Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Clin Sci (Lond) ; 136(1): 139-161, 2022 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-34878093

RESUMEN

Angiotensin-converting enzyme inhibitors (ACEis) have been used to treat anthracycline (ANT)-induced cardiac dysfunction, and they appear beneficial for secondary prevention in high-risk patients. However, it remains unclear whether they truly prevent ANT-induced cardiac damage and provide long-lasting cardioprotection. The present study aimed to examine the cardioprotective effects of perindopril on chronic ANT cardiotoxicity in a rabbit model previously validated with the cardioprotective agent dexrazoxane (DEX) with focus on post-treatment follow-up (FU). Chronic cardiotoxicity was induced by daunorubicin (DAU; 3 mg/kg/week for 10 weeks). Perindopril (0.05 mg/kg/day) was administered before and throughout chronic DAU treatment. After the completion of treatment, significant benefits were observed in perindopril co-treated animals, particularly full prevention of DAU-induced mortality and prevention or significant reductions in cardiac dysfunction, plasma cardiac troponin T (cTnT) levels, morphological damage, and most of the myocardial molecular alterations. However, these benefits significantly waned during 3 weeks of drug-free FU, which was not salvageable by administering a higher perindopril dose. In the longer (10-week) FU period, further worsening of left ventricular function and morphological damage occurred together with heart failure (HF)-related mortality. Continued perindopril treatment in the FU period did not reverse this trend but prevented HF-related mortality and reduced the severity of the progression of cardiac damage. These findings contrasted with the robust long-lasting protection observed previously for DEX in the same model. Hence, in the present study, perindopril provided only temporary control of ANT cardiotoxicity development, which may be associated with the lack of effects on ANT-induced and topoisomerase II ß (TOP2B)-dependent DNA damage responses in the heart.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Cardiotoxicidad/prevención & control , Daunorrubicina/efectos adversos , Perindopril/uso terapéutico , Animales , Antibióticos Antineoplásicos , Cardiotoxicidad/tratamiento farmacológico , Cardiopatías/inducido químicamente , Cardiopatías/prevención & control , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/mortalidad , Masculino , Conejos , Troponina T/sangre
2.
Clin Sci (Lond) ; 135(15): 1897-1914, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34318878

RESUMEN

The anthracycline (ANT) anticancer drugs such as doxorubicin or daunorubicin (DAU) can cause serious myocardial injury and chronic cardiac dysfunction in cancer survivors. A bisdioxopiperazine agent dexrazoxane (DEX) has been developed as a cardioprotective drug to prevent these adverse events, but it is uncertain whether it is the best representative of the class. The present study used a rabbit model of chronic ANT cardiotoxicity to examine another bisdioxopiperazine compound called GK-667 (meso-(butane-2,3-diylbis(2,6-dioxopiperazine-4,1-diyl))bis(methylene)-bis(2-aminoacetate) hydrochloride), a water-soluble prodrug of ICRF-193 (meso-4,4'-(butan-2,3-diyl)bis(piperazine-2,6-dione)), as a potential cardioprotectant. The cardiotoxicity was induced by DAU (3 mg/kg, intravenously, weekly, 10 weeks), and GK-667 (1 or 5 mg/kg, intravenously) was administered before each DAU dose. The treatment with GK-667 was well tolerated and provided full protection against DAU-induced mortality and left ventricular (LV) dysfunction (determined by echocardiography and LV catheterization). Markers of cardiac damage/dysfunction revealed minor cardiac damage in the group co-treated with GK-667 in the lower dose, whereas almost full protection was achieved with the higher dose. This was associated with similar prevention of DAU-induced dysregulation of redox and calcium homeostasis proteins. GK-667 dose-dependently prevented tumor suppressor p53 (p53)-mediated DNA damage response in the LV myocardium not only in the chronic experiment but also after single DAU administration. These effects appear essential for cardioprotection, presumably because of the topoisomerase IIß (TOP2B) inhibition provided by its active metabolite ICRF-193. In addition, GK-667 administration did not alter the plasma pharmacokinetics of DAU and its main metabolite daunorubicinol (DAUol) in rabbits in vivo. Hence, GK-667 merits further investigation as a promising drug candidate for cardioprotection against chronic ANT cardiotoxicity.


Asunto(s)
Cardiomiopatías/prevención & control , Daño del ADN , Dicetopiperazinas/farmacología , Miocitos Cardíacos/efectos de los fármacos , Profármacos/farmacología , Inhibidores de Topoisomerasa II/farmacología , Disfunción Ventricular Izquierda/prevención & control , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Cardiomiopatías/inducido químicamente , Cardiomiopatías/metabolismo , Cardiomiopatías/fisiopatología , Cardiotoxicidad , Enfermedad Crónica , Daunorrubicina , Modelos Animales de Enfermedad , Fibrosis , Células HL-60 , Humanos , Masculino , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Conejos , Proteína p53 Supresora de Tumor/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología
3.
J Pharmacol Exp Ther ; 373(3): 402-415, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32253261

RESUMEN

Bisdioxopiperazine agent dexrazoxane (ICRF-187) has been the only effective and approved drug for prevention of chronic anthracycline cardiotoxicity. However, the structure-activity relationships (SARs) of its cardioprotective effects remain obscure owing to limited investigation of its derivatives/analogs and uncertainties about its mechanism of action. To fill these knowledge gaps, we tested the hypothesis that dexrazoxane derivatives exert cardioprotection via metal chelation and/or modulation of topoisomerase IIß (Top2B) activity in chronic anthracycline cardiotoxicity. Dexrazoxane was alkylated in positions that should not interfere with the metal-chelating mechanism of cardioprotective action; that is, on dioxopiperazine imides or directly on the dioxopiperazine ring. The protective effects of these agents were assessed in vitro in neonatal cardiomyocytes. All studied modifications of dexrazoxane molecule, including simple methylation, were found to abolish the cardioprotective effects. Because this challenged the prevailing mechanistic concept and previously reported data, the two closest derivatives [(±)-4,4'-(propane-1,2-diyl)bis(1-methylpiperazine-2,6-dione) and 4-(2-(3,5-dioxopiperazin-1-yl)ethyl)-3-methylpiperazine-2,6-dione] were thoroughly scrutinized in vivo using a rabbit model of chronic anthracycline cardiotoxicity. In contrast to dexrazoxane, both compounds failed to protect the heart, as demonstrated by mortality, cardiac dysfunction, and myocardial damage parameters, although the pharmacokinetics and metal-chelating properties of their metabolites were comparable to those of dexrazoxane. The loss of cardiac protection was shown to correlate with their abated potential to inhibit and deplete Top2B both in vitro and in vivo. These findings suggest a very tight SAR between bisdioxopiperazine derivatives and their cardioprotective effects and support Top2B as a pivotal upstream druggable target for effective cardioprotection against anthracycline cardiotoxicity. SIGNIFICANCE STATEMENT: This study has revealed the previously unexpected tight structure-activity relationships of cardioprotective effects in derivatives of dexrazoxane, which is the only drug approved for the prevention of cardiomyopathy and heart failure induced by anthracycline anticancer drugs. The data presented in this study also strongly argue against the importance of metal-chelating mechanisms for the induction of this effect and support the viability of topoisomerase IIß as an upstream druggable target for effective and clinically translatable cardioprotection.


Asunto(s)
Antraciclinas/efectos adversos , Cardiotoxicidad/tratamiento farmacológico , ADN-Topoisomerasas de Tipo II/metabolismo , Dexrazoxano/farmacología , Corazón/efectos de los fármacos , Sustancias Protectoras/farmacología , Inhibidores de Topoisomerasa II/farmacología , Animales , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/metabolismo , Línea Celular Tumoral , Células HL-60 , Humanos , Masculino , Modelos Animales , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Conejos , Ratas , Ratas Wistar , Relación Estructura-Actividad
4.
Clin Sci (Lond) ; 133(16): 1827-1844, 2019 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-31409729

RESUMEN

Although proteasome inhibitors (PIs) are modern targeted anticancer drugs, they have been associated with a certain risk of cardiotoxicity and heart failure (HF). Recently, PIs have been combined with anthracyclines (ANTs) to further boost their anticancer efficacy. However, this raised concerns regarding cardiac safety, which were further supported by several in vitro studies on immature cardiomyocytes. In the present study, we investigated the toxicity of clinically used PIs alone (bortezomib (BTZ), carfilzomib (CFZ)) as well as their combinations with an ANT (daunorubicin (DAU)) in both neonatal and adult ventricular cardiomyocytes (NVCMs and AVCMs) and in a chronic rabbit model of DAU-induced HF. Using NVCMs, we found significant cytotoxicity of both PIs around their maximum plasma concentration (cmax) as well as significant augmentation of DAU cytotoxicity. In AVCMs, BTZ did not induce significant cytotoxicity in therapeutic concentrations, whereas the toxicity of CFZ was significant and more profound. Importantly, neither PI significantly augmented the cardiotoxicity of DAU despite even more profound proteasome-inhibitory activity in AVCMs compared with NVCMs. Furthermore, in young adult rabbits, no significant augmentation of chronic ANT cardiotoxicity was noted with respect to any functional, morphological, biochemical or molecular parameter under study, despite significant inhibition of myocardial proteasome activity. Our experimental data show that combination of PIs with ANTs is not accompanied by an exaggerated risk of cardiotoxicity and HF in young adult animal cardiomyocytes and hearts.


Asunto(s)
Antraciclinas/toxicidad , Antineoplásicos/toxicidad , Cardiotoxicidad/etiología , Inhibidores de Proteasoma/toxicidad , Animales , Antraciclinas/administración & dosificación , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Bortezomib/administración & dosificación , Bortezomib/toxicidad , Daunorrubicina/administración & dosificación , Daunorrubicina/toxicidad , Relación Dosis-Respuesta a Droga , Masculino , Miocitos Cardíacos/efectos de los fármacos , Oligopéptidos/administración & dosificación , Oligopéptidos/toxicidad , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/administración & dosificación , Conejos , Ratas , Ratas Wistar
5.
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
6.
Front Pharmacol ; 14: 1298172, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38235109

RESUMEN

Background: Anthracycline cardiotoxicity is a well-known complication of cancer treatment, and miRNAs have emerged as a key driver in the pathogenesis of cardiovascular diseases. This study aimed to investigate the expression of miRNAs in the myocardium in early and late stages of chronic anthracycline induced cardiotoxicity to determine whether this expression is associated with the severity of cardiac damage. Method: Cardiotoxicity was induced in rabbits via daunorubicin administration (daunorubicin, 3 mg/kg/week; for five and 10 weeks), while the control group received saline solution. Myocardial miRNA expression was first screened using TaqMan Advanced miRNA microfluidic card assays, after which 32 miRNAs were selected for targeted analysis using qRT-PCR. Results: The first subclinical signs of cardiotoxicity (significant increase in plasma cardiac troponin T) were observed after 5 weeks of daunorubicin treatment. At this time point, 10 miRNAs (including members of the miRNA-34 and 21 families) showed significant upregulation relative to the control group, with the most intense change observed for miRNA-1298-5p (29-fold change, p < 0.01). After 10 weeks of daunorubicin treatment, when a further rise in cTnT was accompanied by significant left ventricle systolic dysfunction, only miR-504-5p was significantly (p < 0.01) downregulated, whereas 10 miRNAs were significantly upregulated relative to the control group; at this time-point, the most intense change was observed for miR-34a-5p (76-fold change). Strong correlations were found between the expression of multiple miRNAs (including miR-34 and mir-21 family and miR-1298-5p) and quantitative indices of toxic damage in both the early and late phases of cardiotoxicity development. Furthermore, plasma levels of miR-34a-5p were strongly correlated with the myocardial expression of this miRNA. Conclusion: To the best of our knowledge, this is the first study that describes alterations in miRNA expression in the myocardium during the transition from subclinical, ANT-induced cardiotoxicity to an overt cardiotoxic phenotype; we also revealed how these changes in miRNA expression are strongly correlated with quantitative markers of cardiotoxicity.

7.
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
8.
Circ Heart Fail ; 14(11): e008209, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34551586

RESUMEN

BACKGROUND: Anthracycline-induced heart failure has been traditionally attributed to direct iron-catalyzed oxidative damage. Dexrazoxane (DEX)-the only drug approved for its prevention-has been believed to protect the heart via its iron-chelating metabolite ADR-925. However, direct evidence is lacking, and recently proposed TOP2B (topoisomerase II beta) hypothesis challenged the original concept. METHODS: Pharmacokinetically guided study of the cardioprotective effects of clinically used DEX and its chelating metabolite ADR-925 (administered exogenously) was performed together with mechanistic experiments. The cardiotoxicity was induced by daunorubicin in neonatal ventricular cardiomyocytes in vitro and in a chronic rabbit model in vivo (n=50). RESULTS: Intracellular concentrations of ADR-925 in neonatal ventricular cardiomyocytes and rabbit hearts after treatment with exogenous ADR-925 were similar or exceeded those observed after treatment with the parent DEX. However, ADR-925 did not protect neonatal ventricular cardiomyocytes against anthracycline toxicity, whereas DEX exhibited significant protective effects (10-100 µmol/L; P<0.001). Unlike DEX, ADR-925 also had no significant impact on daunorubicin-induced mortality, blood congestion, and biochemical and functional markers of cardiac dysfunction in vivo (eg, end point left ventricular fractional shortening was 32.3±14.7%, 33.5±4.8%, 42.7±1.0%, and 41.5±1.1% for the daunorubicin, ADR-925 [120 mg/kg]+daunorubicin, DEX [60 mg/kg]+daunorubicin, and control groups, respectively; P<0.05). DEX, but not ADR-925, inhibited and depleted TOP2B and prevented daunorubicin-induced genotoxic damage. TOP2B dependency of the cardioprotective effects was probed and supported by experiments with diastereomers of a new DEX derivative. CONCLUSIONS: This study strongly supports a new mechanistic paradigm that attributes clinically effective cardioprotection against anthracycline cardiotoxicity to interactions with TOP2B but not metal chelation and protection against direct oxidative damage.


Asunto(s)
Antraciclinas/farmacología , Cardiotoxicidad/prevención & control , Dexrazoxano/farmacología , Insuficiencia Cardíaca/tratamiento farmacológico , Inhibidores de Topoisomerasa II/metabolismo , Antraciclinas/efectos adversos , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/farmacología , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/metabolismo , ADN-Topoisomerasas de Tipo II/efectos adversos , ADN-Topoisomerasas de Tipo II/metabolismo , Daunorrubicina/metabolismo , Daunorrubicina/farmacología , Dexrazoxano/efectos adversos , Cardiopatías/tratamiento farmacológico , Humanos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/efectos de los fármacos
9.
Chem Res Toxicol ; 22(1): 208-17, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19172757

RESUMEN

Iron (Fe) chelators are used clinically for the treatment of Fe overload disease. Iron also plays a role in the pathology of many other conditions, and these potentially include the cardiotoxicity induced by catecholamines such as isoprenaline (ISO). The current study examined the potential of Fe chelators to prevent ISO cardiotoxicity. This was done as like other catecholamines, ISO contains the classical catechol moiety that binds Fe and may form redox-active and cytotoxic Fe complexes. Studies in vitro used the cardiomyocyte cell line, H9c2, which was treated with ISO in the presence or absence of the chelator, desferrioxamine (DFO), or the lipophilic ligand, 2-pyridylcarboxaldehyde 2-thiophenecarboxyl hydrazone (PCTH). Both of these chelators were not cardiotoxic and significantly reduced ISO cardiotoxicity in vitro. However, PCTH was far more effective than DFO, with the latter showing activity only at a high, clinically unachievable concentration. Further studies in vitro showed that interaction of ISO with Fe(II)/(III) did not increase cytotoxic radical generation, suggesting that this mechanism was not involved. Studies in vivo were initiated using rats pretreated intravenously with DFO or PCTH before subcutaneous administration of ISO (100 mg/kg). DFO at a clinically used dose (50 mg/kg) failed to reduce catecholamine cardiotoxicity, while PCTH at an equimolar dose totally prevented catecholamine-induced mortality and reduced cardiotoxicity. This study demonstrates that PCTH reduced ISO-induced cardiotoxicity in vitro and in vivo, demonstrating that Fe plays a role, in part, in the pathology observed.


Asunto(s)
Quelantes del Hierro/farmacología , Isoproterenol/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Tiofenos/farmacología , Animales , Catecolaminas/antagonistas & inhibidores , Catecolaminas/metabolismo , Catecolaminas/toxicidad , Línea Celular , Deferoxamina/administración & dosificación , Hierro/metabolismo , Isoproterenol/antagonistas & inhibidores , Isoproterenol/metabolismo , Masculino , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Ratas , Ratas Wistar
10.
Biometals ; 22(2): 353-61, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18982411

RESUMEN

Lactoferrin is recently under intense investigation because of its proposed several pharmacologically positive effects. Based on its iron-binding properties and its physiological presence in the human body, it may have a significant impact on pathological conditions associated with iron-catalysed reactive oxygen species (ROS). Its effect on a catecholamine model of myocardial injury, which shares several pathophysiological features with acute myocardial infarction (AMI) in humans, was examined. Male Wistar rats were randomly divided into four groups according to the received medication: control (saline), isoprenaline (ISO, 100 mg kg(-1) s.c.), bovine lactoferrin (La, 50 mg kg(-1) i.v.) or a combination of La + ISO in the above-mentioned doses. After 24 h, haemodynamic functional parameters were measured, a sample of blood was withdrawn and the heart was removed for analysis of various parameters. Lactoferrin premedication reduced some impairment caused by ISO (e.g. a stroke volume decrease, an increase in peripheral resistance and calcium overload). These positive effects were likely to have been mediated by the positive inotropic effect of lactoferrin and by inhibition of ROS formation due to chelation of free iron. The failure of lactoferrin to provide higher protection seems to be associated with the complexity of catecholamine cardiotoxicity and with its hydrophilic character.


Asunto(s)
Catecolaminas/metabolismo , Lactoferrina/química , Animales , Bovinos , Quelantes/farmacología , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Hemodinámica , Hierro/química , Masculino , Infarto del Miocardio/metabolismo , Proyectos Piloto , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno
11.
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
12.
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
13.
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
14.
Toxicology ; 372: 52-63, 2016 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-27816693

RESUMEN

Chronic anthracycline (ANT) cardiotoxicity is a serious complication of cancer chemotherapy. Molsidomine, a NO-releasing drug, has been found cardioprotective in different models of I/R injury and recently in acute high-dose ANT cardiotoxicity. Hence, we examined whether its cardioprotective effects are translatable to chronic ANT cardiotoxicity settings without induction of nitrosative stress and interference with antiproliferative action of ANTs. The effects of molsidomine (0.025 and 0.5mg/kg, i.v.) were studied on the well-established model of chronic ANT cardiotoxicity in rabbits (daunorubicin/DAU/3mg/kg/week for 10 weeks). Molsidomine was unable to significantly attenuate mortality, development of heart failure and morphological damage induced by DAU. Molsidomine did not alter DAU-induced myocardial lipoperoxidation, MnSOD down-regulation, up-regulation of HO-1, IL-6, and molecular markers of cardiac remodeling. Although molsidomine increased 3-nitrotyrosine in the myocardium, this event had no impact on cardiotoxicity development. Using H9c2 myoblasts and isolated cardiomyocytes, it was found that SIN-1 (an active metabolite of molsidomine) induces significant protection against ANT toxicity, but only at high concentrations. In leukemic HL-60 cells, SIN-1 initially augmented ANT cytotoxicity (in low and clinically achievable concentrations), but it protected these cells against ANT in the high concentrations. UHPLC-MS/MS investigation demonstrated that the loss of ANT cytotoxicity after co-incubation of the cells in vitro with high concentrations of SIN-1 is caused by unexpected chemical depletion of DAU molecule. The present study demonstrates that cardioprotective effects of molsidomine are not translatable to clinically relevant chronic form of ANT cardiotoxicity. The augmentation of antineoplastic effects of ANT in low (nM) concentrations may deserve further study.


Asunto(s)
Antraciclinas/toxicidad , Antibióticos Antineoplásicos/toxicidad , Cardiotónicos/farmacología , Cardiopatías/inducido químicamente , Cardiopatías/prevención & control , Molsidomina/farmacología , Donantes de Óxido Nítrico/farmacología , Animales , Cardiotoxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Enfermedad Crónica , Daunorrubicina/toxicidad , Doxorrubicina/toxicidad , Insuficiencia Cardíaca/prevención & control , Peroxidación de Lípido/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Conejos , Especies Reactivas de Oxígeno/metabolismo , Remodelación Ventricular/efectos de los fármacos
15.
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
16.
Eur J Heart Fail ; 6(4): 377-87, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15182761

RESUMEN

BACKGROUND: Cardiac toxicity associated with chronic administration of anthracycline (ANT) antibiotics represents a serious complication of their use in anticancer chemotherapy, but can also serve as a useful experimental model of cardiomyopathy and congestive heart failure. AIMS: In this study, a model of chronic ANT cardiotoxicity induced by repeated i.v. daunorubicin (DAU) administration to rabbits was tested. METHODS: Three groups of animals were used: (1) control group-10 animals received i.v. saline; (2) 11 animals received DAU (3 mg/kg, i.v.); (3) 5 animals received the model cardioprotective agent dexrazoxane (DEX, 60 mg/kg, i.p.), 30 min prior to DAU. All substances were administered once weekly, for 10 weeks. The DAU-induced heart damage and protective action of DEX were determined and quantitated with the use of histopathology, invasive haemodynamic measurements (e.g. left ventricular pressure changes-dP/dt(max), dP/dt(min)), non-invasive systolic function examinations (left ventricular ejection fraction, PEP/LVET index) and biochemical analysis of cardiac troponin T plasma concentrations. RESULTS: All the employed methods showed unambiguously pronounced heart impairment in the DAU group, with the development of both systolic and diastolic heart failure, as well as significant reduction of DAU-cardiotoxicity in DEX-pretreated animals. Other toxicities were acceptable. CONCLUSION: The presented model has been approved to be consistent and reliable and it can serve as a basis for future determinations and comparisons of chronic cardiotoxic effects of various drugs, as well as for the evaluation of potential cardioprotectants.


Asunto(s)
Antraciclinas/efectos adversos , Fármacos Cardiovasculares/uso terapéutico , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/prevención & control , Animales , Antraciclinas/administración & dosificación , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/efectos adversos , Biomarcadores/sangre , Peso Corporal/efectos de los fármacos , Fármacos Cardiovasculares/administración & dosificación , Daunorrubicina/administración & dosificación , Daunorrubicina/efectos adversos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ecocardiografía , Índices de Eritrocitos/efectos de los fármacos , Insuficiencia Cardíaca/mortalidad , Insuficiencia Cardíaca/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Masculino , Modelos Cardiovasculares , Miocardio/citología , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Conejos , Razoxano/administración & dosificación , Razoxano/uso terapéutico , Volumen Sistólico/efectos de los fármacos , Factores de Tiempo
17.
Cancer Chemother Pharmacol ; 52(5): 431-4, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14618274

RESUMEN

PURPOSE: Cardiac troponin T (cTnT) plasma concentration is considered a useful marker of anthracycline-induced cardiomyopathy. In this study we used daunorubicin-treated Chinchilla rabbits as a model to investigate the relationship between left ventricular contractility and cTnT plasma concentrations. METHODS: Two groups of animals were used: a control group (n=8) received i.v. saline, and an experimental group (n=11) received daunorubicin (3 mg/kg, i.v.). The substances were administered once weekly for 10 weeks, and 5-7 days after the last administration, left ventricular cardiac contractility (dP/dtmax) was invasively measured as a contractility index and blood was sampled for cTnT concentration determination (Elecsys Troponin T STAT immunoassay). RESULTS: Cardiac contractility was significantly lower in seven surviving daunorubicin-treated animals than in control animals (745.7+/-69.3 vs 1393.4+/-25.5 kPa/s; P<0.001), while cTnT plasma concentrations were significantly increased (medians 0.278 vs 0.000 ng/ml; P<0.001). When the dP/dtmax values of individual daunorubicin-treated animals were plotted against the corresponding cTnT plasma concentrations, a close negative linear correlation was found (R=-0.910; P<0.005; regression equation: dP/dtmax=-1861*cTnT+1234). CONCLUSIONS: This study suggests that determination of cTnT plasma levels, which is simple and inexpensive, could be used in anthracycline-treated patients for left ventricular systolic function assessment and contractility estimation.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Cardiomiopatías/inducido químicamente , Cardiomiopatías/metabolismo , Miocardio/metabolismo , Troponina T/metabolismo , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/metabolismo , Animales , Biomarcadores , Chinchilla , Daunorrubicina/toxicidad , Masculino , Contracción Miocárdica/efectos de los fármacos , Conejos , Función Ventricular Izquierda/efectos de los fármacos
18.
Acta Histochem ; 104(4): 375-9, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12553706

RESUMEN

The proliferative activity in the subependymal layer of lateral brain ventricles in adulthood is known. We were interested in the reaction of this layer to ibotenic acid lesions, which simulate neurodegenerative processes in Huntington's disease. Animals with a unilateral ibotenic acid lesion were compared with sham-lesioned animals and control animals with intact brains at 5 and 13 weeks after surgery. Five weeks after surgery, increased proliferation was found in most GFAP-positive astrocytes and to a lesser extent in CNPase-positive oligodendrocytes in comparison with controls. Interestingly, a slight increase in proliferation was found as well in the contralateral non-lesioned hemispheres. Moderate elevation of cell proliferation was found after induction of sham-lesions as well. The intensity of the reaction in the subependymal layer decreased in the following 8 weeks. Only a few scattered cells that originated from the subependymal layer had migrated over a short distance to adjacent brain tissue. We conclude that the reaction of the subependymal layer is (a) non-specific, as it is a response to any type of lesion, and (b) slowly decreases in time.


Asunto(s)
Epéndimo/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/toxicidad , Ácido Iboténico/toxicidad , Ventrículos Laterales/efectos de los fármacos , 2',3'-Nucleótido Cíclico Fosfodiesterasas/análisis , Animales , Astrocitos/química , Astrocitos/efectos de los fármacos , Astrocitos/patología , Bromodesoxiuridina/análisis , Epéndimo/química , Epéndimo/patología , Proteína Ácida Fibrilar de la Glía/análisis , Inmunohistoquímica , Antígeno Ki-67/análisis , Ventrículos Laterales/química , Ventrículos Laterales/patología , Oligodendroglía/química , Oligodendroglía/efectos de los fármacos , Oligodendroglía/patología , Antígeno Nuclear de Célula en Proliferación/análisis , Ratas , Ratas Long-Evans
19.
Exp Toxicol Pathol ; 56(3): 181-7, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15625787

RESUMEN

We studied the dose response of pulmonary changes at 3 weeks after 1-25 Gy irradiation and we investigated the effects of an anti-inflammatory drug. Wistar rats were given a single dose of 1-25Gy irradiation to the thorax. Group one was treated with saline only, while group two was administered subcutaneously a combination of pentoxifylline (35 mg/kg) and dexamethasone (1 mg/kg) twice per week. Lungs were examined histochemically and number of neutrophile granulocytes, alveolar septal thickness, air/tissue ratio, number of alveoli per field, number of type II pneumocytes per alveolus, and occludin 1 expression were measured. A significant dose-dependent depletion of type II pneumocytes was found after irradiation with a dose of 1 Gy and higher. Alveolar neutrophils increased after 1 Gy with a dose dependency noted after 10-25Gy and alveolar septa thickening followed 5-25 Gy. A lower occludin 1 expression was observed in animals irradiated with the doses of 5 20 Gy, indicating an effect on vascular permeability. Anti-inflammatory therapy partially inhibited the increase of neutrophils at all radiation doses and the depletion of type II pneumocytes after doses of 1, 10, and 15 Gy. Occludin 1 did not decrease in the lungs of rats treated with the anti-inflammatory drugs as it did in most rats treated only with saline. Our results suggest that pneumocytes depletion is a major factor responsible for radiation pneumonitis development and that these changes may be compensated for provided radiation doses are below the threshold.


Asunto(s)
Permeabilidad Capilar/efectos de la radiación , Alveolos Pulmonares/patología , Neumonitis por Radiación/patología , Animales , Antiinflamatorios/uso terapéutico , Dexametasona/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta en la Radiación , Quimioterapia Combinada , Técnicas para Inmunoenzimas , Técnicas In Vitro , Masculino , Proteínas de la Membrana/metabolismo , Neutrófilos/patología , Neutrófilos/efectos de la radiación , Ocludina , Pentoxifilina/uso terapéutico , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/efectos de la radiación , Neumonitis por Radiación/tratamiento farmacológico , Protectores contra Radiación/uso terapéutico , Ratas , Ratas Wistar
20.
Hum Exp Toxicol ; 21(12): 649-57, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12540035

RESUMEN

This study compares the chronic toxicity of two anthracyclines--daunorubicin and doxorubicin, commonly used for induction of anthracycline cardiomyopathy in the rabbit model. Such a comparative study has not been published until now. Both drugs were administered intravenously to male Chinchilla rabbits in doses at 3 mg/kg (50 mg/m2) once weekly for 10 weeks. Selected biochemical, haematological and cardiovascular parameters and body weights were regularly monitored; additionally, a histological evaluation of heart, kidney and liver was performed at the end of the experiment. In the daunorubicin group, there were marked signs of the progressive development of heart failure, like the significant increases of the pre-ejection period/left ventricular ejection time index values (up to 134%)--and histological changes within the myocardium were also observed. On the other hand, the 10-week doxorubicin administration did not cause these changes that are typical for heart injury. Haematotoxicity, manifested particularly by aplastic anaemia, was apparent in both the experimental groups. Significant body weight loss (by 45.2%) and high premature mortality (100% versus 36.4%) reflected a greater general toxicity, especially nephrotoxicity of doxorubicin in comparison with daunorubicin. Further studies are necessary to find a possible explanation for these findings.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Daunorrubicina/toxicidad , Doxorrubicina/toxicidad , Animales , Peso Corporal/efectos de los fármacos , Pruebas de Química Clínica , Modelos Animales de Enfermedad , Corazón/efectos de los fármacos , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/patología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Hemodinámica/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Miocardio/patología , Conejos , Pruebas de Toxicidad Crónica , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA