Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 175
Filtrar
1.
Alcohol Clin Exp Res ; 38(2): 336-43, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24033682

RESUMEN

BACKGROUND: Alcohol abuse increases the risk for acute lung injury (ALI). In both experimental models and in clinical studies, chronic alcohol ingestion causes airway oxidative stress and glutathione depletion and increases the expression of transforming growth factor beta-1 (TGFß1), a potent inducer of fibrosis, in the lung. Therefore, we hypothesized that alcohol ingestion could promote aberrant fibrosis following experimental ALI and that treatment with the glutathione precursor s-adenosylmethionine (SAMe) could mitigate these effects. METHODS: Three-month-old C57BL/6 mice were fed standard chow ± alcohol (20% v/v) in their drinking water for 8 weeks and ±SAMe (4% w/v) during the last 4 weeks. ALI was induced by intratracheal instillation of bleomycin (2.5 units/kg), and lungs were assessed histologically at 7 and 14 days for fibrosis and at 14 days for the expression of extracellular matrix proteins and TGFß1. RESULTS: Alcohol ingestion had no apparent effect on lung inflammation at 7 days, but at 14 days after bleomycin treatment, it increased lung tissue collagen deposition, hydroxyproline content, and the release of activated TGFß1 into the airway. In contrast, SAMe supplementation completely mitigated alcohol-induced priming of these aberrant fibrotic changes through decreased TGFß1 expression in the lung. In parallel, SAMe decreased alcohol-induced TGFß1 and Smad3 mRNA expressions by lung fibroblasts in vitro. CONCLUSIONS: These new experimental findings demonstrate that chronic alcohol ingestion renders the experimental mouse lung susceptible to fibrosis following bleomycin-induced ALI, and that these effects are likely driven by alcohol-mediated oxidative stress and its induction and activation of TGFß1.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Fibrosis Pulmonar/inducido químicamente , Actinas/biosíntesis , Animales , Antibióticos Antineoplásicos/antagonistas & inhibidores , Bleomicina/antagonistas & inhibidores , Diferenciación Celular/efectos de los fármacos , Depresores del Sistema Nervioso Central/antagonistas & inhibidores , Dieta , Ensayo de Inmunoadsorción Enzimática , Etanol/antagonistas & inhibidores , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Hidroxiprolina/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Neumonía/patología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/prevención & control , ARN Mensajero/biosíntesis , ARN Mensajero/genética , S-Adenosilmetionina/farmacología , Factor de Crecimiento Transformador beta1/biosíntesis
2.
J Cardiovasc Pharmacol ; 63(4): 360-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24336018

RESUMEN

Daunorubicin (DNR) is a widely used chemotherapeutic agent; however, its clinical use is limited because of its cardiotoxicity. This study was aimed to investigate the protective effect of sodium ferulate (SF), an effective component from traditional Chinese herbs, against DNR-induced cardiotoxicity in juvenile rats. DNR was administered intraperitoneally to rats at the dosage of 2.5 mg·kg(-1)·wk(-1) for 5 consecutive weeks (cumulative dose of 12.5 mg/kg) or in combination with intraperitoneal injection of SF at 50 mg·kg(-1)·d(-1) over a period of 30 days. The animals were killed 6 days after the last injection of DNR. SF significantly ameliorated the DNR-induced cardiac dysfunction, structural damage of the myocardium, and release of lactate dehydrogenase and creatine kinase. Treatment with SF also reversed DNR-induced oxidative stress as evidenced by a decrease in malondialdehyde levels with a concomitant increase in myocardical superoxide dismutase activities. Furthermore, SF afforded significant cardioprotection against DNR-induced apoptosis in vivo and effectively suppressed the complex mitochondrion-dependent apoptotic signaling triggered by DNR. This study indicates that SF may improve cardiac function by inhibition of oxidative stress and apoptosis, thus providing a beneficial effect on the prevention of DNR-induced cardiotoxicity.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antibióticos Antineoplásicos/antagonistas & inhibidores , Antibióticos Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Ácidos Cumáricos/uso terapéutico , Daunorrubicina/antagonistas & inhibidores , Daunorrubicina/toxicidad , Cardiopatías/inducido químicamente , Cardiopatías/prevención & control , Mitocondrias/efectos de los fármacos , Animales , Cardiopatías/patología , Hemodinámica , Masculino , Miocardio/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
3.
Pharmazie ; 69(8): 633-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25158576

RESUMEN

Interactions between doxorubicin (DOX) and iron generate reactive oxygen species and contribute to DOX-induced heart failure. Hydrogen, as a selective antioxidant, is a promising potential therapeutic option for the treatment of a variety of diseases. Therefore, we investigated the preventive effects of hydrogen treatment on DOX-induced heart failure in rats. We found that cardiac function was significantly improved and that the plasma levels of oxidative-stress markers and myocardial autophagic activity were decreased in animals treated with hydrogen-containing saline. Therefore, we conclude that hydrogen-containing saline may have beneficial effects for doxorubicin-induced heart failure.


Asunto(s)
Antibióticos Antineoplásicos/antagonistas & inhibidores , Antibióticos Antineoplásicos/toxicidad , Cardiotónicos , Doxorrubicina/antagonistas & inhibidores , Doxorrubicina/toxicidad , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/prevención & control , Hidrógeno/farmacología , Cloruro de Sodio/química , Animales , Autofagia/efectos de los fármacos , Western Blotting , Ecocardiografía , Insuficiencia Cardíaca/diagnóstico por imagen , Hidrógeno/química , Masculino , Microscopía Electrónica de Transmisión , Miocardio/patología , Estrés Oxidativo/efectos de los fármacos , Soluciones Farmacéuticas , Ratas , Ratas Wistar , Sobrevida , Función Ventricular Izquierda/efectos de los fármacos
4.
Am J Physiol Endocrinol Metab ; 305(2): E243-53, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23695218

RESUMEN

Because doxorubicin (DOX)-containing chemotherapy causes left ventricular (LV) dysfunction and remodeling that can progress to heart failure, strategies to alleviate DOX cardiotoxicity are necessary to improve health outcomes of patients surviving cancer. Although clinical evidence suggests that aerobic exercise training (ET) can prevent cardiotoxicity in patients undergoing DOX chemotherapy, the physiological mechanisms involved have not been extensively studied, nor is it known whether compounds [such as resveratrol (RESV)] have similar beneficial effects. With the use of a murine model of chronic DOX exposure, this study compared the efficacy of modest ET to RESV treatment on exercise performance, LV remodeling, and oxidative stress resistance. Mice were divided into four groups that received saline, DOX (8 mg/kg ip, one time per week), DOX + RESV (4 g/kg diet, ad libitum), and DOX + ET (45 min of treadmill exercise, 5 days/wk) for 8 wk. LV function and morphology were evaluated by in vivo echocardiography. DOX caused adverse LV remodeling that was partially attenuated by modest ET and completely prevented by RESV. These effects were paralleled by improvements in exercise performance. The cardioprotective properties of ET and RESV were associated with reduced levels of atrial natriuretic peptide and the lipid peroxidation by-product, 4-hydroxy-2-nonenal. In addition, ET and RESV increased the expression of cardiac sarcoplasmic/endoplasmic reticulum calcium-ATPase 2a, superoxide dismutase, mitochondrial electron transport chain complexes, and mitofusin-1 and -2 in mice administered DOX. Compared with modest ET, RESV more effectively prevented DOX-induced LV remodeling and was associated with the reduction of DOX-induced oxidative stress. Our findings have important implications for protecting patients against DOX-associated cardiac injury.


Asunto(s)
Antibióticos Antineoplásicos/antagonistas & inhibidores , Antibióticos Antineoplásicos/toxicidad , Antioxidantes/farmacología , Doxorrubicina/antagonistas & inhibidores , Doxorrubicina/toxicidad , Cardiopatías/inducido químicamente , Cardiopatías/prevención & control , Condicionamiento Físico Animal/fisiología , Estilbenos/farmacología , Animales , Biomarcadores/metabolismo , Presión Sanguínea/fisiología , Western Blotting , Suplementos Dietéticos , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Femenino , GTP Fosfohidrolasas/metabolismo , Cardiopatías/patología , Peroxidación de Lípido/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Resveratrol , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/prevención & control
5.
J Cardiovasc Pharmacol ; 62(6): 512-23, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24072175

RESUMEN

Doxorubicin (DOX) is a potent available antitumor drug; however, its clinical use is limited by the cardiotoxicity. Salidroside (SLD), with strong antioxidative and cytoprotective actions, is of particular interest in the development of antioxidative therapies for oxidative injury in cardiac diseases. Now, the protection and underlying mechanisms of SLD against DOX-induced cardiotoxicity are still unknown. In the present study, we revealed both antioxidative mechanism and Bcl2-dependent survival signaling involved in SLD's protection. We observed that DOX exposure induced mortality elevation, body weight loss, and cardiac dysfunction in mice, increased lactate dehydrogenase leakage and cardiomyocyte apoptosis, but decreased cell viability and size in cardiac tissues and cultured H9c2 cells, respectively, which were effectively antagonized by SLD supplement. We further observed that SLD significantly reduced the intercellular oxidative stress level, partly by inhibiting NOX1 expression and augmenting the expression and activities of the endogenous antioxidative enzymes, catalase, and manganese superoxide dismutase. In addition, SLD treatment upregulated the antiapoptotic Bcl2 and downregulated the proapoptotic Bax and inhibited a downstream pathway of Bcl2/Bax and caspase-3 activity. Our results indicated that SLD effectively protected the cardiomyocytes against DOX-induced cardiotoxicity by suppressing the excessive oxidative stress and activating a Bcl2-mediated survival signaling pathway.


Asunto(s)
Antibióticos Antineoplásicos/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Cardiotónicos/uso terapéutico , Doxorrubicina/antagonistas & inhibidores , Glucósidos/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Fenoles/uso terapéutico , Disfunción Ventricular/prevención & control , Animales , Antibióticos Antineoplásicos/efectos adversos , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Cardiotónicos/farmacología , Línea Celular , Tamaño de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Clonales , Doxorrubicina/efectos adversos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucósidos/farmacología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Oxidorreductasas/antagonistas & inhibidores , Oxidorreductasas/química , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Fenoles/farmacología , Distribución Aleatoria , Ratas , Disfunción Ventricular/inducido químicamente , Disfunción Ventricular/metabolismo , Disfunción Ventricular/fisiopatología
6.
J Cardiovasc Pharmacol ; 62(2): 130-7, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23519142

RESUMEN

Recombinant human neuregulin-1 (rhNRG-1) improves cardiac function in animal models of doxorubicin (DOX)-induced cardiomyopathy, but the underlying mechanism remains largely unknown. Here, we confirm a role for rhNRG-1 in attenuating DOX-induced autophagy and define the signaling pathways through which it mediates some of its effects. Neonatal rat ventricular myocytes were subjected to different treatments both to induce autophagy and to determine the effects of rhNRG-1 on the process. The rhNRG-1 inhibited DOX-induced autophagy, reduced reactive oxygen species production and increased protein expression of Bcl-2, effects that were recapitulated when the cells were treated with the antioxidant N-acetylcysteine. These effects were blocked by the phosphatidylinositol 3-kinase inhibitor LY294002, pointing to the involvement of the Akt pathway in mediating the process. Inhibition of Bcl-2 expression with small interfering RNA silencing also inhibited rhNRG-1's ability to attenuate DOX-induced autophagy. The rhNRG-1 is a potent inhibitor of DOX-induced autophagy and multiple signaling pathways, including Akt and activation of reactive oxygen species, play important roles in the anti-autophagy effect. The rhNRG-1 is a novel drug that may be effectively therapeutically in protecting further damage in DOX-induced damaged myocardium.


Asunto(s)
Antibióticos Antineoplásicos/antagonistas & inhibidores , Autofagia/efectos de los fármacos , Cardiotónicos/farmacología , Doxorrubicina/antagonistas & inhibidores , Miocitos Cardíacos/efectos de los fármacos , Neurregulina-1/farmacología , Proteínas Recombinantes/farmacología , Animales , Animales Recién Nacidos , Antibióticos Antineoplásicos/efectos adversos , Antioxidantes/farmacología , Cardiomiopatías/inducido químicamente , Cardiomiopatías/prevención & control , Células Cultivadas , Doxorrubicina/efectos adversos , Inhibidores Enzimáticos/farmacología , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Neurregulina-1/genética , Estrés Oxidativo/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
7.
J Appl Toxicol ; 33(3): 202-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21996788

RESUMEN

The aim of this study was to investigate the antioxidant and anti-apoptotic effects of onion (Allium cepa) extracts (ACE) on doxorubicin (DOX)-induced cardiotoxicity. The rats in the ACE-pretreated group were given a daily dose of 1 ml ACE for 14 days. To induce cardiotoxicity, DOX (30 mg kg(-1) body weight) was injected intraperitoneally by a single dose and the rats were sacrificed after 48 h. To date, no such studies have been performed on the cardioprotective and anti-apoptotic potential of ACE on DOX-induced cardiotoxicity. Our data indicate a significant reduction in the activity of in situ identification of apoptosis using terminal dUTP nick end-labeling in cardiomyocytes of the DOX-treated group with ACE therapy. The DOX-treated with ACE groups showed a significant decrease in malondialdehyde levels, and increased activities of superoxide dismutase, glutathione and glutathione peroxidase in comparison with the DOX-treated group. Creatine kinase, creatine kinase MB, lactate dehydrogenase activities and cardiac troponin I levels were significantly decreased in the DOX + ACE group in comparison with the DOX group. These biochemical and histological disturbances were effectively attenuated on pretreatment with ACE. The present study showed that ACE may be a suitable cardioprotector against toxic effects of DOX.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Doxorrubicina/toxicidad , Cardiopatías/tratamiento farmacológico , Cebollas/química , Extractos Vegetales/farmacología , Animales , Antibióticos Antineoplásicos/antagonistas & inhibidores , Doxorrubicina/antagonistas & inhibidores , Antagonismo de Drogas , Corazón/efectos de los fármacos , Cardiopatías/inducido químicamente , Cardiopatías/patología , Masculino , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Ratas , Ratas Sprague-Dawley
8.
Pharm Biol ; 51(3): 339-44, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23134235

RESUMEN

CONTEXT: Doxorubicin (Dox) is an anthracycline antibiotic used as anticancer agent. However, its use is limited due to its cardiotoxicity which is mainly attributed to accumulation of reactive oxygen species. OBJECTIVE: This study was conducted to assess whether the antioxidant, proanthocyanidins (Pro) can ameliorate Dox-induced cardiotoxicity in rats. MATERIALS AND METHODS: Male Sprague-Dawely rats were divided into four groups. Group I was control. Group II received Pro (70 mg/kg, orally) once daily for 10 days. Group III received doxorubicin 15 mg/kg i.p. as a single dose on the 7th day and Group IV animals were treated with Pro once daily for 10 days and Dox on the 7th day. The parameters of study were serum biomarkers, cardiac tissue antioxidant status, ECG, and effect on aconitine-induced cardiotoxicity. RESULTS: Cardiac toxicity of doxorubicin was manifested as a significant increase in heart rate, elevation of the ST segment, prolongation of the QT interval and an increase in T wave amplitude. In addition, Dox enhanced aconitine-induced cardiotoxicity by a significant decrease in the aconitine dose producing ventricular tachycardia (VT). Administration of Pro significantly suppressed Dox-induced ECG changes and normalized the aconitine dose producing VT. The toxicity of Dox was also confirmed biochemically by significant elevation of serum CK-MB and LDH activities as well as myocardial MDA and GSH contents and decrease in serum catalase and myocardial SOD activities. Administration of Pro significantly suppressed these biochemical changes. DISCUSSION AND CONCLUSION: These results suggest that proanthocyanidins might be a potential cardioprotective agent against Dox-induced cardiotoxicity due to its antioxidant properties.


Asunto(s)
Antioxidantes/uso terapéutico , Cardiomiopatía Dilatada/prevención & control , Cardiotónicos/uso terapéutico , Cardiotoxinas/antagonistas & inhibidores , Doxorrubicina/antagonistas & inhibidores , Extracto de Semillas de Uva/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Proantocianidinas/uso terapéutico , Aconitina/administración & dosificación , Aconitina/antagonistas & inhibidores , Aconitina/toxicidad , Animales , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/antagonistas & inhibidores , Arritmias Cardíacas/etiología , Arritmias Cardíacas/prevención & control , Biomarcadores/sangre , Cardiomiopatía Dilatada/inducido químicamente , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/fisiopatología , Cardiotoxinas/efectos adversos , Doxorrubicina/efectos adversos , Resistencia a Medicamentos/efectos de los fármacos , Corazón/efectos de los fármacos , Corazón/fisiopatología , Masculino , Miocardio/metabolismo , Fitoterapia , Ratas , Ratas Sprague-Dawley , Taquicardia/inducido químicamente , Taquicardia/prevención & control , Agonistas del Canal de Sodio Activado por Voltaje/administración & dosificación , Agonistas del Canal de Sodio Activado por Voltaje/toxicidad
9.
J Med Assoc Thai ; 95 Suppl 1: S109-14, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23964452

RESUMEN

BACKGROUND: Nowadays, anticarcinogenic potential of pigmented brown rice and rice bran varieties have been increasingly stated. However, their mechanisms of action are still inconclusive. One of which might be their antigenotoxic activity that no study in human cells was reported before. OBJECTIVE: To evaluate the antigenotoxic activities of Thai Sangyod red rice extracts against a chemotherapeutic agent, doxorubicin, by sister chromatid exchange (SCE) assay in human lymphocytes in vitro. MATERIAL AND METHOD: Two fractions of water-soluble of Sangyod rice extracts were used: (i) the washed water extract of brown rice (WWBR) and (ii) the water extract of rice bran (WERB). Human lymphocytes were pretreated with each extracts at concentrations of 6.2, 12.5, 25, 50 and 100 microg/ml for 2 h followed by a genotoxic agent, doxorubicin (DXR) (0.1 microg/ml) for 2 h. SCE level, mitotic index (MI) and proliferation index (PI) were evaluated. Statistical analysis by Dunnett's t-test was performed. RESULTS: The results indicated that the pretreatment of WERB fraction only at concentration of 6.2 microg/ml could significantly decrease SCE level as compared to that of the DXR treated alone (p < 0.05). On the other hand, WERB fraction at other concentrations and all WWBR pretreatments could not. In addition, there was no significant difference in MI and PI levels between all pretreated extracts as compared to the DXR treated alone. CONCLUSION: Our data revealed that WERB pretreatment only at specific low concentration of 6.2 microg/ml possessed the antigenotoxic potential against genotoxic damage but not anticytotoxic induced by DXR. Further work is still needed to clarify more the antigenotoxic and anticytotoxic potentials from other fractions of Sangyod rice extracts.


Asunto(s)
Antibióticos Antineoplásicos/antagonistas & inhibidores , Doxorrubicina/antagonistas & inhibidores , Linfocitos/efectos de los fármacos , Oryza , Extractos Vegetales/farmacología , Intercambio de Cromátides Hermanas/efectos de los fármacos , Adulto , Antibióticos Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Doxorrubicina/farmacología , Humanos , Índice Mitótico , Pruebas de Mutagenicidad , Pigmentos Biológicos/farmacología , Tailandia
10.
J Pharmacol Exp Ther ; 335(1): 223-30, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20668052

RESUMEN

The anthracycline doxorubicin (Dox) is an effective antitumor agent. However, its use is limited because of its toxicity in the heart. N-Benzyladriamycin-14-valerate (AD 198) is a modified anthracycline with antitumor efficacy similar to that of Dox, but with significantly less cardiotoxicity and potentially cardioprotective elements. In the present study, we investigated the possibility of in vivo protective effects of low-dose AD 198 against Dox-induced cardiomyopathy. To do this, rats were divided into four groups: vehicle, Dox (20 mg/kg; single injection day 1), AD 198 (0.3 mg/kg per injection; injections on days 1, 2, and 3), or a combination treatment of Dox + AD 198. Seventy-two hours after beginning treatment, hearts from the Dox group had decreased phosphorylation of AMP kinase and troponin I and reduced poly(ADP-ribose) polymerase, ß-tubulin, and serum albumin expression. Dox also increased the phosphorylation of phospholamban and expression of inducible nitric-oxide synthase in hearts. Each of these Dox-induced molecular changes was attenuated in the Dox + AD 198 group. In addition, excised hearts from rats treated with Dox had a 25% decrease in left ventricular developed pressure (LVDP) and a higher than normal increase in LVDP when perfused with a high extracellular Ca(2+) solution. The Dox-induced decrease in baseline LVDP and hyper-responsiveness to [Ca(2+)] was not observed in hearts from the Dox + AD 198 group. Thus Dox, with well established and efficient antitumor protocols, in combination with low levels of AD 198, to counter anthracycline cardiotoxicity, may be a promising next step in chemotherapy.


Asunto(s)
Antibióticos Antineoplásicos/antagonistas & inhibidores , Antibióticos Antineoplásicos/toxicidad , Cardiomiopatías/inducido químicamente , Cardiomiopatías/prevención & control , Cardiotónicos/farmacología , Doxorrubicina/antagonistas & inhibidores , Doxorrubicina/toxicidad , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Western Blotting , Calcio/farmacología , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacología , Electroforesis en Gel de Poliacrilamida , Indicadores y Reactivos , Masculino , Espectrometría de Masas , Miocitos Cardíacos/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Fosforilación , Poli Adenosina Difosfato Ribosa/farmacología , Proteína Quinasa C-epsilon/antagonistas & inhibidores , Proteínas Quinasas/metabolismo , Ratas , Ratas Wistar
11.
J Pharmacol Exp Ther ; 334(3): 1023-30, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20543097

RESUMEN

Doxorubicin (DOX) is one of the most effective anticancer drugs. However, its cardiotoxicity remains a clinical concern that severely restricts its therapeutic usage. We designed this study to investigate whether tadalafil, a long-acting phosphodiesterase-5 (PDE-5) inhibitor, protects against DOX-induced cardiotoxicity. We also sought to delineate the cellular and molecular mechanisms underlying tadalafil-induced cardioprotection. Male CF-1 outbred mice were randomized into three groups (n = 15-24/group) to receive either saline (0.2 ml i.p.), DOX (15 mg/kg, given by a single intraperitoneal injection), or tadalafil (4 mg/kg p.o. daily for 9 days) plus DOX. Left ventricular function was subsequently assessed by transthoracic echocardiography and Millar conductance catheter. Cardiac contractile function was impaired by DOX, and it was significantly improved by cotreatment with tadalafil. Tadalafil attenuated DOX-induced apoptosis and depletion of prosurvival proteins, including Bcl-2 and GATA-4, in myocardium. Cardiac oxidative stress was attenuated and antioxidant capacity was enhanced by tadalafil possibly via up-regulation of mitochondrial superoxide dismutase (MnSOD). Moreover, the tadalafil-treated group demonstrated increased cardiac cGMP level and protein kinase G (PKG) activity. Tadalafil did not interfere with the efficacy of DOX in killing human osteosarcoma cells in vitro or its antitumor effect in vivo in tumor xenograft model. We conclude that tadalafil improved left ventricular function and prevented cardiomyocyte apoptosis in DOX-induced cardiomyopathy through mechanisms involving up-regulation of cGMP, PKG activity, and MnSOD level without interfering with the chemotherapeutic benefits of DOX.


Asunto(s)
Antibióticos Antineoplásicos/antagonistas & inhibidores , Antibióticos Antineoplásicos/toxicidad , Carbolinas/farmacología , Cardiomiopatías/inducido químicamente , Cardiomiopatías/tratamiento farmacológico , Doxorrubicina/antagonistas & inhibidores , Doxorrubicina/toxicidad , Inhibidores de Fosfodiesterasa 5 , Inhibidores de Fosfodiesterasa/farmacología , Animales , Antibióticos Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Western Blotting , Carbolinas/farmacocinética , Cardiomiopatías/diagnóstico por imagen , Línea Celular Tumoral , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Doxorrubicina/uso terapéutico , Factor de Transcripción GATA4/biosíntesis , Genes bcl-2/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Miocitos Cardíacos/efectos de los fármacos , Trasplante de Neoplasias , Inhibidores de Fosfodiesterasa/farmacocinética , Superóxido Dismutasa/biosíntesis , Tadalafilo , Ultrasonografía , Función Ventricular Izquierda/efectos de los fármacos
12.
J Pharmacol Exp Ther ; 334(3): 729-38, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20571059

RESUMEN

The mechanism of action of TNP-470 [O-(chloroacetyl-carbamoyl) fumagillol], which potently and selectively inhibits the proliferation of endothelial cells, is incompletely understood. Previous studies have established its binding protein and the most distal effector of its growth arrest activity as methionine aminopeptidase 2 (MetAP-2) and p21(WAF1/CIP1), respectively. However, the mechanistic steps between these two effectors have not been identified. We have found that addition of exogenous guanine and guanine-containing nucleosides to culture medium will completely reverse the cytostatic effect of TNP-470 on both cultured bovine aortic and mouse pulmonary endothelial cells. Western blotting showed that supplementation with exogenous guanosine reverses the induction of p21(WAF1/CIP1) by TNP-470. This "rescue" by guanine/guanosine was abolished when the guanine salvage pathway of nucleotide biosynthesis was inhibited with Immucillin H, suggesting that TNP-470 might reduce de novo guanine synthesis in endothelial cells. However, an analysis of inosine 5'-monophosphate dehydrogenase, the rate-limiting enzyme in de novo guanine synthesis and target of the antiangiogenic drug mycophenolic acid, showed no TNP-470-induced changes. Curiously, quantitation of cellular nucleotides confirmed that GTP levels were not reduced after TNP-470 treatment. Addition of guanosine at the start of G(1) phase causes a doubling in GTP levels that persists to the G(1)/S phase transition, where commitment to TNP-470 growth arrest occurs. Thus, guanine rescue involves an augmentation of cellular GTP beyond physiological levels rather than a restoration of a drug-induced GTP deficit. Determining the mechanism whereby this causes restoration of endothelial cell proliferation is an ongoing investigation.


Asunto(s)
Antibióticos Antineoplásicos/antagonistas & inhibidores , Antibióticos Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Ciclohexanos/antagonistas & inhibidores , Ciclohexanos/farmacología , Células Endoteliales/efectos de los fármacos , Nucleótidos de Guanina/farmacología , Guanina/farmacología , Sesquiterpenos/antagonistas & inhibidores , Sesquiterpenos/farmacología , Adenosina Trifosfato/biosíntesis , Animales , Western Blotting , Bovinos , Ciclo Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Inducción Enzimática/efectos de los fármacos , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ratones , Ácido Micofenólico/farmacología , O-(Cloroacetilcarbamoil) Fumagilol , Timidina/metabolismo , Proteína p53 Supresora de Tumor/biosíntesis , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/biosíntesis
13.
Horm Metab Res ; 42(5): 311-7, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20213584

RESUMEN

It has been reported that the immunosuppressant rapamycin decreases the viability of pancreatic beta cells. In contrast, exendin-4, an analogue of glucagon-like peptide-1, has been found to inhibit beta cell death and to increase beta cell mass. We investigated the effects of exendin-4 on the cytotoxic effect of rapamycin in beta cells. Incubation with 10 nM rapamycin induced cell death in 12 h in murine beta cell line MIN6 cells and Wistar rat islets, but not when coincubated with 10 nM exendin-4. Rapamycin was found to increase phosphorylation of c-Jun amino-terminal kinase (JNK) and p38 in 30 minutes in MIN6 cells and Wistar rat islets while exendin-4 decreased their phosphorylation. Akt and extracellular signal-regulated kinase (ERK) were not involved in the cytoprotective effect of exendin-4. These results indicate that exendin-4 may exert its protective effect against rapamycin-induced cell death in pancreatic beta cells by inhibiting JNK and p38 signaling.


Asunto(s)
Antibióticos Antineoplásicos/antagonistas & inhibidores , Antibióticos Antineoplásicos/toxicidad , Células Secretoras de Insulina/efectos de los fármacos , MAP Quinasa Quinasa 4/antagonistas & inhibidores , Péptidos/farmacología , Sirolimus/antagonistas & inhibidores , Sirolimus/toxicidad , Ponzoñas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Western Blotting , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Relación Dosis-Respuesta a Droga , Exenatida , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Citometría de Flujo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/patología , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación/efectos de los fármacos , Ratas , Ratas Wistar
14.
Circ Res ; 103(11): 1270-9, 2008 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-18948619

RESUMEN

Doxorubicin (DOX) is a widely used antitumor drug, but its application is limited because of its cardiotoxic side effects. Heat shock protein (Hsp)20 has been recently shown to protect cardiomyocytes against apoptosis, induced by ischemia/reperfusion injury or by prolonged beta-agonist stimulation. However, it is not clear whether Hsp20 would exert similar protective effects against DOX-induced cardiac injury. Actually, DOX treatment was associated with downregulation of Hsp20 in the heart. To elucidate the role of Hsp20 in DOX-triggered cardiac toxicity, Hsp20 was first overexpressed ex vivo by adenovirus-mediated gene delivery. Increased Hsp20 levels conferred higher resistance to DOX-induced cell death, compared to green fluorescent protein control. Furthermore, cardiac-specific overexpression of Hsp20 in vivo significantly ameliorated acute DOX-triggered cardiomyocyte apoptosis and animal mortality. Hsp20 transgenic mice also showed improved cardiac function and prolonged survival after chronic administration of DOX. The mechanisms underlying these beneficial effects were associated with preserved Akt phosphorylation/activity and attenuation of DOX-induced oxidative stress. Coimmunoprecipitation studies revealed an interaction between Hsp20 and phosphorylated Akt. Accordingly, BAD phosphorylation was preserved, and cleaved caspase-3 was decreased in DOX-treated Hsp20 transgenic hearts, consistent with the antiapoptotic effects of Hsp20. Parallel ex vivo experiments showed that either infection with a dominant-negative Akt adenovirus or preincubation of cardiomyocytes with the phosphatidylinositol 3-kinase inhibitors significantly attenuated the protective effects of Hsp20. Taken together, our findings indicate that overexpression of Hsp20 inhibits DOX-triggered cardiac injury, and these beneficial effects appear to be dependent on Akt activation. Thus, Hsp20 may constitute a new therapeutic target in ameliorating the cardiotoxic effects of DOX treatment in cancer patients.


Asunto(s)
Cardiomiopatías/inducido químicamente , Doxorrubicina/toxicidad , Proteínas del Choque Térmico HSP20/fisiología , Corazón/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Antibióticos Antineoplásicos/antagonistas & inhibidores , Antibióticos Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Cardiomiopatías/prevención & control , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/antagonistas & inhibidores , Proteínas del Choque Térmico HSP20/efectos de los fármacos , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Etiquetado Corte-Fin in Situ , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Ratas
15.
Am J Physiol Heart Circ Physiol ; 297(1): H37-46, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19429816

RESUMEN

Human cytochrome P-450 (CYP)2J2 is abundant in heart and active in biosynthesis of epoxyeicosatrienoic acids (EETs). Recently, we demonstrated that these eicosanoid products protect myocardium from ischemia-reperfusion injury. The present study utilized transgenic (Tr) mice with cardiomyocyte-specific overexpression of human CYP2J2 to investigate protection toward toxicity resulting from acute (0, 5, or 15 mg/kg daily for 3 days, followed by 24-h recovery) or chronic (0, 1.5, or 3.0 mg/kg biweekly for 5 wk, followed by 2-wk recovery) doxorubicin (Dox) administration. Acute treatment resulted in marked elevations of serum lactate dehydrogenase and creatine kinase levels that were significantly greater in wild-type (WT) than CYP2J2 Tr mice. Acute treatment also resulted in less activation of stress response enzymes in CYP2J2 Tr mice (catalase 750% vs. 300% of baseline, caspase-3 235% vs. 165% of baseline in WT vs. CYP2J2 Tr mice). Moreover, CYP2J2 Tr hearts exhibited less Dox-induced cardiomyocytes apoptosis (measured by TUNEL) compared with WT hearts. After chronic treatment, comparable decreases in body weight were observed in WT and CYP2J2 Tr mice. However, cardiac function, assessed by measurement of fractional shortening with M-mode transthoracic echocardiography, was significantly higher in CYP2J2 Tr than WT hearts after chronic Dox treatment (WT 37 +/- 2%, CYP2J2 Tr 47 +/- 1%). WT mice also had larger increases in beta-myosin heavy chain and cardiac ankryin repeat protein compared with CYP2J2 Tr mice. CYP2J2 Tr hearts had a significantly higher rate of Dox metabolism than WT hearts (2.2 +/- 0.25 vs. 1.6 +/- 0.50 ng.min(-1).100 microg protein(-1)). In vitro data from H9c2 cells demonstrated that EETs attenuated Dox-induced mitochondrial damage. Together, these data suggest that cardiac-specific overexpression of CYP2J2 limited Dox-induced toxicity.


Asunto(s)
Antibióticos Antineoplásicos/antagonistas & inhibidores , Antibióticos Antineoplásicos/toxicidad , Sistema Enzimático del Citocromo P-450/fisiología , Doxorrubicina/antagonistas & inhibidores , Doxorrubicina/toxicidad , Cardiopatías/inducido químicamente , Cardiopatías/fisiopatología , Animales , Biomarcadores , Creatina Quinasa/metabolismo , Citocromo P-450 CYP2J2 , Sistema Enzimático del Citocromo P-450/biosíntesis , Sistema Enzimático del Citocromo P-450/genética , Ecocardiografía , Femenino , Expresión Génica/genética , Pruebas de Función Cardíaca , Humanos , Etiquetado Corte-Fin in Situ , Técnicas In Vitro , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mitocondrias Cardíacas/efectos de los fármacos
16.
Int Immunopharmacol ; 9(1): 70-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18976720

RESUMEN

OBJECTIVE: Abnormal angiogenesis is a central hallmark for the development and progression of idiopathic pulmonary fibrosis. It has been shown that vascular endothelial growth factor (VEGF) is one of the critical angiogenic factors in angiogenesis. The aim of the present study was to assess whether disruption of VEGF pathway would attenuate bleomycin-induced pulmonary fibrosis. METHODS: Bleomycin-induced pulmonary fibrosis mice were treated intraperitoneally with VEGF receptor tyrosine kinase inhibitor SU5416 at different phases after bleomycin infusion. We measured angiogenesis and inflammatory response in both bleomycin-treated and control mice, and correlated these levels with pulmonary fibrosis. RESULTS: The increased expressions of VEGF/VEGFR (Flk-1) were correlated to a larger number of microvessels and a higher score of pulmonary fibrosis. Early administration of SU5416 inhibited pulmonary collagen deposition, histopathologic fibroplasias and the activation of TGF-beta1/Smad3 signaling pathway in bleomycin-stimulated lung. These were also paralleled by a reduction of VEGF/VEGFR-2 (Flk-1) expression and microvessel numbers in lung. Furthermore, SU5416 inhibited inflammatory cell numbers and LDH activity in BALF and IL-13 expression in lung tissue at early inflammatory phase of bleomycin-induced pulmonary fibrosis. CONCLUSION: These results suggest that the VEGFR-2 inhibitor, SU5416, attenuates histopathologic fibroplasias and collagen deposition by regulating angiogenesis and inflammation in the lung.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antibióticos Antineoplásicos/antagonistas & inhibidores , Antibióticos Antineoplásicos/toxicidad , Bleomicina/antagonistas & inhibidores , Bleomicina/toxicidad , Indoles/farmacología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/prevención & control , Pirroles/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Western Blotting , Líquido del Lavado Bronquioalveolar/citología , Citometría de Flujo , Hidroxiprolina/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/patología , Neovascularización Patológica/prevención & control , Fibrosis Pulmonar/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Proteína smad3/biosíntesis , Proteína smad3/genética , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Factor A de Crecimiento Endotelial Vascular/biosíntesis
17.
Toxicology ; 255(1-2): 72-9, 2009 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19010377

RESUMEN

Anthracycline-induced cardiomyopathy is a major problem in anti-cancer therapy. The only approved agent for alleviating this serious dose limiting side effect is ICRF-187 (dexrazoxane). The current thinking is that the ring-opened hydrolysis product of this agent, ADR-925, which is formed inside cardiomyocytes, removes iron from its complexes with anthracyclines, hereby reducing the concentration of highly toxic iron-anthracycline complexes that damage cardiomyocytes by semiquinone redox recycling and the production of free radicals. However, the 2 carbon linker ICRF-187 is also is a catalytic inhibitor of topoisomerase II, resulting in the risk of additional myelosuppression in patients receiving ICRF-187 as a cardioprotectant in combination with doxorubicin. The development of a topoisomerase II-inactive iron chelating compound thus appeared attractive. In the present paper we evaluate the topoisomerase II-inactive 3 carbon linker bisdioxopiperazine analog ICRF-161 as a cardioprotectant. We demonstrate that this compound does chelate iron and protects against doxorubicin-induced LDH release from primary rat cardiomyocytes in vitro, similarly to ICRF-187. The compound does not target topoisomerase II in vitro or in cells, it is well tolerated and shows similar exposure to ICRF-187 in rodents, and it does not induce myelosuppression when given at high doses to mice as opposed to ICRF-187. However, when tested in a model of chronic anthracycline-induced cardiomyopathy in spontaneously hypertensive rats, ICRF-161 was not capable of protecting against the cardiotoxic effects of doxorubicin. Modulation of the activity of the beta isoform of the topoisomerase II enzyme by ICRF-187 has recently been proposed as the mechanism behind its cardioprotection. This concept is thus supported by the present study in that iron chelation alone does not appear to be sufficient for protection against anthracycline-induced cardiomyopathy.


Asunto(s)
Antibióticos Antineoplásicos/antagonistas & inhibidores , Antibióticos Antineoplásicos/toxicidad , Antineoplásicos/farmacología , Cardiomiopatías/inducido químicamente , Cardiomiopatías/prevención & control , ADN-Topoisomerasas de Tipo II/metabolismo , Doxorrubicina/antagonistas & inhibidores , Doxorrubicina/toxicidad , Razoxano/farmacología , Animales , Animales Recién Nacidos , Antineoplásicos/farmacocinética , Cardiomiopatías/patología , Ensayo de Unidades Formadoras de Colonias , Crithidia fasciculata/metabolismo , ADN/efectos de los fármacos , Compuestos Férricos/farmacología , Estimación de Kaplan-Meier , L-Lactato Deshidrogenasa/metabolismo , Ratones , Mitocondrias Cardíacas/efectos de los fármacos , Miocardio/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Ratas , Ratas Endogámicas SHR , Razoxano/farmacocinética , Retículo Sarcoplasmático/efectos de los fármacos , Retículo Sarcoplasmático/patología , Troponina I/metabolismo
18.
Cancer Res ; 67(18): 8839-46, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17875725

RESUMEN

Doxorubicin is among the most effective and widely used anticancer drugs in the clinic. However, cardiotoxicity is one of the life-threatening side effects of doxorubicin-based therapy. Dexrazoxane (Zinecard, also known as ICRF-187) has been used in the clinic as a cardioprotectant against doxorubicin cardiotoxicity. The molecular basis for doxorubicin cardiotoxicity and the cardioprotective effect of dexrazoxane, however, is not fully understood. In the present study, we showed that dexrazoxane specifically abolished the DNA damage signal gamma-H2AX induced by doxorubicin, but not camptothecin or hydrogen peroxide, in H9C2 cardiomyocytes. Doxorubicin-induced DNA damage was also specifically abolished by the proteasome inhibitors bortezomib and MG132 and much reduced in top2beta(-/-) mouse embryonic fibroblasts (MEF) compared with TOP2beta(+/+) MEFs, suggesting the involvement of proteasome and DNA topoisomerase IIbeta (Top2beta). Furthermore, in addition to antagonizing Top2 cleavage complex formation, dexrazoxane also induced rapid degradation of Top2beta, which paralleled the reduction of doxorubicin-induced DNA damage. Together, our results suggest that dexrazoxane antagonizes doxorubicin-induced DNA damage through its interference with Top2beta, which could implicate Top2beta in doxorubicin cardiotoxicity. The specific involvement of proteasome and Top2beta in doxorubicin-induced DNA damage is consistent with a model in which proteasomal processing of doxorubicin-induced Top2beta-DNA covalent complexes exposes the Top2beta-concealed DNA double-strand breaks.


Asunto(s)
Roturas del ADN de Doble Cadena , Doxorrubicina/antagonistas & inhibidores , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Razoxano/farmacología , Animales , Antibióticos Antineoplásicos/antagonistas & inhibidores , Antibióticos Antineoplásicos/toxicidad , ADN-Topoisomerasas de Tipo II/química , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Doxorrubicina/toxicidad , Interacciones Farmacológicas , Cardiopatías/inducido químicamente , Cardiopatías/enzimología , Cardiopatías/prevención & control , Histonas/metabolismo , Humanos , Ratones , Modelos Moleculares , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Conformación Proteica , Inhibidores de Topoisomerasa II
19.
Eur J Cancer ; 44(3): 334-40, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18194856

RESUMEN

Nebivolol is a cardioselective beta-blocker (BB) currently used for the treatment of hypertension. It has mild vasodilating properties attributed to its interaction with the L-arginine/nitric oxide pathway, a property not shared by other BBs. Carvedilol is a nonselective ss-adrenergic receptor antagonist that also blocks alpha1-adrenergic receptors and is a potent antioxidant. Anthracyclines (ANTs), daunorubicin and doxorubicin, are commonly used in the treatment of several tumours, but their cardiac toxicity prevents their use at maximum myelotoxic doses, representing an important problem. In this study, we have evaluated the role of these BBs administered in combination with ANTs (daunorubicin and doxorubicin) on a reduction in cardiac toxicity. The combination of BB and ANTs has reduced the release of GSSG and GSH; in particular, co-treatment with nebivolol to ANTs has shown a significant reduction. The total integrated creatine kinase and troponin T activities were improved by BB and ANTs co-treatment. A significant reduction of their release was observed when hearts were treated with nebivolol. Cardiac tissue activity of gluthatione reductase was not significant and similar among experimental groups. In contrast, gluthatione peroxidise, Mn-superoxide dismutase and nitrite/nitrate release were increased after co-treatment with nebivolol. Finally, three parameters have been used to evaluate the cardiac toxicity of ANTs: the left ventricular pressure developed under a constant perfusion pressure (LVDP), the rate of variation of this parameter during systole (contractility) (LV/dt)max and during diastole (relaxation) (LV(dP/dt)min. Combination with BB has shown a reduction in cardiac toxicity; in particular, nebivolol has exerted the most significant cardioprotective effect.


Asunto(s)
Antagonistas Adrenérgicos beta/administración & dosificación , Antibióticos Antineoplásicos/antagonistas & inhibidores , Benzopiranos/administración & dosificación , Daunorrubicina/antagonistas & inhibidores , Etanolaminas/administración & dosificación , Cardiopatías/prevención & control , Análisis de Varianza , Animales , Antibióticos Antineoplásicos/toxicidad , Carbazoles/administración & dosificación , Carvedilol , Daunorrubicina/toxicidad , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos , Glutatión/metabolismo , Cardiopatías/inducido químicamente , Masculino , Nebivolol , Nitratos/metabolismo , Estrés Oxidativo , Propanolaminas/administración & dosificación , Ratas , Ratas Sprague-Dawley
20.
Eur J Heart Fail ; 10(1): 39-46, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18164246

RESUMEN

Contractile dysfunction and cardiomyopathies secondary to apoptotic cell death are limiting factors for treating cancer with doxorubicin. Inhibition of volume-sensitive chloride currents (I(Cl,vol)) has been reported to blunt doxorubicin-induced apoptosis in cardiomyocytes. To investigate cellular contractility during acute induction of apoptosis by doxorubicin and to determine whether I(Cl,vol) inhibitors are able to prevent the subsequent contractile dysfunction, electrically paced ventricular myocytes freshly isolated from adult rabbits were acutely exposed to doxorubicin in the presence and absence of I(Cl,vol) inhibitors IAA-94 or DIDS. Doxorubicin induced increases in both annexin V labelling and caspase-3 activity and decreases in cell volume. Alteration in cardiac contractility was observed after doxorubicin exposure. Both IAA-94 and DIDS abolished the doxorubicin-induced decreases in peak shortening and cell volume as well as the increases in caspase-3 activity and annexin V labelling. These protective effects of I(Cl,vol) inhibitors were abolished by previous inhibition of PI(3)kinase, Akt and Erk 1/2. Thus, I(Cl,vol) inhibitors prevent doxorubicin-induced apoptosis and subsequent contractile dysfunction through PI(3)kinase/Akt and Erk 1/2. Inhibition of I(Cl,vol) may represent a new pharmacological strategy for developing cytoprotective drugs against apoptotic cell death and contractile dysfunction.


Asunto(s)
Antibióticos Antineoplásicos/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Cardiomiopatías/prevención & control , Canales de Cloruro/antagonistas & inhibidores , Doxorrubicina/antagonistas & inhibidores , Contracción Miocárdica/efectos de los fármacos , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Animales , Antibióticos Antineoplásicos/efectos adversos , Doxorrubicina/efectos adversos , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Glicolatos/farmacología , Corazón/efectos de los fármacos , Masculino , Contracción Miocárdica/genética , Miocardio/enzimología , Miocitos Cardíacos/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Conejos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA