Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Nat Commun ; 13(1): 81, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013172

RESUMEN

Despite the high prevalence of ischemic heart diseases worldwide, no antibody-based treatment currently exists. Starting from the evidence that a specific isoform of the Bone Morphogenetic Protein 1 (BMP1.3) is particularly elevated in both patients and animal models of myocardial infarction, here we assess whether its inhibition by a specific monoclonal antibody reduces cardiac fibrosis. We find that this treatment reduces collagen deposition and cross-linking, paralleled by enhanced cardiomyocyte survival, both in vivo and in primary cultures of cardiac cells. Mechanistically, we show that the anti-BMP1.3 monoclonal antibody inhibits Transforming Growth Factor ß pathway, thus reducing myofibroblast activation and inducing cardioprotection through BMP5. Collectively, these data support the therapeutic use of anti-BMP1.3 antibodies to prevent cardiomyocyte apoptosis, reduce collagen deposition and preserve cardiac function after ischemia.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Proteína Morfogenética Ósea 1/genética , Cardiotónicos/farmacología , Cicatriz/genética , Fibrosis Endomiocárdica/genética , Infarto del Miocardio/genética , Animales , Proteína Morfogenética Ósea 1/antagonistas & inhibidores , Proteína Morfogenética Ósea 1/metabolismo , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 5/genética , Proteína Morfogenética Ósea 5/metabolismo , Estudios de Casos y Controles , Supervivencia Celular/efectos de los fármacos , Cicatriz/etiología , Cicatriz/metabolismo , Cicatriz/prevención & control , Modelos Animales de Enfermedad , Fibrosis Endomiocárdica/etiología , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/prevención & control , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Cultivo Primario de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Troponina T/genética , Troponina T/metabolismo
2.
Nat Commun ; 13(1): 149, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013221

RESUMEN

Cachexia is associated with poor prognosis in chronic heart failure patients, but the underlying mechanisms of cachexia triggered disease progression remain poorly understood. Here, we investigate whether the dysregulation of myokine expression from wasting skeletal muscle exaggerates heart failure. RNA sequencing from wasting skeletal muscles of mice with heart failure reveals a reduced expression of Ostn, which encodes the secreted myokine Musclin, previously implicated in the enhancement of natriuretic peptide signaling. By generating skeletal muscle specific Ostn knock-out and overexpressing mice, we demonstrate that reduced skeletal muscle Musclin levels exaggerate, while its overexpression in muscle attenuates cardiac dysfunction and myocardial fibrosis during pressure overload. Mechanistically, Musclin enhances the abundance of C-type natriuretic peptide (CNP), thereby promoting cardiomyocyte contractility through protein kinase A and inhibiting fibroblast activation through protein kinase G signaling. Because we also find reduced OSTN expression in skeletal muscle of heart failure patients, augmentation of Musclin might serve as therapeutic strategy.


Asunto(s)
Caquexia/genética , Fibrosis Endomiocárdica/genética , Insuficiencia Cardíaca/genética , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Factores de Transcripción/genética , 2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/genética , 2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Caquexia/metabolismo , Caquexia/fisiopatología , Caquexia/prevención & control , Estudios de Casos y Controles , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/fisiopatología , Fibrosis Endomiocárdica/prevención & control , Femenino , Regulación de la Expresión Génica , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/prevención & control , Pruebas de Función Cardíaca , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/agonistas , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/deficiencia , Atrofia Muscular/metabolismo , Atrofia Muscular/fisiopatología , Atrofia Muscular/prevención & control , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factores de Transcripción/agonistas , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/deficiencia
3.
Bull Exp Biol Med ; 171(3): 305-311, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34302205

RESUMEN

We studied the effects and mechanisms of action of conophylline in different concentrations in the original in vitro model of myocardial fibrosis (treatment of cardiac fibroblasts isolated form the hearts of newborn rats with angiotensin II). Viability, collagen content, and expression of related protein in cardiac fibroblasts were assessed using the MTT-test, Sircol assay, and Western blotting, respectively. Conophylline markedly protected the cultured cells against the development of angiotensin II-induced fibrosis, which was seen from reduced viability of fibroblasts, decreased collagen content, and down-regulation of the expression of α-smooth muscle actin (α-SMA). Conophylline did not affect the TGF-ß pathway altered by angiotensin II, but markedly decreased the level of bone morphogenetic protein-4 (BMP4) enhanced by angiotensin II and BMP4 itself. Conophylline produced no effect on phosphorylation of α-SMA and Smad homologue-1/5/8, the classic BMP4 downstream pathway elements, but reduced the level of c-Jun N-terminal kinase (JNK) elevated by BMP4. Conophylline did not inhibit the development of myocardial fibrosis in the presence of JNK activator anisomycin. Thus, conophylline inhibited angiotensin II-provoked myocardial fibrosis via the BMP4/JNK pathway.


Asunto(s)
Angiotensina II/farmacología , Antifibróticos/farmacología , Proteína Morfogenética Ósea 4/genética , Fibroblastos/efectos de los fármacos , MAP Quinasa Quinasa 4/genética , Alcaloides de la Vinca/farmacología , Animales , Animales Recién Nacidos , Proteína Morfogenética Ósea 4/antagonistas & inhibidores , Proteína Morfogenética Ósea 4/metabolismo , Colágeno/genética , Colágeno/metabolismo , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/prevención & control , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , Modelos Biológicos , Miocardio/metabolismo , Miocardio/patología , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Ratas , Ratas Wistar , Transducción de Señal , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Proteína Smad8/genética , Proteína Smad8/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Int J Mol Med ; 47(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33649779

RESUMEN

Oxidative stress serves a key role in doxorubicin (DOX)­induced cardiotoxicity. The peptide Szeto­Schiller (SS)31 is an efficacious antioxidant with the capacity to reduce mitochondrial reactive oxygen species (ROS) levels and scavenge free radicals. Although SS31 is involved in the pathophysiological process of various cardiovascular diseases, the role of SS31 in DOX­induced cardiotoxicity remains unclear. To explore the effects of SS31 in DOX­induced cardiotoxicity, the present study first constructed DOX­induced cardiotoxicity models, in which H9c2 cells were incubated with 1 µM DOX for 24 h and C57BL/6 mice were administered DOX (20 mg/kg cumulative dose). The results of various assays in these models demonstrated that SS31 exhibited a cardioprotective effect in vitro and in vivo by attenuating the level of ROS, stabilizing the mitochondrial membrane potential and ameliorating myocardial apoptosis as well as fibrosis following treatment with DOX. Mechanistically, the results of the present study revealed that the p38 MAPK signaling pathway was inhibited by SS31 in DOX­treated H9c2 cells, which was associated with the cardioprotective function of SS31. In addition, P79350, a selective agonist of p38 MAPK, reversed the protective effects of SS31. Taken together, these results demonstrated the effects of SS31 on ameliorating DOX­induced cardiotoxicity and indicated its potential as a drug for the treatment of DOX­induced cardiotoxicity.


Asunto(s)
Cardiotónicos/uso terapéutico , Doxorrubicina/toxicidad , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Miocardio/patología , Oligopéptidos/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Antioxidantes/uso terapéutico , Apoptosis/efectos de los fármacos , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/prevención & control , Fibrosis Endomiocárdica/tratamiento farmacológico , Fibrosis Endomiocárdica/prevención & control , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo
5.
Probl Radiac Med Radiobiol ; 25: 56-74, 2020 Dec.
Artículo en Inglés, Ucraniano | MEDLINE | ID: mdl-33361829

RESUMEN

The review is devoted to the current issues of radiation-induced cardiovascular complications, their diagnostics andincidence depending on the radiation doses and exposure regimens, potential efficiency of the screening strategiesfor cardiotoxicity monitoring after radiotherapy in cancer patients by analyzing the data from literature and clinical trials, based on recommendations of European Society of Cardiology and European Society of Medical Oncology.


Asunto(s)
Cardiomiopatías/patología , Cardiotoxicidad/patología , Fibrosis Endomiocárdica/patología , Válvulas Cardíacas/efectos de la radiación , Corazón/efectos de la radiación , Cardiomiopatías/etiología , Cardiomiopatías/prevención & control , Cardiotoxicidad/etiología , Cardiotoxicidad/prevención & control , Relación Dosis-Respuesta en la Radiación , Fibrosis Endomiocárdica/etiología , Fibrosis Endomiocárdica/prevención & control , Endotelio Vascular/patología , Endotelio Vascular/efectos de la radiación , Corazón/efectos de los fármacos , Corazón/fisiopatología , Válvulas Cardíacas/efectos de los fármacos , Válvulas Cardíacas/patología , Humanos , Neoplasias/patología , Neoplasias/radioterapia , Radiación Ionizante , Protectores contra Radiación/uso terapéutico
6.
Bull Exp Biol Med ; 169(4): 549-557, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32910392

RESUMEN

Regenerative potential of multipotent mesenchymal stromal cells from the human umbilical cord (MMSC-UC) in the suspension and spheroid form was revealed during the progression of experimental small focal myocardial infarction in rats. In isoproterenol-induced myocardial infarction, foci of necrosis and inflammatory infiltrate and at later terms fibrosis foci were found mainly in the left ventricle of rat heart. In rats receiving MMSC-UC, destructive changes in the myocardium, fibrous scars, and inflammatory process were less pronounced. MMSC-UC also contributed to normalization of the morphofunctional parameters of the heart. Spheroids exhibited higher efficiency in comparison with cell suspension.


Asunto(s)
Fibrosis Endomiocárdica/prevención & control , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Infarto del Miocardio/terapia , Regeneración/fisiología , Esferoides Celulares/trasplante , Animales , Modelos Animales de Enfermedad , Fibrosis Endomiocárdica/inducido químicamente , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/fisiopatología , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/ultraestructura , Humanos , Isoproterenol/administración & dosificación , Masculino , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/inducido químicamente , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Miocardio/ultraestructura , Miocitos Cardíacos/patología , Miocitos Cardíacos/ultraestructura , Cultivo Primario de Células , Ratas , Ratas Wistar , Esferoides Celulares/citología , Esferoides Celulares/fisiología , Trasplante Heterólogo , Resultado del Tratamiento , Cordón Umbilical/citología , Cordón Umbilical/metabolismo
7.
J Am Heart Assoc ; 9(16): e016270, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32791029

RESUMEN

Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce the risk of cardiovascular death and hospitalization for heart failure in patients with chronic heart failure. Initially, these drugs were believed to have a profile similar to diuretics or hemodynamically active drugs, but they do not rapidly reduce natriuretic peptides or cardiac filling pressures, and they exert little early benefit on symptoms, exercise tolerance, quality of life, or signs of congestion. Clinically, the profile of SGLT2 inhibitors resembles that of neurohormonal antagonists, whose benefits emerge gradually during sustained therapy. In experimental models, SGLT2 inhibitors produce a characteristic pattern of cellular effects, which includes amelioration of oxidative stress, mitigation of mitochondrial dysfunction, attenuation of proinflammatory pathways, and a reduction in myocardial fibrosis. These cellular effects are similar to those produced by angiotensin converting enzyme inhibitors, ß-blockers, mineralocorticoid receptor antagonists, and neprilysin inhibitors. At a molecular level, SGLT2 inhibitors induce transcriptional reprogramming of cardiomyocytes that closely mimics that seen during nutrient deprivation. This shift in signaling activates the housekeeping pathway of autophagy, which clears the cytosol of dangerous cytosolic constituents that are responsible for cellular stress, thereby ameliorating the development of cardiomyopathy. Interestingly, similar changes in cellular signaling and autophagic flux have been seen with inhibitors of the renin-angiotensin system, ß-blockers, mineralocorticoid receptor antagonists, and neprilysin inhibitors. The striking parallelism of these molecular, cellular, and clinical profiles supports the premise that SGLT2 inhibitors should be regarded as neurohormonal antagonists when prescribed for the treatment of heart failure with a reduced ejection fraction.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Antagonistas de Hormonas/farmacología , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Antagonistas Adrenérgicos beta/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Autofagia/efectos de los fármacos , Autofagia/fisiología , Cardiomiopatías/prevención & control , Cardiotónicos/farmacología , Reprogramación Celular/efectos de los fármacos , Enfermedad Crónica , Fibrosis Endomiocárdica/prevención & control , Insuficiencia Cardíaca/etiología , Hemodinámica/efectos de los fármacos , Humanos , Inflamación/prevención & control , Antagonistas de Receptores de Mineralocorticoides/farmacología , Enfermedades Mitocondriales/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Neprilisina/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos
8.
ESC Heart Fail ; 7(5): 2838-2852, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32662949

RESUMEN

AIMS: Left ventricular (LV) dysfunction in viral myocarditis is attributed to myocardial inflammation and fibrosis, inducing acute and long-time cardiac damage. Interventions are not established. On the basis of the link between inflammation, fibrosis, aldosterone, and extracellular matrix regulation, we aimed to investigate the effect of an early intervention with the mineralocorticoid receptor antagonist (MRA) eplerenone on cardiac remodelling in a murine model of persistent coxsackievirus B3 (CVB3)-induced myocarditis. METHODS AND RESULTS: SWR/J mice were infected with 5 × 104 plaque-forming units of CVB3 (Nancy strain) and daily treated either with eplerenone (200 mg/kg body weight) or with placebo starting from Day 1. At Day 8 or 28 post infection, mice were haemodynamically characterized and subsequently sacrificed for immunohistological and molecular biology analyses. Eplerenone did not influence CVB3 load. Already at Day 8, 1.8-fold (P < 0.05), 1.4-fold (P < 0.05), 3.2-fold (P < 0.01), and 2.1-fold (P < 0.001) reduction in LV intercellular adhesion molecule 1 expression, presence of monocytes/macrophages, oxidative stress, and apoptosis, respectively, was observed in eplerenone-treated vs. untreated CVB3-infected mice. In vitro, eplerenone led to 1.4-fold (P < 0.01) and 1.2-fold (P < 0.01) less CVB3-induced cardiomyocyte oxidative stress and apoptosis. Furthermore, collagen production was 1.1-fold (P < 0.05) decreased in cardiac fibroblasts cultured with medium of eplerenone-treated vs. untreated CVB3-infected HL-1 cardiomyocytes. These ameliorations were in vivo translated into prevention of cardiac fibrosis, as shown by 1.4-fold (P < 0.01) and 2.1-fold (P < 0.001) lower collagen content in the LV of eplerenone-treated vs. untreated CVB3-infected mice at Days 8 and 28, respectively. This resulted in an early and long-lasting improvement of LV dimension and function, as indicated by reduced LV end-systolic volume and end-diastolic volume, and an increase in LV contractility (dP/dtmax ) and LV relaxation (dP/dtmin ), respectively (P < 0.05). CONCLUSIONS: Early intervention with the MRA eplerenone modulates the acute host and defence reaction and prevents cardiac disease progression in experimental CVB3-induced myocarditis without aggravation of viral load. The findings advocate for an initiation of therapy of viral myocarditis as early as possible, even before the onset of inflammation-induced myocardial dysfunction. This may also have implications for coronavirus disease-19 therapy.


Asunto(s)
Fibrosis Endomiocárdica/prevención & control , Enterovirus Humano B/patogenicidad , Eplerenona/farmacología , Miocarditis/tratamiento farmacológico , Miocarditis/virología , Disfunción Ventricular Izquierda/virología , Análisis de Varianza , Animales , Biopsia con Aguja , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis Endomiocárdica/patología , Inmunohistoquímica , Masculino , Metaloproteinasas de la Matriz/efectos de los fármacos , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Transgénicos , Miocarditis/prevención & control , Distribución Aleatoria , Valores de Referencia , Resultado del Tratamiento , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
9.
Med Sci Monit ; 25: 318-323, 2019 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-30631031

RESUMEN

BACKGROUND Hypertension is a leading global disease, and myocardial fibrosis is an important adverse effect of hypertension, seriously threatening human health. The IL-6/STAT3 pathway and endothelin-1 (ET-1) were previously suggested to play a part in myocardial fibrosis. MATERIAL AND METHODS To investigate the role of Atorvastatin (Ato) in spontaneous hypertension, systolic blood pressure (SBP) and left ventricular mass index (LVMI) were measured, and Masson trichrome staining was performed. Furthermore, the relative protein levels of the IL-6/STAT3/ET-1 pathway were tested. RESULTS Ato prevented myocardial fibrosis in spontaneous hypertension rats, especially at the dosage of 50 mg/kg/d. The IL-6/STAT3 pathway was observed to be suppressed by Ato, and ET-1 level in myocardial tissues was also downregulated by Ato. The phosphorylation status of STAT3 was tested after Ato treatment, showing that Ato mainly stimulated the tyr-705 phosphorylation of STAT3. CONCLUSIONS Results of this study may help promote myocardial fibrosis therapy and provide insights into the IL-6/STAT3/ET-1-mediated mechanism in Ato-induced myocardial fibrosis inhibition.


Asunto(s)
Atorvastatina/farmacología , Hipertensión/metabolismo , Animales , Atorvastatina/metabolismo , Presión Sanguínea , Cardiomiopatías/metabolismo , Modelos Animales de Enfermedad , Fibrosis Endomiocárdica/tratamiento farmacológico , Fibrosis Endomiocárdica/prevención & control , Endotelina-1/metabolismo , Fibrosis/patología , Hipertensión/complicaciones , Interleucina-6/metabolismo , Masculino , Miocardio/patología , Fosforilación , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Ann Thorac Surg ; 107(5): 1448-1455, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30552887

RESUMEN

BACKGROUND: The pedicled greater omentum has been shown to offer benefit in ischemic heart disease for both animal models and human patients. The impact of cardio-omentopexy in a pressure overload model of left ventricular hypertrophy (LVH) is unknown. METHODS: LVH was created in rats by banding the ascending aorta after right thoracotomy (n = 23). Sham surgery was performed in 12 additional rats. Six weeks after banding, surviving LVH rats were assigned to cardio-omentopexy by left thoracotomy (LVH+Om, n = 8) or sham left thoracotomy (LVH, n = 8). Sham rats also underwent left thoracotomy for cardio-omentopexy (Sham+Om, n = 6); the remaining rats underwent sham left thoracotomy (Sham, n = 6). RESULTS: Echocardiography 10 weeks after cardio-omentopexy revealed LV end-systolic diameter, cardiomyocyte diamter, and myocardial fibrosis in the LVH group were significantly increased compared with the LVH+Om, Sham+Om, and Sham groups (p < 0.01). LV ejection fraction of the LVH group was lower than the LVH+Om group (p < 0.01). Gene expression analysis revealed significantly lower levels of sarcoendoplasmic reticulum calcium adenosine triphosphatase 2b in LVH rats than in the LVH+Om, Sham+Om, and Sham groups (p < 0.01). In contrast, collagen type 1 α 1 chain, lysyl oxidase-like protein 1, nuclear protein-1, and transforming growth factor- ß1 in the LVH group were significantly higher than in the LVH+Om cohort (p < 0.01), consistent with a reduced fibrotic phenotype after omentopexy. Lectin staining showed myocardial capillary density of the LVH group was significantly lower than all other groups (p < 0.01). CONCLUSIONS: Cardio-omentopexy reduced cardiac dilation, contractile dysfunction, cardiomyocyte hypertrophy, and myocardial fibrosis, while maintaining other molecular indicators of contractile function in this LVH model.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos , Fibrosis Endomiocárdica/prevención & control , Insuficiencia Cardíaca/prevención & control , Hipertrofia Ventricular Izquierda/cirugía , Epiplón/cirugía , Animales , Cadena alfa 1 del Colágeno Tipo I , Modelos Animales de Enfermedad , Fibrosis Endomiocárdica/etiología , Insuficiencia Cardíaca/etiología , Hipertrofia Ventricular Izquierda/complicaciones , Hipertrofia Ventricular Izquierda/patología , Masculino , Miocardio , Ratas , Ratas Sprague-Dawley , Volumen Sistólico
11.
DNA Cell Biol ; 37(10): 821-830, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30227089

RESUMEN

Premenopausal women have a reduced risk for cardiovascular disease. Estrogen deficiency augments cardiac inflammation and oxidative stress and, thereby, aggravates myocardial fibrosis (MF) and diastolic dysfunction in hypertensive female rats. However, estrogen replacement therapy has no effect on myocardial infarction and cardiac fibrosis in postmenopausal women. Further clinical studies showed that high blood glucose levels in patients with diabetes is an important cause of MF, but the underlying mechanism is unclear. To experimentally address this issue, diabetes mellitus (DM) was induced by injecting streptozotocin and administering a high-fat diet in ovariectomized (OVX) rats. High degrees of fibrosis and apoptosis were detected in the cardiac tissue of these rats, together with increased expression of iNOS. Further treatment with the G protein-coupled estrogen receptor 30 (GPR30) agonist G1 decreased iNOS expression and the apoptosis rate in cardiac tissue significantly and inhibited cardiac fibroblast (CF) proliferation. Similar trends were observed in cultured CFs treated with high concentrations of fat and glucose. In addition, treatment with the iNOS-specific inhibitor W1400 attenuated iNOS and vimentin expression, which is associated with a marked reduction in MF. These results suggest that GPR30 activation inhibits MF in diabetic OVX female rats by suppressing cardiac iNOS activity and consequently NO levels. Thus, GPR30 activation may provide novel cardioprotection strategies for postmenopausal women, especially those with DM.


Asunto(s)
Diabetes Mellitus Experimental/genética , Fibrosis Endomiocárdica/genética , Fibroblastos/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Ovariectomía , Receptores Acoplados a Proteínas G/genética , Amidinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Glucemia/metabolismo , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Dieta Alta en Grasa , Fibrosis Endomiocárdica/etiología , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/prevención & control , Inhibidores Enzimáticos/farmacología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Regulación de la Expresión Génica , Humanos , Miocardio/metabolismo , Miocardio/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Estreptozocina , Vimentina/genética , Vimentina/metabolismo
12.
Cardiovasc Ther ; 36(3): e12321, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29380561

RESUMEN

INTRODUCTION: Atrial structural remodeling in the form of fibrosis contributes to the arrhythmic substrate in atrial fibrillation (AF). The aim of this study was to investigate the effects of doxycycline on chronic intermittent hypoxia (CIH)-induced atrial fibrosis and the pathophysiological mechanisms underlying such changes. METHODS: A total of 30 Sprague Dawley rats were randomized into three groups: control group, CIH group, and CIH with doxycycline treatment (CIH-D) group. CIH lasted 5 hours per day for 4 weeks. CIH-D rats were administrated doxycycline for 4 weeks, while they received CIH. Masson's trichrome staining was used to determine collagen deposit in the atrial myocardium. Protein and mRNA levels of Matrix Metalloproteinase-2 (MMP-2) and -9 (MMP-9), microRNA-21 (miR-21) and its downstream target Sprouty1 (Spry1), and extracellular signal-regulated kinases 1/2 (ERK1/2) were measured using Western blotting or real-time qRT-PCR, respectively. RESULTS: Compared to the control group, the CIH group showed higher interstitial collagen fraction, increased MMP-9, miR-21, and p-ERK1/2 levels, and decreased MMP-2 and Spry1 levels. Doxycycline treatment attenuated CIH-induced atrial fibrosis, reduced MMP-2, MMP-9, miR-21, and p-ERK1/2, and increased Spry1. CONCLUSIONS: CIH treatment induced significant atrial fibrosis in our rat model, which was attenuated by doxycycline. These changes can be explained by alterations in the MMP and miR-21/ERK signaling pathways.


Asunto(s)
Antibacterianos/uso terapéutico , Doxiciclina/uso terapéutico , Fibrosis Endomiocárdica/prevención & control , Cardiopatías/etiología , Cardiopatías/prevención & control , Hipoxia/complicaciones , Animales , Enfermedad Crónica , Ecocardiografía , Fibrosis Endomiocárdica/diagnóstico por imagen , Cardiopatías/diagnóstico por imagen , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Metaloproteinasas de la Matriz/metabolismo , MicroARNs/genética , Proteínas del Tejido Nervioso/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
13.
Biochem Biophys Res Commun ; 491(2): 329-336, 2017 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-28733035

RESUMEN

Hyperglycemia plays a crucial role in the pathogenesis of diabetic complications; however, the mechanisms underlying diabetic cardiac fibrosis remain unclear. Endothelial cells are known to contribute to cardiac fibrosis through endothelial-mesenchymal transition (EndMT) under high glucose stimulation. Here we investigated the expression of miR-18a-5p and examined its functional role in human aortic valvular endothelial cells (HAVECs). Using HAVECs, we revealed that miR-18a-5p regulated high glucose-induced EndMT. Moreover, high glucose levels induced Notch2 expression, which promoted EndMT, resulting in the downregulation of vascular endothelial cadherin and CD31 and upregulation of fibroblast-specific protein-1, α-smooth muscle actin, fibronectin, and vimentin. Furthermore, Notch2 was identified as a target of miR-18a-5p. Our data showed that the overexpression of miR-18a-5p could downregulate Notch2 expression and subsequently suppress EndMT. In conclusion, our findings demonstrated that miR-18a-5p/Notch2 signaling pathway participates in the regulation of high glucose-induced EndMT, and may act as a novel promising target for myocardial fibrosis in diabetic cardiomyopathy.


Asunto(s)
Dependovirus/genética , Cardiomiopatías Diabéticas/genética , Fibrosis Endomiocárdica/genética , Transición Epitelial-Mesenquimal , MicroARNs/genética , Receptor Notch2/genética , Actinas/genética , Actinas/metabolismo , Animales , Válvula Aórtica/citología , Válvula Aórtica/efectos de los fármacos , Válvula Aórtica/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Línea Celular , Dependovirus/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Cardiomiopatías Diabéticas/inducido químicamente , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/prevención & control , Fibrosis Endomiocárdica/inducido químicamente , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/prevención & control , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Regulación de la Expresión Génica , Vectores Genéticos/administración & dosificación , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Glucosa/farmacología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Humanos , Masculino , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptor Notch2/metabolismo , Proteína de Unión al Calcio S100A4 , Transducción de Señal , Estreptozocina , Vimentina/genética , Vimentina/metabolismo
14.
Cell Physiol Biochem ; 41(3): 849-864, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28214892

RESUMEN

BACKGROUND/AIMS: Baicalin has been shown to be effective for various animal models of cardiovascular diseases, such as pulmonary hypertension, atherosclerosis and myocardial ischaemic injury. However, whether baicalin plays a role in cardiac hypertrophy remains unknown. Here we investigated the protective effects of baicalin on cardiac hypertrophy induced by pressure overload and explored the potential mechanisms involved. METHODS: C57BL/6J-mice were treated with baicalin or vehicle following transverse aortic constriction or Sham surgery for up to 8 weeks, and at different time points, cardiac function and heart size measurement and histological and biochemical examination were performed. RESULTS: Mice under pressure overload exhibited cardiac dysfunction, high mortality, myocardial hypertrophy, increased apoptosis and fibrosis markers, and suppressed cardiac expression of PPARα and PPARß/δ. However, oral administration of baicalin improved cardiac dysfunction, decreased mortality, and attenuated histological and biochemical changes described above. These protective effects of baicalin were associated with reduced heart and cardiomyocyte size, lower fetal genes expression, attenuated cardiac fibrosis, lower expression of profibrotic markers, and decreased apoptosis signals in heart tissue. Moreover, we found that baicalin induced PPARα and PPARß/δ expression in vivo and in vitro. Subsequent experiments demonstrated that long-term baicalin treatment presented no obvious cardiac lipotoxicity. CONCLUSIONS: The present results demonstrated that baicalin attenuates pressure overload induced cardiac dysfunction and ventricular remodeling, which would be due to suppressed cardiac hypertrophy, fibrosis, apoptosis and metabolic abnormality.


Asunto(s)
Cardiotónicos/farmacología , Fibrosis Endomiocárdica/prevención & control , Flavonoides/farmacología , Hipertrofia Ventricular Izquierda/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Disfunción Ventricular Izquierda/prevención & control , Animales , Línea Celular , Tamaño de la Célula , Modelos Animales de Enfermedad , Fibrosis Endomiocárdica/etiología , Fibrosis Endomiocárdica/mortalidad , Fibrosis Endomiocárdica/patología , Regulación de la Expresión Génica , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/mortalidad , Hipertrofia Ventricular Izquierda/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , PPAR alfa/agonistas , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR delta/agonistas , PPAR delta/genética , PPAR delta/metabolismo , PPAR-beta/agonistas , PPAR-beta/genética , PPAR-beta/metabolismo , Presión/efectos adversos , Transducción de Señal , Análisis de Supervivencia , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/mortalidad , Disfunción Ventricular Izquierda/patología
15.
Biochem J ; 474(3): 399-410, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27803247

RESUMEN

Cardiac fibrosis and chronic inflammation are common complications in type 2 diabetes mellitus (T2D). Since nucleotide oligomerization-binding domain 1 (NOD1), an innate immune receptor, is involved in the pathogenesis of insulin resistance and diabetes outcomes, we sought to investigate its involvement in cardiac fibrosis. Here, we show that selective staining of cardiac fibroblasts from T2D (db/db;db) mice exhibits up-regulation and activation of the NOD1 pathway, resulting in enhanced NF-κB and TGF-ß signalling. Activation of the TGF-ß pathway in cardiac fibroblasts from db mice was prevented after inhibition of NF-κB with BAY-11-7082 (BAY). Moreover, fibrosis progression in db mice was also prevented by BAY treatment. Enhanced TGF-ß signalling and cardiac fibrosis of db mice was dependent, at least in part, on the sequential activation of NOD1 and NF-κB since treatment of db mice with a selective NOD1 agonist induced activation of the TGF-ß pathway, but co-administration of a NOD1 agonist plus BAY, or a NOD1 inhibitor prevented the NOD1-induced fibrosis. Therefore, NOD1 is involved in cardiac fibrosis associated with diabetes, and establishes a new mechanism for the development of heart fibrosis linked to T2D.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Fibrosis Endomiocárdica/metabolismo , Miocardio/metabolismo , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Ácido Diaminopimélico/análogos & derivados , Ácido Diaminopimélico/farmacología , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/prevención & control , Regulación de la Expresión Génica , Humanos , Insulina/sangre , Resistencia a la Insulina , Ratones , Ratones Transgénicos , Miocardio/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Células 3T3 NIH , Nitrilos/farmacología , Proteína Adaptadora de Señalización NOD1/agonistas , Proteína Adaptadora de Señalización NOD1/genética , Transducción de Señal , Sulfonas/farmacología , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
16.
Int Heart J ; 57(3): 336-40, 2016 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-27149999

RESUMEN

Chronic inflammation is known to occur in diabetes mellitus (DM) and contributes to atrial fibrosis, possible substrates for atrial fibrillation. We tested the hypothesis that dipeptidyl peptidase (DPP)-4 inhibitors prevent the formation of atrial fibrosis through their anti-inflammatory activity, beyond the effects of controlling blood glucose.DM models obtained by administration of streptozotocin (STZ) were divided into 3 groups: with PKF275-055, a DPP-4 inhibitor in group D, glibenclamide in group SU, and no additional drug in group P. At 8 weeks after STZ administration, the heart was subjected to Masson trichrome staining and immunohistochemistry with anti-ED2, ED3, and smooth muscle actin antibody.The % area of fibrosis in atria of group P accounted for 14.7% ± 4.1%, showing a significant increase in fibrosis when compared with the control group. In group SU, the % area accounted for 7.9% ± 2.9%, indicating significant deceased fibrosis by sulfonylurea. Meanwhile, we could not find significant differences in group D when compared to group P or group SU. While ED3-positive cells increased in group P (1.12% ± 0.24%), they were significantly decreased in groups D and SU (0.41% ± 0.22% and 0.55% ± 0.29%, respectively). Between group D and SU, however, there were no significant differences in the amount of cells positive to ED2, ED3, and smooth muscle actin antibodies.In STZ-induced DM rats, administration of sulfonylurea and DPP-4 inhibitors inhibited inflammation and fibrosis of the atria. However, no significant differences were observed between the 2 antidiabetic drugs.


Asunto(s)
Adamantano/análogos & derivados , Fibrilación Atrial , Diabetes Mellitus Experimental , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Fibrosis Endomiocárdica , Gliburida/farmacología , Atrios Cardíacos , Nitrilos/farmacología , Pirrolidinas/farmacología , Adamantano/farmacología , Animales , Antiinflamatorios/farmacología , Fibrilación Atrial/fisiopatología , Fibrilación Atrial/prevención & control , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/fisiopatología , Fibrosis Endomiocárdica/etiología , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/prevención & control , Femenino , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/patología , Atrios Cardíacos/fisiopatología , Hipoglucemiantes/farmacología , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/prevención & control , Ratas , Ratas Wistar , Estadística como Asunto
17.
J Atheroscler Thromb ; 23(11): 1294-1306, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27052784

RESUMEN

AIM: Endoplasmic reticulum stress (ERS) and inflammation participate in cardiac fibrosis. Importantly, a novel paracrine/autocrine peptide intermedin1-53 (IMD1-53) in the heart inhibits myocardial fibrosis in rats. However, the mechanisms are yet to be fully elucidated. METHODS: Myocardial fibrosis in apolipoprotein E-deficient (ApoE -/-) mice and neonatal rat cardiac fibroblasts (CFs) were induced using homocysteine (Hcy). RESULTS: IMD1-53 inhibited myocardial fibrosis in vivo and in vitro. Picrosirius red staining showed that IMD1-53 reduced myocardial interstitial collagen deposition in ApoE-/- mice treated with Hcy and decreased the expression of myocardial collagen I and III, which was further verified in rat CFs. IMD1-53 attenuated myocardial hypertrophy, as shown by cardiomyocyte cross-sectional area, ratio of heart weight to body weight, and mRNA levels of atrial natriuretic peptide and brain natriuretic peptide. IMD1-53 inhibited the upregulation of ERS hallmarkers such as glucose-regulated protein 78 (GRP78), GRP94, activating transcription factor 6 (ATF6), ATF4, inositol-requiring enzyme 1α, spliced-X-box-binding protein-1, protein kinase receptor-like ER kinase, and eukaryotic translation initiation factor 2α in mouse myocardium and rat CFs treated with Hcy. In addition, IMD1-53 decreased the production of inflammatory factors such as tumor necrosis factor-α, monocyte chemotactic protein-1, interleukin-6 (IL-6), and IL-1ß in the mouse myocardium and rat CFs treated with Hcy. Concurrently, IMD1-53 ameliorated the expression of nuclear factor-κB, transforming growth factor-ß1, and c-Jun N-terminal kinase in the mouse myocardium and rat CFs treated with Hcy. CONCLUSIONS: IMD potentially protects against myocardial fibrosis induced by Hcy in ApoE-/- mice, possibly via attenuating myocardial ERS and inflammation.


Asunto(s)
Apolipoproteínas E/deficiencia , Fibrosis Endomiocárdica/prevención & control , Estrés del Retículo Endoplásmico/efectos de los fármacos , Homocisteína/efectos adversos , Inflamación/prevención & control , Errores Innatos del Metabolismo Lipídico/metabolismo , Neuropéptidos/fisiología , Animales , Animales Recién Nacidos , Apolipoproteínas E/metabolismo , Western Blotting , Células Cultivadas , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Chaperón BiP del Retículo Endoplásmico , Técnica del Anticuerpo Fluorescente , Inflamación/metabolismo , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Cardiology ; 133(2): 97-108, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26513353

RESUMEN

OBJECTIVES: The aim of this study was to investigate the protective role of erythropoietin (EPO) against myocardial fibrosis (MF). METHODS: Pressure-overloaded rats were established by abdominal aortic constriction, the rats were randomly divided in a double-blind manner into 3 groups (n = 12 for each group): sham-operated rats (sham), operated rats receiving physiological saline (vehicle) and operated rats receiving 4,000 U/kg rhEPO (EPO group). The vehicle and drugs were administered to rats by intraperitoneal injection. In addition, cultured adult rat cardiac fibroblasts (CFs) were utilized to investigate the role of EPO in CF proliferation and collagen secretion. RESULTS: After 4 weeks, besides an increase in blood pressure, myocardial hypertrophy, collagen deposition in the myocardium and decreased cardiac function were observed in the pressure-overloaded rats. The expression of NADPH oxidase (Nox2 and Nox4) and inflammatory cytokines (CD45, F4/80 and MCP-1) was also significantly increased. All these alterations were prevented by EPO. TGF-ß promoted CF proliferation, collagen secretion, ROS production and Nox2/Nox4 expression, which was inhibited by EPO. In addition, the TGF-ß-induced increase of ERK1/2 phosphorylation and NF-x03BA;B expression were attenuated by EPO. CONCLUSION: EPO inhibited rat MF induced by pressure overload and improved myocardial function by decreasing CF proliferation and differentiation via inhibition of the NADPH-ERK-NF-x03BA;B pathway.


Asunto(s)
Fibrosis Endomiocárdica/tratamiento farmacológico , Fibrosis Endomiocárdica/prevención & control , Eritropoyetina/uso terapéutico , Factor de Crecimiento Transformador beta/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Western Blotting , Células Cultivadas , Método Doble Ciego , Fibrosis Endomiocárdica/fisiopatología , Eritropoyetina/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Inmunohistoquímica , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Miocardio/patología , NADP/efectos de los fármacos , NADP/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
19.
Toxicol Lett ; 241: 9-18, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26581635

RESUMEN

Sunitinib (Su) is currently approved for treatment of several malignances. However, along with the benefits of disease stabilization, cardiovascular toxicities have also been increasingly recognized. The aim of this study was to analyze which mechanisms are involved in the cardiotoxicity caused by Su, as well as to explore the potential cardioprotective effects of l-carnitine (LC). To this end, four groups of Wistar rats were used: (1) control; (2) rats treated with 400mg LC/kg/day; (3) rats treated with 25mg Su/kg/day; and (4) rats treated with LC+Su simultaneously. In addition, cultured rat cardiomyocytes were treated with an inhibitor of nuclear factor kappa B (NF-κB), in order to examine the role of this transcription factor in this process. An elevation in the myocardial expression of pro-inflammatory cytokines, together with an increase in the mRNA expression of NF-κB, was observed in Su-treated rats. These results were accompanied by an increase in the expression of pro-fibrotic factors, nitrotyrosine and NOX 2 subunit of NADPH oxidase; and by a decrease in that of collagen degradation factor. Higher blood pressure and heart rate levels were also found in Su-treated rats. All these alterations were inhibited by co-administration of LC. Furthermore, cardiotoxic effects of Su were blocked by NF-κB inhibition. Our results suggest that: (i) inflammatory and fibrotic processes are involved in the cardiac toxicity observed following treatment with Su; (ii) these processes might be mediated by the transcription factor NF-κB; (iii) LC exerts a protective effect against arterial hypertension, cardiac inflammation and fibrosis, which are all observed after Su treatment.


Asunto(s)
Antineoplásicos/toxicidad , Cardiotónicos/farmacología , Carnitina/farmacología , Cardiopatías/inducido químicamente , Cardiopatías/prevención & control , Indoles/antagonistas & inhibidores , Indoles/toxicidad , Miocarditis/inducido químicamente , Miocarditis/prevención & control , Pirroles/antagonistas & inhibidores , Pirroles/toxicidad , Animales , Presión Sanguínea/efectos de los fármacos , Cardiotoxicidad , Citocinas/biosíntesis , Fibrosis Endomiocárdica/inducido químicamente , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/prevención & control , Expresión Génica/efectos de los fármacos , Cardiopatías/patología , Masculino , Miocarditis/patología , Miocitos Cardíacos/efectos de los fármacos , FN-kappa B/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Wistar , Sunitinib
20.
Radiat Res ; 184(6): 611-20, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26579943

RESUMEN

Radiation-induced heart injury is one of the major side effects of radiotherapy for thoracic malignancies. Previous studies have shown that radiotherapy induced myocardial fibrosis and intensified myocardial remodeling. In this study, we investigated whether atorvastatin could inhibit radiation-induced heart fibrosis in Sprague-Dawley rats, which were randomly divided into six groups: control; radiation only; and four treatment groups receiving atorvastatin plus radiation (E1, E2, E3 and E4). All rats, except the control group, received local heart irradiation in 7 daily fractions of 3 Gy for a total of 21 Gy. Rats in groups E1 (10 mg/kg/day) and E2 (20 mg/kg/day) received atorvastatin and radiation treatment until week 12 after exposure. Rats in groups E3 (10 mg/kg/day) and E4 (20 mg/kg/day) received atorvastatin treatment from 3 months before irradiation to week 12 after irradiation. The expressions of TGF-ß1, Smad2, Smad3, fibronectin, ROCK I and p-Akt in heart tissues were evaluated using real-time PCR or Western blot analyses. Atorvastatin significantly reduced the expression of TGF-ß1, Smad3/P-Smad3, ROCK I and p-Akt in rats of the E1-E4 groups and in a dose-dependent manner. Fibronectin exhibited a similar pattern of expression changes. In addition, echocardiography showed that atorvastatin treatment can inhibit the increase of left ventricular end-diastolic dimension, left ventricular end-systolic diameter and left ventricular posterior wall thickness, and prevent the decrease of ejection fraction and fraction shortening in E1-E4 groups compared with the radiation only group. This study demonstrated that radiation exposure increased the expression of fibronectin in cardiac fibroblasts and induced cardiac fibrosis through activation of the TGF-ß1/Smad3, RhoA/ROCK, and PI3K/AKT signaling pathways. Statins ameliorated radiation-induced cardiac fibrosis in Sprague-Dawley rats. Our results suggest that atorvastatin is effective for the treatment of radiation-induced cardiac fibrosis, especially with longer and higher dose atorvastatin treatment, as demonstrated in experimental group E4.


Asunto(s)
Atorvastatina/administración & dosificación , Cardiotoxicidad/prevención & control , Cardiotoxicidad/fisiopatología , Fibrosis Endomiocárdica/prevención & control , Fibrosis Endomiocárdica/fisiopatología , Factores Inmunológicos/metabolismo , Animales , Cardiotoxicidad/etiología , Relación Dosis-Respuesta a Droga , Fibrosis Endomiocárdica/etiología , Masculino , Dosis de Radiación , Radiación Ionizante , Protectores contra Radiación/administración & dosificación , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA