Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biofactors ; 42(2): 133-48, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26876101

RESUMEN

Tn is a unique translational biomarker in cardiology whose potential has not been diminished in the new era of high sensitive assays. cTns can be valuable markers in cardiac diseases as well as in infectious diseases and respiratory diseases. Furthermore, the role of cTns is growing in the routine evaluation of cardioxicity and in determining the efficacy/safety ratio of novel cardioprotective strategies in clinical settings. cTns can detect myocardial injury not only in a wide spectrum of laboratory animals in experimental studies in vivo, but also in isolated heart models or cardiomyocytes in vitro. The crucial issue regarding the cross-species usage of cardiac troponin investigation remains the choice of cardiac troponin testing. This review summarizes the recent proteomic data on aminoacid sequences of cTnT and cTnI in various species, as well as selected analytical characteristics of human cardiac troponin high-sensitivity assays. Due to the highly phylogenetically conserved structure of troponins, the same bioindicator can be investigated using the same method in both clinical and experimental cardiology, thus contributing to a better understanding of the pathogenesis of cardiac diseases as well as to increased effectiveness of troponin use in clinical practice. Measuring cardiac troponins using commercially available human high-sensitivity cardiac troponin tests with convenient antibodies selected on the basis of adequate proteomic knowledge can solve many issues which would otherwise be difficult to address in clinical settings for various ethical and practical reasons. Our survey could help elaborate the practical guidelines for optimizing the choice of cTns assay in cardiology.


Asunto(s)
Biomarcadores/metabolismo , Cardiopatías/diagnóstico , Troponina I/aislamiento & purificación , Troponina T/aislamiento & purificación , Bioensayo/métodos , Cardiopatías/metabolismo , Cardiopatías/patología , Humanos , Miocardio/metabolismo , Miocardio/patología , Proteómica , Troponina I/metabolismo , Troponina T/metabolismo
2.
J Mol Cell Cardiol ; 91: 92-103, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26724189

RESUMEN

Dexrazoxane (DEX) is a clinically available cardioprotectant that reduces the toxicity induced by anthracycline (ANT) anticancer drugs; however, DEX is seldom used and its action is poorly understood. Inorganic nitrate/nitrite has shown promising results in myocardial ischemia-reperfusion injury and recently in acute high-dose ANT cardiotoxicity. However, the utility of this approach for overcoming clinically more relevant chronic forms of cardiotoxicity remains elusive. Hence, in this study, the protective potential of inorganic nitrate and nitrite against chronic ANT cardiotoxicity was investigated, and the results were compared to those using DEX. Chronic cardiotoxicity was induced in rabbits with daunorubicin (DAU). Sodium nitrate (1g/L) was administered daily in drinking water, while sodium nitrite (0.15 or 5mg/kg) or DEX (60mg/kg) was administered parenterally before each DAU dose. Although oral nitrate induced a marked increase in plasma NOx, it showed no improvement in DAU-induced mortality, myocardial damage or heart failure. Instead, the higher nitrite dose reduced the incidence of end-stage cardiotoxicity, prevented related premature deaths and significantly ameliorated several molecular and cellular perturbations induced by DAU, particularly those concerning mitochondria. The latter result was also confirmed in vitro. Nevertheless, inorganic nitrite failed to prevent DAU-induced cardiac dysfunction and molecular remodeling in vivo and failed to overcome the cytotoxicity of DAU to cardiomyocytes in vitro. In contrast, DEX completely prevented all of the investigated molecular, cellular and functional perturbations that were induced by DAU. Our data suggest that the difference in cardioprotective efficacy between DEX and inorganic nitrite may be related to their different abilities to address a recently proposed upstream target for ANT in the heart - topoisomerase IIß.


Asunto(s)
Cardiotónicos/farmacología , Cardiotoxicidad/prevención & control , Dexrazoxano/farmacología , Nitratos/farmacología , Nitrito de Sodio/farmacología , Animales , Antibióticos Antineoplásicos/efectos adversos , Cardiotoxicidad/metabolismo , Cardiotoxicidad/patología , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Daunorrubicina/efectos adversos , Esquema de Medicación , Infusiones Intravenosas , Masculino , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Conejos
3.
Int J Cardiol ; 201: 358-67, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26310978

RESUMEN

BACKGROUND: Cardiac troponins (cTns) seem to be more sensitive for the detection of anthracycline cardiotoxicity than the currently recommended method of monitoring LV systolic function. However, the optimal timing of blood sampling remains unknown. Hence, the aims of the present study were to determine the precise diagnostic window for cTns during the development of chronic anthracycline cardiotoxicity and to evaluate their predictive value. METHODS: Cardiotoxicity was induced in rabbits with daunorubicin (3mg/kg, weekly, for 8 weeks). Blood samples were collected 2-168 h after the 1st, 5th and 8th drug administrations, and concentrations of cTns were determined using highly sensitive assays: hs cTnT (Roche) and hs cTnI (Abbott). RESULTS: The plasma levels of cTns progressively increased with the rising number of chemotherapy cycles. While only a mild non-significant increase in both cTn levels occurred after the first daunorubicin dose, a significant rise was observed after the 5th and 8th administrations. Two hours after these administrations, a significant increase occurred with a peak between 4-6h and a decline until 24h. Discrete cTn release continued even after cessation of the therapy. While greater variability of cTn levels was observed around the peak concentrations, the values did not correspond well with the severity of LV systolic dysfunction. Unlike AMI in cardiotoxicity, cTn elevations may be better associated with cumulative dose and concentrations at steady state than cmax. CONCLUSIONS: To the best of our knowledge, this is the first study to precisely describe the diagnostic window and predictive value of cTns in anthracycline cardiotoxicity.


Asunto(s)
Antraciclinas/toxicidad , Cardiotoxicidad/sangre , Troponina I/sangre , Troponina T/sangre , Animales , Antibióticos Antineoplásicos/toxicidad , Biomarcadores/sangre , Biomarcadores/metabolismo , Cardiomiopatías/sangre , Cardiomiopatías/inducido químicamente , Cardiotoxicidad/diagnóstico por imagen , Daunorrubicina/toxicidad , Modelos Animales de Enfermedad , Ecocardiografía , Corazón/efectos de los fármacos , Corazón/fisiología , Masculino , Valor Predictivo de las Pruebas , Conejos , Análisis de Regresión , Sístole/efectos de los fármacos , Sístole/fisiología
4.
PLoS One ; 9(5): e96055, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24804796

RESUMEN

Chronic anthracycline cardiotoxicity is a serious clinical issue with well characterized functional and histopathological hallmarks. However, molecular determinants of the toxic damage and associated myocardial remodeling remain to be established. Furthermore, details on the different propensity of the left and right ventricle (LV and RV, respectively) to the cardiotoxicity development are unknown. Hence, the aim of the investigation was to study molecular changes associated with remodeling of the LV and RV in chronic anthracycline cardiotoxicity and post-treatment follow up. The cardiotoxicity was induced in rabbits with daunorubicin (3 mg/kg/week for 10 weeks) and animals were sacrificed either at the end of the treatment or after an additional 10 weeks. Daunorubicin induced severe and irreversible cardiotoxicity associated with LV dysfunction and typical morphological alterations, whereas the myocardium of the RV showed only mild changes. Both ventricles also showed different expression of ANP after daunorubicin treatment. Daunorubicin impaired the expression of several sarcomeric proteins in the LV, which was not the case of the RV. In particular, a significant drop was found in titin and thick filament proteins at both mRNA and protein level and this might be connected with persistent LV down-regulation of GATA-4. In addition, the LV was more affected by treatment-induced perturbations in calcium handling proteins. LV cardiomyocytes showed marked up-regulation of desmin after the treatment and vimentin was mainly induced in LV fibroblasts, whereas only weaker changes were observed in the RV. Remodeling of extracellular matrix was almost exclusively found in the LV with particular induction of collagen I and IV. Hence, the present study describes profound molecular remodeling of myocytes, non-myocyte cells and extracellular matrix in response to chronic anthracycline treatment with marked asymmetry between LV and RV.


Asunto(s)
Antraciclinas/toxicidad , Miocardio/metabolismo , Remodelación Ventricular/fisiología , Animales , Western Blotting , Daunorrubicina/farmacología , Ecocardiografía , Inmunohistoquímica , Filamentos Intermedios/metabolismo , Masculino , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción/metabolismo , Troponina T/metabolismo , Remodelación Ventricular/efectos de los fármacos
5.
Toxicology ; 311(3): 191-204, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23831762

RESUMEN

Despite incomplete understanding to its mechanism of action, dexrazoxane (DEX) is still the only clearly effective cardioprotectant against chronic anthracycline (ANT) cardiotoxicity. However, its clinical use is currently restricted to patients exceeding significant ANT cumulative dose (300mg/m(2)), although each ANT cycle may induce certain potentially irreversible myocardial damage. Therefore, the aim of this study was to compare early and delayed DEX intervention against chronic ANT cardiotoxicity and study the molecular events involved. The cardiotoxicity was induced in rabbits with daunorubicin (DAU; 3mg/kg/week for 10 weeks); DEX (60mg/kg) was administered either before the 1st or 7th DAU dose (i.e. after ≈300mg/m(2) cumulative dose). While both DEX administration schedules prevented DAU-induced premature deaths and severe congestive heart failure, only the early intervention completely prevented the left ventricular dysfunction, myocardial morphological changes and mitochondrial damage. Further molecular analyses did not support the assumption that DEX cardioprotection is based and directly proportional to protection from DAU-induced oxidative damage and/or deletions in mtDNA. Nevertheless, DAU induced significant up-regulation of heme oxygenase 1 pathway while heme synthesis was inversely regulated and both changes were schedule-of-administration preventable by DEX. Early and delayed DEX interventions also differed in ability to prevent DAU-induced down-regulation of expression of mitochondrial proteins encoded by both nuclear and mitochondrial genome. Hence, the present functional, morphological as well as the molecular data highlights the enormous cardioprotective effects of DEX and provides novel insights into the molecular events involved. Furthermore, the data suggests that currently recommended delayed intervention may not be able to take advantage of the full cardioprotective potential of the drug.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Cardiotónicos/administración & dosificación , Daunorrubicina/toxicidad , Cardiopatías/prevención & control , Razoxano/administración & dosificación , Animales , Citrato (si)-Sintasa/metabolismo , Cardiopatías/inducido químicamente , Cardiopatías/metabolismo , Cardiopatías/patología , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Conejos , Troponina T/sangre , Función Ventricular Izquierda/efectos de los fármacos
6.
Antioxid Redox Signal ; 18(8): 899-929, 2013 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-22794198

RESUMEN

SIGNIFICANCE: Anthracyclines (doxorubicin, daunorubicin, or epirubicin) rank among the most effective anticancer drugs, but their clinical usefulness is hampered by the risk of cardiotoxicity. The most feared are the chronic forms of cardiotoxicity, characterized by irreversible cardiac damage and congestive heart failure. Although the pathogenesis of anthracycline cardiotoxicity seems to be complex, the pivotal role has been traditionally attributed to the iron-mediated formation of reactive oxygen species (ROS). In clinics, the bisdioxopiperazine agent dexrazoxane (ICRF-187) reduces the risk of anthracycline cardiotoxicity without a significant effect on response to chemotherapy. The prevailing concept describes dexrazoxane as a prodrug undergoing bioactivation to an iron-chelating agent ADR-925, which may inhibit anthracycline-induced ROS formation and oxidative damage to cardiomyocytes. RECENT ADVANCES: A considerable body of evidence points to mitochondria as the key targets for anthracycline cardiotoxicity, and therefore it could be also crucial for effective cardioprotection. Numerous antioxidants and several iron chelators have been tested in vitro and in vivo with variable outcomes. None of these compounds have matched or even surpassed the effectiveness of dexrazoxane in chronic anthracycline cardiotoxicity settings, despite being stronger chelators and/or antioxidants. CRITICAL ISSUES: The interpretation of many findings is complicated by the heterogeneity of experimental models and frequent employment of acute high-dose treatments with limited translatability to clinical practice. FUTURE DIRECTIONS: Dexrazoxane may be the key to the enigma of anthracycline cardiotoxicity, and therefore it warrants further investigation, including the search for alternative/complementary modes of cardioprotective action beyond simple iron chelation.


Asunto(s)
Antraciclinas/efectos adversos , Quelantes/farmacología , Corazón/efectos de los fármacos , Metales/efectos adversos , Miocardio/metabolismo , Estrés Oxidativo , Transducción de Señal , Antraciclinas/química , Antraciclinas/farmacología , Antineoplásicos/efectos adversos , Antineoplásicos/química , Antineoplásicos/farmacología , Antioxidantes/química , Antioxidantes/farmacología , Cardiotónicos/efectos adversos , Cardiotónicos/química , Cardiotónicos/farmacología , Quelantes/efectos adversos , Quelantes/química , Humanos , Oxidación-Reducción , Razoxano/efectos adversos , Razoxano/química , Razoxano/farmacología , Especies Reactivas de Oxígeno/metabolismo
7.
Can J Physiol Pharmacol ; 90(9): 1303-10, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22913659

RESUMEN

Dexrazoxane (DEX), an inhibitor of topoisomerase II and intracellular iron chelator, is believed to reduce the formation of reactive oxygen species (ROS) and protects the heart from the toxicity of anthracycline antineoplastics. As ROS also play a role in the pathogenesis of cardiac ischaemia/reperfusion (I/R) injury, the aim was to find out whether DEX can improve cardiac ischaemic tolerance. DEX in a dose of 50, 150, or 450 mg·(kg body mass)(-1) was administered intravenously to rats 60 min before ischaemia. Myocardial infarct size and ventricular arrhythmias were assessed in anaesthetized open-chest animals subjected to 20 min coronary artery occlusion and 3 h reperfusion. Arrhythmias induced by I/R were also assessed in isolated perfused hearts. Only the highest dose of DEX significantly reduced infarct size from 53.9% ± 4.7% of the area at risk in controls to 37.5% ± 4.3% without affecting the myocardial markers of oxidative stress. On the other hand, the significant protective effect against reperfusion arrhythmias occurred only in perfused hearts with the dose of DEX of 150 mg·kg(-1), which also tended to limit the incidence of ischaemic arrhythmias. It is concluded that DEX in a narrow dose range can suppress arrhythmias in isolated hearts subjected to I/R, while a higher dose is needed to limit myocardial infarct size in open-chest rats.


Asunto(s)
Arritmias Cardíacas/prevención & control , Fármacos Cardiovasculares/uso terapéutico , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Razoxano/uso terapéutico , Enfermedad Aguda , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Fármacos Cardiovasculares/administración & dosificación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Electrocardiografía , Técnicas In Vitro , Masculino , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/metabolismo , Perfusión , Ratas , Ratas Wistar , Razoxano/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Resultado del Tratamiento
8.
J Pharmacol Exp Ther ; 343(2): 468-78, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22915767

RESUMEN

Anthracycline anticancer drugs (e.g., doxorubicin or daunorubicin) can induce chronic cardiotoxicity and heart failure (HF), both of which are believed to be based on oxidative injury and mitochondrial damage. In this study, molecular and functional changes induced by chronic anthracycline treatment with progression into HF in post-treatment follow-up were analyzed with special emphasis on nuclear factor erythroid 2-related factor 2 (Nrf2) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α) pathways. Chronic cardiotoxicity was induced in rabbits with daunorubicin (3 mg/kg, weekly for 10 weeks), and the animals were followed for another 10 weeks. Echocardiography revealed a significant drop in left ventricular (LV) systolic function during the treatment with marked progression to LV dilation and congestive HF in the follow-up. Although daunorubicin-induced LV lipoperoxidation was found, it was only loosely associated with cardiac performance. Furthermore, although LV oxidized glutathione content was increased, the oxidized-to-reduced glutathione ratio itself remained unchanged. Neither Nrf2, the master regulator of antioxidant response, nor the majority of its target genes showed up-regulation in the study. However, down-regulation of manganese superoxide dismutase and NAD(P)H dehydrogenase [quinone] 1 were observed together with heme oxygenase 1 up-regulation. Although marked perturbations in mitochondrial functions were found, no induction of PGC1α-controlled mitochondrial biogenesis pathway was revealed. Instead, especially in the post-treatment period, an impaired regulation of this pathway was observed along with down-regulation of the expression of mitochondrial genes. These results imply that global oxidative stress need not be a factor responsible for the development of anthracycline-induced HF, whereas suppression of mitochondrial biogenesis might be involved.


Asunto(s)
Antraciclinas/toxicidad , Antibióticos Antineoplásicos/toxicidad , Cardiopatías/inducido químicamente , Cardiopatías/metabolismo , Mitocondrias Cardíacas/metabolismo , Factor 2 Relacionado con NF-E2/biosíntesis , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Daunorrubicina/farmacología , Ecocardiografía , Glutatión/metabolismo , Pruebas de Función Cardíaca , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Mitocondrias Cardíacas/efectos de los fármacos , Miocardio/patología , Estrés Oxidativo/efectos de los fármacos , Conejos , Reacción en Cadena en Tiempo Real de la Polimerasa , Sobrevida , Factores de Transcripción/metabolismo , Troponina T/metabolismo
9.
Histol Histopathol ; 26(5): 551-62, 2011 05.
Artículo en Inglés | MEDLINE | ID: mdl-21432770

RESUMEN

Anthracyclines are one of the most effective anticancer drugs ever developed, but their clinical use has been hampered by the risk of severe cardiotoxicity. In this study, we investigated whether rabbits exposed to a different cumulative dose of anthracycline suffer from immunohistochemically detectable vascular toxicity and endothelial dysfunction. Daunorubicin (3 mg/kg, i.e. 50 mg/m²) was administered i.v. to rabbits once weekly for 1-10 weeks to reach different cumulative doses of the drug (50-500 mg/m²), while control rabbits received saline. The rabbits were sacrificed either 24 hours or 7 days after reaching each particular cumulative dose, and aortas and right femoral arteries were collected for immunohistochemical analysis. Immunohistochemical analysis showed ICAM-1 staining in many aortas from both saline and daunorubicin-treated rabbits without any relationship to the anthracycline treatment. On the contrary, unlike in the lipopolysaccharide-treated or hypercholesterolemic rabbits, no distinct immunoreactivity for other markers of inflammation, oxidative and nitrosative stress (VCAM-1, 4-HNE, iNOS and nitrotyrosine) were detected in aortas and femoral arteries from either control or daunorubicin-treated animals. No relationship to the cumulative dose of the drug or post-expose set up of harvesting was found. In this study, we have demonstrated that daunorubicin does not induce gross histopathological changes in the studied arteries and it fails to induce immunohistochemically detectable endothelial dysfunction. Thus, we propose that endothelial cells are much less susceptible to anthracycline toxicity than cardiac myocytes. In addition, our data suggest that vascular toxicity of anthracyclines plays rather a minor role in the cardiovascular complications of anthracycline chemotherapy.


Asunto(s)
Aorta/metabolismo , Daunorrubicina/farmacología , Células Endoteliales/citología , Arteria Femoral/metabolismo , Animales , Antraciclinas/farmacología , Antibióticos Antineoplásicos/farmacología , Aorta/efectos de los fármacos , Apoptosis , Inmunohistoquímica/métodos , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Miocitos Cardíacos/efectos de los fármacos , Selectina-P/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Conejos , Enfermedades Vasculares/inducido químicamente
10.
J Mol Cell Cardiol ; 50(5): 849-62, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21284945

RESUMEN

Chronic anthracycline cardiotoxicity is a feared complication of cancer chemotherapy. However, despite several decades of primarily hypothesis-driven research, the molecular basis of this phenomenon remains poorly understood. The aim of this study was to obtain integrative molecular insights into chronic anthracycline cardiotoxicity and the resulting heart failure. Cardiotoxicity was induced in rabbits (daunorubicin 3mg/kg, weekly, 10weeks) and changes in the left ventricular proteome were analyzed by 2D-DIGE. The protein spots with significant changes (p<0.01, >1.5-fold) were identified using MALDI-TOF/TOF. Key data were corroborated by immunohistochemistry, qRT-PCR and enzyme activity determination and compared with functional, morphological and biochemical data. The most important alterations were found in mitochondria - especially in proteins crucial for oxidative phosphorylation, energy channeling, antioxidant defense and mitochondrial stress. Furthermore, the intermediate filament desmin, which interacts with mitochondria, was determined to be distinctly up-regulated and disorganized in its expression pattern. Interestingly, the latter changes reflected the intensity of toxic damage in whole hearts as well as in individual cells. In addition, a marked drop in myosin light chain isoforms, activation of proteolytic machinery (including the proteasome system), increased abundance of chaperones and proteins involved in chaperone-mediated autophagy, membrane repair as well as apoptosis were found. In addition, dramatic changes in proteins of basement membrane and extracellular matrix were documented. In conclusion, for the first time, the complex proteomic signature of chronic anthracycline cardiotoxicity was revealed which enhances our understanding of the basis for this phenomenon and it may enhance efforts in targeting its reduction.


Asunto(s)
Antraciclinas/toxicidad , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/metabolismo , Ventrículos Cardíacos/metabolismo , Miocardio/metabolismo , Animales , Western Blotting , Daunorrubicina/toxicidad , Ecocardiografía , Electroforesis en Gel Bidimensional , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Ventrículos Cardíacos/efectos de los fármacos , Inmunohistoquímica , Malondialdehído/metabolismo , Proteínas Mitocondriales/metabolismo , Proteómica , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Troponina I/metabolismo , Vimentina/metabolismo
11.
Arch Toxicol ; 85(5): 525-35, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21046361

RESUMEN

The clinical usefulness of anthracycline antineoplastic drugs is limited by their cardiotoxicity. Its mechanisms have not been fully understood, although the induction of oxidative stress is widely believed to play the principal role. Glutathione is the dominant cellular antioxidant, while glutathione peroxidase (GPx) together with glutathione reductase (GR) constitutes the major enzymatic system protecting the cardiac cells from oxidative damage. Therefore, this study aimed to assess their roles in anthracycline cardiotoxicity. Ten-week intravenous administration of daunorubicin (DAU, 3 mg/kg weekly) to rabbits induced heart failure, which was evident from decreased left ventricular ejection fraction and release of cardiac troponins to circulation. However, no significant changes in either total or oxidized glutathione contents or GR activity were detected in left ventricular tissue of DAU-treated rabbits when compared with control animals. GPx activity in the cardiac tissue significantly increased. In H9c2 rat cardiac cells, 24-h DAU exposure (0.1-10 µM) induced significant dose-dependent toxicity. Cellular content of reduced glutathione was insignificantly decreased, oxidized glutathione and GR activity were unaffected, and GPx activity was significantly increased. Neither buthionine sulfoximine (BSO, glutathione biosynthesis inhibitor) nor 2-oxo-4-thiazolidine-carboxylic acid (OTC, glutathione biosynthetic precursor) had significant effects on DAU cytotoxicity. This contrasted with model oxidative injury induced by hydrogen peroxide, which cytotoxicity was increased by BSO and decreased by OTC. In conclusion, our results suggest that the dysfunction of glutathione antioxidant system does not play a causative role in anthracycline cardiotoxicity.


Asunto(s)
Antioxidantes/farmacología , Daunorrubicina/efectos adversos , Glutatión/farmacología , Cardiopatías/inducido químicamente , Animales , Antibióticos Antineoplásicos , Butionina Sulfoximina/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Glutatión Peroxidasa/metabolismo , Glutatión Reductasa/metabolismo , Corazón/efectos de los fármacos , Peróxido de Hidrógeno/toxicidad , Masculino , Modelos Animales , Estrés Oxidativo/efectos de los fármacos , Ácido Pirrolidona Carboxílico/metabolismo , Conejos , Ratas , Tiazolidinas/metabolismo
12.
Pharmacol Rep ; 61(1): 154-71, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19307704

RESUMEN

The risk of cardiotoxicity is the most serious drawback to the clinical usefulness of anthracycline antineoplastic antibiotics, which include doxorubicin (adriamycin), daunorubicin or epirubicin. Nevertheless, these compounds remain among the most widely used anticancer drugs. The molecular pathogenesis of anthracycline cardiotoxicity remains highly controversial, although the oxidative stress-based hypothesis involving intramyocardial production of reactive oxygen species (ROS) has gained the widest acceptance. Anthracyclines may promote the formation of ROS through redox cycling of their aglycones as well as their anthracycline-iron complexes. This proposed mechanism has become particularly popular in light of the high cardioprotective efficacy of dexrazoxane (ICRF-187). The mechanism of action of this drug has been attributed to its hydrolytic transformation into the iron-chelating metabolite ADR-925, which may act by displacing iron from anthracycline-iron complexes or by chelating free or loosely bound cellular iron, thus preventing site-specific iron-catalyzed ROS damage. However, during the last decade, calls for the critical reassessment of this "ROS and iron" hypothesis have emerged. Numerous antioxidants, although efficient in cellular or acute animal experiments, have failed to alleviate anthracycline cardiotoxicity in clinically relevant chronic animal models or clinical trials. In addition, studies with chelators that are stronger and more selective for iron than ADR-925 have also yielded negative or, at best, mixed outcomes. Hence, several lines of evidence suggest that mechanisms other than the traditionally emphasized "ROS and iron" hypothesis are involved in anthracycline-induced cardiotoxicity and that these alternative mechanisms may be better bases for designing approaches to achieve efficient and safe cardioprotection.


Asunto(s)
Antraciclinas/efectos adversos , Antibióticos Antineoplásicos/efectos adversos , Cardiopatías/inducido químicamente , Animales , Antraciclinas/farmacología , Antibióticos Antineoplásicos/farmacología , Ensayos Clínicos como Asunto , Cardiopatías/fisiopatología , Humanos , Hierro/metabolismo , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Factores de Riesgo
13.
J Pharmacol Exp Ther ; 326(1): 259-69, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18434588

RESUMEN

Anthracycline cardiotoxicity ranks among the most severe complications of cancer chemotherapy. Although its pathogenesis is only incompletely understood, "reactive oxygen species (ROS) and iron" hypothesis has gained the widest acceptance. Besides dexrazoxane, novel oral iron chelator deferiprone has been recently reported to afford significant cardioprotection in both in vitro and ex vivo conditions. Therefore, the aim of this study was to assess whether deferiprone 1) has any effect on the anticancer action of daunorubicin and 2) whether it can overcome or significantly reduce the chronic anthracycline cardiotoxicity in the in vivo rabbit model (daunorubicin, 3 mg/kg i.v., weekly for 10 weeks). First, using the leukemic cell line, deferiprone (1-300 microM) was shown not to blunt the antiproliferative effect of daunorubicin. Instead, in clinically relevant concentrations (>10 microM), deferiprone augmented the antiproliferative action of daunorubicin. However, deferiprone (10 or 50 mg/kg administered p.o. before each daunorubicin dose) failed to afford significant protection against daunorubicin-induced mortality, left ventricular lipoperoxidation, cardiac dysfunction, and morphological cardiac deteriorations, as well as an increase in plasma cardiac troponin T. Hence, this first in vivo study changes the current view on deferiprone as a potential cardioprotectant against anthracycline cardiotoxicity. In addition, these results, together with our previous findings, further suggest that the role of iron and its chelation in anthracycline cardiotoxicity is not as trivial as originally believed and/or other mechanisms unrelated to iron-catalyzed ROS production are involved.


Asunto(s)
Antraciclinas/administración & dosificación , Antraciclinas/toxicidad , Cardiopatías/inducido químicamente , Piridonas/uso terapéutico , Animales , Antraciclinas/antagonistas & inhibidores , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Cardiotónicos/uso terapéutico , Cardiotónicos/toxicidad , Proliferación Celular/efectos de los fármacos , Daunorrubicina/antagonistas & inhibidores , Deferiprona , Células HL-60 , Cardiopatías/mortalidad , Cardiopatías/patología , Humanos , Masculino , Conejos
14.
Hemoglobin ; 32(1-2): 207-15, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18274998

RESUMEN

The risk of cardiotoxicity is the main drawback of anthracycline antibiotics. However, these drugs remain among the most effective and frequently used anti cancer drugs. In this study we aimed to assess the cardioprotective effects of aroylhydrazone iron (FE) chelators: pyridoxal isonicotinoyl hydrazone (PIH) and its two analogs: salicyladehyde isonicotinoyl hydrazone (SIH) and pyridoxal o-chlorbenzoyl hydrazone (o-108). In rabbits, chronic treatment with daunorubicin (DAU) (3 mg/kg weekly for 10 weeks) induced mortality (33%) as well as left ventricular (LV) dysfunction. Co-administrations of PIH (25 mg/kg, i.p.), SIH hydrochloride [1 mg/kg, iv] as well as o-108 (10 mg/kg, i.p.), fully prevented premature deaths and most of the DAU-induced functional impairments were significantly suppressed. However, when 2- to 2.5-fold higher doses of the chelators were used, they led to rather paradoxical and mostly negative results regarding both cardioprotection and overall mortality.


Asunto(s)
Cardiotónicos/uso terapéutico , Daunorrubicina/toxicidad , Cardiopatías/inducido químicamente , Cardiopatías/prevención & control , Quelantes del Hierro/uso terapéutico , Isoniazida/análogos & derivados , Piridoxal/análogos & derivados , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/toxicidad , Cardiotónicos/administración & dosificación , Cardiotónicos/química , Daunorrubicina/administración & dosificación , Cardiopatías/mortalidad , Quelantes del Hierro/administración & dosificación , Quelantes del Hierro/química , Isoniazida/administración & dosificación , Isoniazida/química , Isoniazida/uso terapéutico , Piridoxal/administración & dosificación , Piridoxal/química , Piridoxal/uso terapéutico , Conejos
15.
Acta Medica (Hradec Kralove) ; 50(2): 109-11, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18035747

RESUMEN

Matrix metalloproteinases (MMPs), activated by oxidative stress, play a key role during cardiac remodeling. In the present study we aimed to assess the role of MMPs in experimental cardiomyopathy induced by repeated 10-week administration of daunorubicin (3 mg/kg i.v.) to rabbits. In the daunorubicin group, the plasma cardiac troponin T levels (cTnT - a marker of myocardial necrosis) were significantly increased (p<0.05), commencing with the 8th administration compared with the controls. The amount of collagen (an estimate of fibrosis) was also significantly higher in the daunorubicin group (13.39 +/- 0.97 mg/g wet weight) compared to the control group (10.03 +/- 0.65 mg/g wet weight). In both groups, the LV MMP-activity was observed only in the gelatine substrate in the 70 kDa region (MMP-2), while no MMPs activities were detectable either in the casein or collagen containing zymograms. At the end of the experiment, the MMP-2 activity was slightly up-regulated (by 16 %) compared with the controls.


Asunto(s)
Cardiomiopatías/enzimología , Daunorrubicina/toxicidad , Metaloproteinasas de la Matriz/metabolismo , Animales , Cardiomiopatías/inducido químicamente , Cardiomiopatías/metabolismo , Cardiomiopatías/fisiopatología , Colágeno/metabolismo , Conejos , Troponina T/metabolismo , Remodelación Ventricular
16.
Cardiovasc Toxicol ; 7(2): 145-50, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17652820

RESUMEN

The use of anthracycline anticancer drugs is limited by a cumulative, dose-dependent cardiac toxicity. Iron chelation has long been considered as a promising strategy to limit this unfavorable side effect, either by restoring the disturbed cellular iron homeostasis or by removing redox-active iron, which may promote anthracycline-induced oxidative stress. Aroylhydrazone lipophilic iron chelators have shown promising results in the rabbit model of daunorubicin-induced cardiomyopathy as well as in cellular models. The lack of interference with the antiproliferative effects of the anthracyclines also favors their use in clinical settings. The dose, however, should be carefully titrated to prevent iron depletion, which apparently also applies for other strong iron chelators. We have shown that a mere ability of a compound to chelate iron is not the sole determinant of a good cardioprotector and the protective potential does not directly correlate with the ability of the chelators to prevent hydroxyl radical formation. These findings, however, do not weaken the role of iron in doxorubicin cardiotoxicity as such, they rather appeal for further investigations into the molecular mechanisms how anthracyclines interact with iron and how iron chelation may interfere with these processes.


Asunto(s)
Antraciclinas/toxicidad , Antibióticos Antineoplásicos/toxicidad , Cardiotónicos , Cardiopatías/inducido químicamente , Cardiopatías/metabolismo , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Hierro/fisiología , Animales , Antraciclinas/efectos adversos , Antibióticos Antineoplásicos/efectos adversos , Cardiopatías/prevención & control , Humanos , Quelantes del Hierro/efectos adversos , Estrés Oxidativo/efectos de los fármacos
17.
Toxicology ; 237(1-3): 218-228, 2007 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-17587482

RESUMEN

Cardiac troponin T (cTnT) and troponin I (cTnI) are becoming acknowledged as useful biochemical markers of drug-induced cardiotoxicity. In this study we examined the release kinetics of cTnT and cTnI using an in vitro model of isolated rat neonatal ventricular cardiomyocytes (NVCM, 72h treatment with 0.1-3microM of daunorubicin) and compared it with data from a rabbit model of chronic anthracycline-induced cardiomyopathy in vivo (3mg/kg of daunorubicin weekly, 10 weeks). In cell-culture media, the cTnI and cTnT concentrations were concentration- and time-dependently increasing in response to daunorubicin exposure and were negatively exponentially related to cardiomyocyte viability. With 3microM daunorubicin, the relative increase of AUC of cTnT and cTnI was 2.4- and 5.3-fold higher than the increase of LDH activity, respectively. In rabbits, the daunorubicin-induced cardiomyopathy was associated with progressive increase of both cTnT and cTnI. Although the correlation between cTnT and cTnI cumulative release (AUCs) was found (R=0.81; P<0.01) and both cardiac troponins corresponded well with the echocardiographically-assessed systolic dysfunction (R=0.83 and 0.81 for cTnT and cTnI, respectively; P<0.001), the first significant increase in cTnI levels was observed earlier (at a cumulative daunorubicin dose of 200mg/m(2)) than with cTnT (350mg/m(2)). In conclusion, our study has confirmed cTnT and cTnI as very sensitive and specific markers of anthracycline-induced cardiotoxicity. The troponins can become not only the bridge between the clinical and experimental studies of drug-induced cardiotoxicity but also the linkage between the preclinical experiments in vitro and in vivo.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Cardiomiopatías/inducido químicamente , Daunorrubicina/efectos adversos , Miocitos Cardíacos , Troponina I/sangre , Troponina T/sangre , Animales , Animales Recién Nacidos , Biomarcadores/sangre , Western Blotting , Cardiomiopatías/sangre , Cardiomiopatías/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Conejos , Ratas
18.
Toxicology ; 235(3): 150-66, 2007 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-17459556

RESUMEN

Pyridoxal-derived aroylhydrazone iron chelators have been previously shown as effective cardioprotectants against chronic anthracycline cardiotoxicity. In this study we focused on a novel salicylaldehyde analogue (salicylaldehyde isonicotinoyl hydrazone, SIH), which has been recently demonstrated to possess marked and dose-dependent protective effects against oxidative injury of cardiomyocytes. Therefore, in the present study the cardioprotective potential of SIH against daunorubicin (DAU) cardiotoxicity was assessed in vitro (isolated rat ventricular cardiomyocytes; DAU 10 microM, 48 h exposure) as well as in vivo (chronic DAU-induced cardiomyopathy in rabbits; DAU 3mg/kg, i.v. weekly, 10 weeks). In vitro, SIH (3-100 microM) was able to partially, but significantly decrease the LDH leakage from cardiomyocytes. In vivo, SIH co-administration was capable to reduce (SIH dose of 0.5mg/kg, i.v.) or even to completely prevent (1.0mg/kg, i.v.) the DAU-induced mortality. Moreover, the latter dose of the chelator significantly improved the left ventricular function (LV dP/dt(max)=1185+/-80 kPa/s versus 783+/-53 kPa/s in the DAU group; P<0.05) and decreased the severity of the myocardial morphological changes as well as the plasma levels of cardiac troponin T. Unfortunately, further escalation of the SIH dose (to 2.5mg/kg) resulted in a nearly complete reversal of the protective effects as judged by the overall mortality, functional, morphological as well as biochemical examinations. Hence, this study points out that aroylhydrazone iron chelators can induce a significant cardioprotection against anthracycline cardiotoxicity; however, they share the curious dose-response relationship which is unrelated to the chemical structure or the route of the administration of the chelator.


Asunto(s)
Aldehídos/farmacología , Cardiopatías/prevención & control , Hidrazonas/farmacología , Quelantes del Hierro/farmacología , Animales , Células Cultivadas , Daunorrubicina , Cardiopatías/inducido químicamente , Cardiopatías/patología , Cardiopatías/fisiopatología , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Masculino , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Conejos , Ratas , Troponina T/sangre
19.
J Pharmacol Exp Ther ; 319(3): 1336-47, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17003229

RESUMEN

Iron chelation is the only pharmacological intervention against anthracycline cardiotoxicity whose effectiveness has been well documented both experimentally and clinically. In this study, we aimed to assess whether pyridoxal 2-chlorobenzoyl hydrazone (o-108, a strong iron chelator) can provide effective protection against daunorubicin (DAU)-induced chronic cardiotoxicity in rabbits. First, using the HL-60 leukemic cell line, it was shown that o-108 has no potential to blunt the antiproliferative efficacy of DAU. Instead, o-108 itself moderately inhibited cell proliferation. In vivo, chronic DAU treatment (3 mg/kg weekly for 10 weeks) induced mortality (33%), left ventricular (LV) dysfunction, a troponin T rise, and typical morphological LV damage. In contrast, all animals treated with 10 mg/kg o-108 before DAU survived without a significant drop in the LV ejection fraction (63.2 +/- 0.5 versus 59.2 +/- 1.0%, beginning versus end, not significant), and their cardiac contractility (dP/dt(max)) was significantly higher than in the DAU-only group (1131 +/- 125 versus 783 +/- 53 kPa/s, p < 0.05), which corresponded with histologically assessed lower extent and intensity of myocardial damage. Although higher o-108 dose (25 mg/kg) was well tolerated when administered alone, in combination with DAU it led to rather paradoxical and mostly negative results regarding both cardioprotection and overall mortality. In conclusion, we show that shielding of free intracellular iron using a potent lipophilic iron chelator is able to offer a meaningful protection against chronic anthracycline cardiotoxicity. However, this approach lost its potential with the higher chelator dose, which suggests that iron might play more complex role in the pathogenesis of this disease than previously assumed.


Asunto(s)
Antibióticos Antineoplásicos , Cardiotónicos , Daunorrubicina , Cardiopatías/inducido químicamente , Cardiopatías/prevención & control , Hidrazonas/farmacología , Quelantes del Hierro/farmacología , Piridoxal/análogos & derivados , Animales , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Gasto Cardíaco/efectos de los fármacos , División Celular/efectos de los fármacos , Chinchilla , Electrocardiografía/efectos de los fármacos , Células HL-60 , Cardiopatías/patología , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Contracción Miocárdica/efectos de los fármacos , Miocardio/metabolismo , Miocardio/patología , Piridoxal/farmacología , Conejos , Volumen Sistólico/efectos de los fármacos , Troponina T/metabolismo , Función Ventricular Izquierda/efectos de los fármacos
20.
Artículo en Inglés | MEDLINE | ID: mdl-16725389

RESUMEN

A high performance liquid chromatographic method for the determination of a biocompatible iron chelator, pyridoxal 2-chlorobenzoyl hydrazone (o-108), in rabbit plasma was developed and validated. The separation was achieved on a C18 column with the mobile phase composed of a mixture of 0.01 M phosphate buffer (pH 6) with the addition of EDTA (2 mM), methanol and acetonitrile (42:24:14; v/v/v). The method was validated with respect to selectivity, linearity (0.8-150 microg/mL), intra- and inter-day variability and stability. This method was successfully applied to the analysis of the samples obtained from a pilot pharmacokinetic experiment, in which the chelator was administered intravenously to rabbits.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Hidrazonas/sangre , Quelantes del Hierro/análisis , Animales , Área Bajo la Curva , Estabilidad de Medicamentos , Hidrazonas/química , Hidrazonas/farmacocinética , Quelantes del Hierro/química , Quelantes del Hierro/farmacocinética , Masculino , Estructura Molecular , Proyectos Piloto , Piridoxal/análogos & derivados , Piridoxal/sangre , Piridoxal/farmacocinética , Conejos , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA