IκBα expression without significant influence on the expression of ERK1/2 and JNK. Meanwhile, the NF-κB inhibitor PDTC significantly attenuated the effects of CRP on KChIP2 and Ito.f current density. In conclusion, CRP could significantly down-regulate KChIP2 expression and reduce current density of Ito.f partly through NF-κB pathway, suggesting that CRP may directly or indirectly influence QTc interval and arrhythmia via influencing KChIP2 expression and Ito.f current density of cardiomyocytes.

13.
Int J Cardiovasc Imaging ; 31(2): 329-37, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25392055

RESUMEN

Left ventricular noncompaction (LVNC) is a rare cardiomyopathy with high incidence of heart failure (HF). It is unclear whether LVNC patients with desynchronized HF would benefit from cardiac resynchronization therapy (CRT). In order to evaluate the effect of CRT on LVNC, this study explored left ventricular (LV) remodeling and mechanical synchronicity before and after CRT in LVNC patients, and compare with that in idiopathic dilated cardiomyopathy (DCM) patients. We collected 15 LVNC and 30 matched DCM patients. All the patients underwent clinical evaluation,electrocardiogram and echocardiography before CRT and ≥6 months later. LV response was defined as ≥15 % decrease in LV end-systolic volume (LVESV). Longitudinal synchronicity was quantified by YU-index using tissue Doppler imaging. The time delay of peak radial strain from anteroseptal to posterior wall, which derived from speckle tracking imaging, was used to quantify radial synchronicity. In LVNC group, LV ejection fraction increased from 27.6 ± 5.5 to 39.1 ± 7.0 % (P < 0.01) during follow-up, but LV volumes did not change significantly (both P > 0.05). Five LVNC patients (33.3 %) responded to CRT, and all of them were super-responders (reduction in LVESV > 30 %). In addition, the number of noncompacted segments and the thickness ratio of noncompacted to compacted myocardium decreased (both P < 0.05). Inter-ventricular, longitudinal and radial intra-ventricular dyssynchrony also reduced significantly (all P < 0.05). Compared with DCM group, there was no significant difference in LV response rate (33.3 vs. 60.0 %, P = 0.092), improvement of LV function and dyssynchrony index (all P < 0.05). In conclusion, CRT improved heart function, morphology and mechanical dyssynchrony in LVNC patients.


Asunto(s)
Terapia de Resincronización Cardíaca , Insuficiencia Cardíaca/terapia , No Compactación Aislada del Miocardio Ventricular/terapia , Función Ventricular Izquierda , Remodelación Ventricular , Anciano , Dispositivos de Terapia de Resincronización Cardíaca , Cardiomiopatía Dilatada/fisiopatología , Cardiomiopatía Dilatada/terapia , Estudios de Casos y Controles , China , Ecocardiografía Doppler en Color , Femenino , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/fisiopatología , Humanos , No Compactación Aislada del Miocardio Ventricular/complicaciones , No Compactación Aislada del Miocardio Ventricular/diagnóstico , No Compactación Aislada del Miocardio Ventricular/fisiopatología , Masculino , Persona de Mediana Edad , Recuperación de la Función , Volumen Sistólico , Factores de Tiempo , Resultado del Tratamiento
14.
Circ J ; 79(1): 201-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25373595

RESUMEN

BACKGROUND: Endothelial-mesenchymal transition (EndMT) plays a pivotal role in cardiac fibrosis. However, it is unclear whether EndMT is involved in dyssynchronous heart failure (DHF). METHODS AND RESULTS: Twelve dogs received 3-week rapid right ventricular pacing (RVP) to develop DHF and then were randomly divided into a RVP group (n=6; RVP for another 3 weeks) and a biventricular pacing (BiVP) group (n=6; BiVP for 3 weeks), and another 6 dogs were in the control group. Contractile function in BiVP group was a little better than that in RVP group (P<0.05), but significant heart failure remained in 2 groups. RVP induced more significant cardiac fibrosis and higher collagen 1A2 expression in the left ventricular lateral wall (late-contracting and high-stress) than that in the anterior wall, and for those in the BiVP group, it was much lower. CD31, S100A4, α-smooth muscle actin and collagen 1A2 were used to evaluate EndMT. EndMT levels, transforming growth factor-ß (TGF-ß)/snail signaling, collagen 1A2 and integrin ß1 expression were much higher in the endothelial cells from the RVP lateral wall than that from BiVP. In this in vitro study, cyclic stretch could independently induce EndMT and enhance the pro-EndMT effect of TGF-ß in HUVECs, which could be partly blocked by integrin ß1 siRNA. CONCLUSIONS: RVP-induced DHF could aggravate fibrosis due to regional heterogeneity of mechanical stress, and it was better in the BiVP group where mechanical stress-induced EndMT might play a pivotal role through the integrin ß1 pathway.


Asunto(s)
Estimulación Cardíaca Artificial/efectos adversos , Transdiferenciación Celular/fisiología , Endotelio/patología , Insuficiencia Cardíaca/fisiopatología , Mesodermo/patología , Miocardio/patología , Animales , Terapia de Resincronización Cardíaca/efectos adversos , Colágeno Tipo I/biosíntesis , Perros , Células Endoteliales/metabolismo , Fibrosis , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Ventrículos Cardíacos/fisiopatología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina beta1/biosíntesis , Integrina beta1/genética , Masculino , Contracción Miocárdica , Miocardio/metabolismo , Distribución Aleatoria , Transducción de Señal/fisiología , Factores de Transcripción de la Familia Snail , Estrés Mecánico , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/biosíntesis
15.
J Cell Mol Med ; 16(4): 657-71, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22004043

RESUMEN

Transplantation of bone marrow-derived mesenchymal stem cells (MSCs) is safe and may improve cardiac function and structural remodelling in patients following myocardial infarction (MI). Cardiovascular cell differentiation and paracrine effects to promote endogenous cardiac regeneration, neovascularization, anti-inflammation, anti-apoptosis, anti-remodelling and cardiac contractility, may contribute to MSC-based cardiac repair following MI. However, current evidence indicates that the efficacy of MSC transplantation was unsatisfactory, due to the poor viability and massive death of the engrafted MSCs in the infarcted myocardium. MicroRNAs are short endogenous, conserved, non-coding RNAs and important regulators involved in numerous facets of cardiac pathophysiologic processes. There is an obvious involvement of microRNAs in almost every facet of putative repair mechanisms of MSC-based therapy in MI, such as stem cell differentiation, neovascularization, apoptosis, cardiac remodelling, cardiac contractility and arrhythmias, and others. It is proposed that therapeutic modulation of individual cardiovascular microRNA of MSCs, either mimicking or antagonizing microRNA actions, will hopefully enhance MSC therapeutic efficacy. In addition, MSCs may be manipulated to enhance functional microRNA expression or to inhibit aberrant microRNA levels in a paracrine manner. We hypothesize that microRNAs may be used as novel regulators in MSC-based therapy in MI and MSC transplantation by microRNA regulation may represent promising therapeutic strategy for MI patients in the future.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Mesenquimatosas/citología , MicroARNs/uso terapéutico , Infarto del Miocardio/patología , Trasplante de Células Madre , Apoptosis , Diferenciación Celular , Humanos , MicroARNs/fisiología , Infarto del Miocardio/cirugía , Neovascularización Fisiológica
16.
Chin Med J (Engl) ; 123(17): 2295-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21034537

RESUMEN

BACKGROUND: Cardiac resynchronization therapy (CRT) could improve heart function, symptom status, quality of life and reduce hospitalization and mortality in patients with severe heart failure (HF) with optimal medical management. However, the possible adverse effects of CRT are often ignored by clinicians. METHOD: A retrospective analysis of CRT over a 6-year period was made in a single cardiac center. RESULTS: Fifty-four patients were treated with CRT(D) device, aged (57 ± 11) years, with left ventricular ejection fraction of (32.1 ± 9.8)%, of which 4 (7%) developed ventricular tachycardia/ventricular fibrillation (VT/VF) or junctional tachycardia after operation. Except for one with frequent ventricular premature beat before operation, the others had no previous history of ventricular arrhythmia. Of the 4 patients, 3 had dilated cardiomyopathy and 1 had ischemic cardiomyopathy, and tachycardia occurred within 3 days after operation. Sustained, refractory VT and subsequent VF occurred in one patient, frequent nonsustained VT in two patients and nonparoxysmal atrioventricular junctional tachycardia in one patient. VT was managed by amiodarone in two patients, amiodarone together with beta-blocker in one patient, and junctional tachycardia was terminated by overdrive pacing. During over 12-month follow-up, except for one patient's death due to refractory heart and respiratory failure in hospital, the others remain alive and arrhythmia-free. CONCLUSIONS: New-onset VT/VF or junctional tachycardia may occur in a minority of patients with or without prior history of tachycardia after biventricular pacing. Arrhythmia can be managed by conventional therapy, but may require temporary discontinuation of pacing. More observational studies should be performed to determine the potential proarrhythmic effect of CRT.


Asunto(s)
Terapia de Resincronización Cardíaca/efectos adversos , Taquicardia Ventricular/etiología , Fibrilación Ventricular/etiología , Humanos , Periodo Perioperatorio , Estudios Retrospectivos
18.
FEBS Lett ; 583(17): 2811-8, 2009 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-19631649

RESUMEN

It was found that C-reactive protein (CRP) could significantly increase the expression and activity of tissue factor (TF), but decrease that of tissue factor pathway inhibitor (TFPI) in human umbilical vein endothelial cells (HUVECs) in dose- and time-dependent manners, which could be antagonized by PDTC and U0126. CRP could also increase protein expression of phosphorylated nuclear factor-kappaB (NF-kappaB), IkappaB-alpha and ERK1/2 in dose- and time-dependent manner. In addition, neutralizing antibody to CD32 (FcgammaR II) could significantly attenuate the expression and activity of TF and TFPI induced by CRP. These results suggest that CRP may promote coagulation by enhancing the expression and activity of TF and reducing that of TFPI by activating NF-kappaB and extracellular signal-regulated kinase via FcgammaR II.


Asunto(s)
Proteína C-Reactiva/metabolismo , Inhibidores Enzimáticos/metabolismo , Lipoproteínas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/metabolismo , Tromboplastina/metabolismo , Anticuerpos/metabolismo , Proteína C-Reactiva/genética , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Lipoproteínas/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , FN-kappa B/genética , Receptores de IgG/metabolismo , Tromboplastina/genética
19.
Zhonghua Nei Ke Za Zhi ; 48(3): 205-7, 2009 Mar.
Artículo en Chino | MEDLINE | ID: mdl-19576087

RESUMEN

OBJECTIVE: To investigate the expression and significance of activator protein-1 (AP-1) and matrix metalloproteinases (MMPs) in acute myocardial infarction (AMI) subjects. METHODS: Immunohistochemical techniques were used to detect the subunit of AP-1 (c-Jun), MMP-2 and MMP-9 in human AMI and normal heart tissue and the expressions of c-Jun and MMPs were measured with computer image analysis system. RESULTS: (1) There were expressions of c-Jun, MMP-2 and MMP-9 in normal heart tissue, mainly in myocardial cells and cardiac fibroblasts, and their expressions in AMI myocardial tissues were all significantly higher than those in normal myocardial tissues (P < 0.05). (2) The level of MMP-9 expression was significantly and positively correlated with c-Jun in AMI heart tissue (r = 0.773, P < 0.01). CONCLUSIONS: The expressions of AP-1 and MMPs increase in human myocardial infarction. These findings suggest that AP-1 transcription activation pathway and MMPs may play an important role in ventricular remodeling of myocardial infarction.


Asunto(s)
Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Infarto del Miocardio/metabolismo , Factor de Transcripción AP-1/metabolismo , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/fisiopatología , Remodelación Ventricular
20.
Zhonghua Nei Ke Za Zhi ; 48(1): 28-30, 2009 Jan.
Artículo en Chino | MEDLINE | ID: mdl-19484973

RESUMEN

OBJECTIVE: Implantable cardioverter defibrillator (ICD) can effectively treat life-threatening ventricular arrhythmias. The most common side effect is inappropriate discharge. This study analyzes the incidence and causes of inappropriate discharges of ICD in our hospital. METHODS: Forty-three patients implanted with ICD in our hospital from November 2001 to October 2007 were involved in our study. Patients were followed-up regularly. All episodes recorded and stored in the ICD were analyzed. RESULTS: Seven of the 43 patients underwent ninety-six inappropriate discharges. Inappropriate discharges in six patients were caused by supraventricular tachyarrhythmias (SVT). In one patient the discharge was caused by noise. Most inappropriate discharges occurred in the first year after implantation. The history of atrial fibrillation before implantation is an independent predictor of inappropriate discharges. CONCLUSIONS: The incidence of inappropriate discharge is 16.3% in our study and the most common cause is SVT. Most inappropriate discharges occur in the first year after implantation. Patients with atrial fibrillation history have a higher risk of inappropriate discharges.


Asunto(s)
Desfibriladores Implantables/efectos adversos , Anciano , Falla de Equipo , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...