Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicol Sci ; 198(2): 288-302, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38290791

RESUMO

Anthracyclines, such as doxorubicin (adriamycin), daunorubicin, or epirubicin, rank among the most effective agents in classical anticancer chemotherapy. However, cardiotoxicity remains the main limitation of their clinical use. Topoisomerase IIß has recently been identified as a plausible target of anthracyclines in cardiomyocytes. We examined the putative topoisomerase IIß selective agent XK469 as a potential cardioprotective and designed several new analogs. In our experiments, XK469 inhibited both topoisomerase isoforms (α and ß) and did not induce topoisomerase II covalent complexes in isolated cardiomyocytes and HL-60, but induced proteasomal degradation of topoisomerase II in these cell types. The cardioprotective potential of XK469 was studied on rat neonatal cardiomyocytes, where dexrazoxane (ICRF-187), the only clinically approved cardioprotective, was effective. Initially, XK469 prevented daunorubicin-induced toxicity and p53 phosphorylation in cardiomyocytes. However, it only partially prevented the phosphorylation of H2AX and did not affect DNA damage measured by Comet Assay. It also did not compromise the daunorubicin antiproliferative effect in HL-60 leukemic cells. When administered to rabbits to evaluate its cardioprotective potential in vivo, XK469 failed to prevent the daunorubicin-induced cardiac toxicity in either acute or chronic settings. In the following in vitro analysis, we found that prolonged and continuous exposure of rat neonatal cardiomyocytes to XK469 led to significant toxicity. In conclusion, this study provides important evidence on the effects of XK469 and its combination with daunorubicin in clinically relevant doses in cardiomyocytes. Despite its promising characteristics, long-term treatments and in vivo experiments have not confirmed its cardioprotective potential.


Assuntos
Antraciclinas , Quinoxalinas , Inibidores da Topoisomerase II , Ratos , Animais , Coelhos , Inibidores da Topoisomerase II/toxicidade , Inibidores da Topoisomerase II/uso terapêutico , Antraciclinas/toxicidade , Antraciclinas/uso terapêutico , Cardiotoxicidade , Daunorrubicina/toxicidade , Daunorrubicina/uso terapêutico , Doxorrubicina/toxicidade , Antibióticos Antineoplásicos/toxicidade , DNA Topoisomerases Tipo II/metabolismo , DNA Topoisomerases Tipo II/uso terapêutico , Dano ao DNA
2.
Circ Heart Fail ; 14(11): e008209, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34551586

RESUMO

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.


Assuntos
Antraciclinas/farmacologia , Cardiotoxicidade/prevenção & controle , Dexrazoxano/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Inibidores da Topoisomerase II/metabolismo , Antraciclinas/efeitos adversos , Antibióticos Antineoplásicos/efeitos adversos , Antibióticos Antineoplásicos/farmacologia , Cardiotoxicidade/tratamento farmacológico , Cardiotoxicidade/metabolismo , DNA Topoisomerases Tipo II/efeitos adversos , DNA Topoisomerases Tipo II/metabolismo , Daunorrubicina/metabolismo , Daunorrubicina/farmacologia , Dexrazoxano/efeitos adversos , Cardiopatias/tratamento farmacológico , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Estresse Oxidativo/efeitos dos fármacos
3.
Clin Sci (Lond) ; 135(15): 1897-1914, 2021 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-34318878

RESUMO

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.


Assuntos
Cardiomiopatias/prevenção & controle , Dano ao DNA , Dicetopiperazinas/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Pró-Fármacos/farmacologia , Inibidores da Topoisomerase II/farmacologia , Disfunção Ventricular Esquerda/prevenção & controle , Função Ventricular Esquerda/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/metabolismo , Cardiomiopatias/fisiopatologia , Cardiotoxicidade , Doença Crônica , Daunorrubicina , Modelos Animais de Doenças , Fibrose , Células HL-60 , Humanos , Masculino , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Coelhos , Proteína Supressora de Tumor p53/metabolismo , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/fisiopatologia
4.
J Med Chem ; 64(7): 3997-4019, 2021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33750129

RESUMO

Cardioprotective activity of dexrazoxane (ICRF-187), the only clinically approved drug against anthracycline-induced cardiotoxicity, has traditionally been attributed to its iron-chelating metabolite. However, recent experimental evidence suggested that the inhibition and/or depletion of topoisomerase IIß (TOP2B) by dexrazoxane could be cardioprotective. Hence, we evaluated a series of dexrazoxane analogues and found that their cardioprotective activity strongly correlated with their interaction with TOP2B in cardiomyocytes, but was independent of their iron chelation ability. Very tight structure-activity relationships were demonstrated on stereoisomeric forms of 4,4'-(butane-2,3-diyl)bis(piperazine-2,6-dione). In contrast to its rac-form 12, meso-derivative 11 (ICRF-193) showed a favorable binding mode to topoisomerase II in silico, inhibited and depleted TOP2B in cardiomyocytes more efficiently than dexrazoxane, and showed the highest cardioprotective efficiency. Importantly, the observed ICRF-193 cardioprotection did not interfere with the antiproliferative activity of anthracycline. Hence, this study identifies ICRF-193 as the new lead compound in the development of efficient cardioprotective agents.


Assuntos
Cardiotônicos/uso terapêutico , Cardiotoxicidade/tratamento farmacológico , Piperazinas/uso terapêutico , Inibidores da Topoisomerase II/uso terapêutico , Animais , Animais Recém-Nascidos , Cardiotônicos/síntese química , Cardiotônicos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Daunorrubicina/toxicidade , Dicetopiperazinas , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Estrutura Molecular , Miócitos Cardíacos/efeitos dos fármacos , Piperazinas/síntese química , Piperazinas/metabolismo , Ligação Proteica , Ratos Wistar , Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Relação Estrutura-Atividade , Inibidores da Topoisomerase II/síntese química , Inibidores da Topoisomerase II/metabolismo
5.
Sci Rep ; 11(1): 4456, 2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33627707

RESUMO

The bisdioxopiperazine topoisomerase IIß inhibitor ICRF-193 has been previously identified as a more potent analog of dexrazoxane (ICRF-187), a drug used in clinical practice against anthracycline cardiotoxicity. However, the poor aqueous solubility of ICRF-193 has precluded its further in vivo development as a cardioprotective agent. To overcome this issue, water-soluble prodrugs of ICRF-193 were prepared, their abilities to release ICRF-193 were investigated using a novel UHPLC-MS/MS assay, and their cytoprotective effects against anthracycline cardiotoxicity were tested in vitro in neonatal ventricular cardiomyocytes (NVCMs). Based on the obtained results, the bis(2-aminoacetoxymethyl)-type prodrug GK-667 was selected for advanced investigations due to its straightforward synthesis, sufficient solubility, low cytotoxicity and favorable ICRF-193 release. Upon administration of GK-667 to NVCMs, the released ICRF-193 penetrated well into the cells, reached sufficient intracellular concentrations and provided effective cytoprotection against anthracycline toxicity. The pharmacokinetics of the prodrug, ICRF-193 and its rings-opened metabolite was estimated in vivo after administration of GK-667 to rabbits. The plasma concentrations of ICRF-193 reached were found to be adequate to achieve cardioprotective effects in vivo. Hence, GK-667 was demonstrated to be a pharmaceutically acceptable prodrug of ICRF-193 and a promising drug candidate for further evaluation as a potential cardioprotectant against chronic anthracycline toxicity.


Assuntos
Antraciclinas/efeitos adversos , Cardiotônicos/farmacologia , Cardiotoxicidade/tratamento farmacológico , DNA Topoisomerases Tipo II/metabolismo , Dicetopiperazinas/farmacologia , Piperazina/farmacologia , Inibidores da Topoisomerase II/farmacologia , Animais , Cardiotônicos/química , Cardiotoxicidade/metabolismo , Dexrazoxano/química , Dexrazoxano/farmacologia , Dicetopiperazinas/química , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Piperazina/química , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Coelhos , Razoxano/química , Razoxano/farmacologia , Inibidores da Topoisomerase II/química , Água/química
6.
J Pharmacol Exp Ther ; 373(3): 402-415, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32253261

RESUMO

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.


Assuntos
Antraciclinas/efeitos adversos , Cardiotoxicidade/tratamento farmacológico , DNA Topoisomerases Tipo II/metabolismo , Dexrazoxano/farmacologia , Coração/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Inibidores da Topoisomerase II/farmacologia , Animais , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/metabolismo , Linhagem Celular Tumoral , Células HL-60 , Humanos , Masculino , Modelos Animais , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Coelhos , Ratos , Ratos Wistar , Relação Estrutura-Atividade
7.
Clin Sci (Lond) ; 133(16): 1827-1844, 2019 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-31409729

RESUMO

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.


Assuntos
Antraciclinas/toxicidade , Antineoplásicos/toxicidade , Cardiotoxicidade/etiologia , Inibidores de Proteassoma/toxicidade , Animais , Antraciclinas/administração & dosagem , Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Bortezomib/administração & dosagem , Bortezomib/toxicidade , Daunorrubicina/administração & dosagem , Daunorrubicina/toxicidade , Relação Dose-Resposta a Droga , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Oligopeptídeos/administração & dosagem , Oligopeptídeos/toxicidade , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/administração & dosagem , Coelhos , Ratos , Ratos Wistar
8.
Sci Rep ; 9(1): 4524, 2019 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-30872754

RESUMO

Sobuzoxane (MST-16) is an approved anticancer agent, a pro-drug of bisdioxopiperazine analog ICRF-154. Due to the structural similarity of ICRF-154 to dexrazoxane (ICRF-187), MST-16 deserves attention as a cardioprotective drug. This study presents for the first time UHPLC-MS/MS assay of MST-16, ICRF-154 and its metabolite (EDTA-diamide) in cell culture medium, buffer, plasma and cardiac cells and provides data on MST-16 bioactivation under conditions relevant to investigation of cardioprotection of this drug. The analysis of these compounds that differ considerably in their lipophilicity was achieved on the Zorbax SB-Aq column using a mixture of aqueous ammonium formate and methanol as a mobile phase. The biological samples were either diluted or precipitated with methanol, which was followed by acidification for the assay of MST-16. The method was validated for determination of all compounds in the biological materials. The application of the method for analysis of samples from in vitro experiments provided important findings, namely, that (1) MST-16 is quickly decomposed in biological environments, (2) the cardiac cells actively metabolize MST-16, and (3) MST-16 readily penetrates into the cardiac cells and is converted into ICRF-154 and EDTA-diamide. These data are useful for the in-depth examination of the cardioprotective potential of this drug.


Assuntos
Antineoplásicos/análise , Ácido Edético/química , Piperazinas/análise , Razoxano/análogos & derivados , Animais , Antineoplásicos/metabolismo , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Ratos , Ratos Wistar , Razoxano/química , Razoxano/metabolismo , Espectrometria de Massas em Tandem
9.
J Pharmacol Exp Ther ; 364(3): 433-446, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29273587

RESUMO

Dexrazoxane (DEX), the only cardioprotectant approved against anthracycline cardiotoxicity, has been traditionally deemed to be a prodrug of the iron-chelating metabolite ADR-925. However, pharmacokinetic profile of both agents, particularly with respect to the cells and tissues essential for its action (cardiomyocytes/myocardium), remains poorly understood. The aim of this study is to characterize the conversion and disposition of DEX to ADR-925 in vitro (primary cardiomyocytes) and in vivo (rabbits) under conditions where DEX is clearly cardioprotective against anthracycline cardiotoxicity. Our results show that DEX is hydrolyzed to ADR-925 in cell media independently of the presence of cardiomyocytes or their lysate. Furthermore, ADR-925 directly penetrates into the cells with contribution of active transport, and detectable concentrations occur earlier than after DEX incubation. In rabbits, ADR-925 was detected rapidly in plasma after DEX administration to form sustained concentrations thereafter. ADR-925 was not markedly retained in the myocardium, and its relative exposure was 5.7-fold lower than for DEX. Unlike liver tissue, myocardium homogenates did not accelerate the conversion of DEX to ADR-925 in vitro, suggesting that myocardial concentrations in vivo may originate from its distribution from the central compartment. The pharmacokinetic parameters for both DEX and ADR-925 were determined by both noncompartmental analyses and population pharmacokinetics (including joint parent-metabolite model). Importantly, all determined parameters were closer to human than to rodent data. The present results open venues for the direct assessment of the cardioprotective effects of ADR-925 in vitro and in vivo to establish whether DEX is a drug or prodrug.


Assuntos
Cardiotônicos/farmacocinética , Dexrazoxano/farmacocinética , Etilenodiaminas/farmacocinética , Glicina/análogos & derivados , Miócitos Cardíacos/metabolismo , Animais , Cardiotônicos/sangue , Cardiotônicos/metabolismo , Cardiotônicos/farmacologia , Dexrazoxano/sangue , Dexrazoxano/metabolismo , Dexrazoxano/urina , Etilenodiaminas/metabolismo , Glicina/metabolismo , Glicina/farmacocinética , Coelhos , Ratos , Distribuição Tecidual
10.
Toxicology ; 392: 1-10, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28941780

RESUMO

Novel dexrazoxane derivative JR-311 was prepared to investigate structure-activity relationships and mechanism(s) of protection against anthracycline cardiotoxicity. Its cardioprotective, antiproliferative, iron (Fe) chelation and inhibitory and/or depletory activities on topoisomerase IIbeta (TOP2B) were examined and compared with dexrazoxane. While in standard assay, JR-311 failed in both cardioprotection and depletion of TOP2B, its repeated administration to cell culture media led to depletion of TOP2B and significant protection of isolated rat neonatal ventricular cardiomyocytes from daunorubicin-induced damage. This effect was explained by a focused analytical investigation that revealed rapid JR-311 decomposition, resulting in negligible intracellular concentrations of the parent compound but high exposure of cells to the decomposition products, including Fe-chelating JR-H2. Although chemical instability is an obstacle for the development of JR-311, this study identified a novel dexrazoxane analogue with preserved pharmacodynamic properties, contributed to the investigation of structure-activity relationships and suggested that the cardioprotection of bis-dioxopiperazines is likely attributed to TOP2B activity of the parent compound rather than Fe chelation of their hydrolytic metabolites/degradation products. Moreover, this study highlights the importance of early stability testing during future development of novel dexrazoxane analogues.


Assuntos
Cardiotônicos/farmacologia , DNA Topoisomerases Tipo II/metabolismo , Dexrazoxano/farmacologia , Quelantes de Ferro/farmacologia , Animais , Animais Recém-Nascidos , Antraciclinas/toxicidade , Cardiotoxicidade/tratamento farmacológico , Cardiotoxicidade/etiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Daunorrubicina/toxicidade , Dexrazoxano/análogos & derivados , Dicetopiperazinas/farmacologia , Ferro/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Ratos Wistar , Relação Estrutura-Atividade
11.
Toxicology ; 371: 17-28, 2016 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-27744045

RESUMO

Catecholamines may undergo iron-promoted oxidation resulting in formation of reactive intermediates (aminochromes) capable of redox cycling and reactive oxygen species (ROS) formation. Both of them induce oxidative stress resulting in cellular damage and death. Iron chelation has been recently shown as a suitable tool of cardioprotection with considerable potential to protect cardiac cells against catecholamine-induced cardiotoxicity. However, prolonged exposure of cells to classical chelators may interfere with physiological iron homeostasis. Prochelators represent a more advanced approach to decrease oxidative injury by forming a chelating agent only under the disease-specific conditions associated with oxidative stress. Novel prochelator (lacking any iron chelating properties) BHAPI [(E)-N-(1-(2-((4-(4,4,5,5-tetramethyl-1,2,3-dioxoborolan-2-yl)benzyl)oxy)phenyl)ethylidene) isonicotinohydrazide] is converted by ROS to active chelator HAPI with strong iron binding capacity that efficiently inhibits iron-catalyzed hydroxyl radical generation. Our results confirmed redox activity of oxidation products of catecholamines isoprenaline and epinephrine, that were able to activate BHAPI to HAPI that chelates iron ions inside H9c2 cardiomyoblasts. Both HAPI and BHAPI were able to efficiently protect the cells against intracellular ROS formation, depletion of reduced glutathione and toxicity induced by catecholamines and their oxidation products. Hence, both HAPI and BHAPI have shown considerable potential to protect cardiac cells by both inhibition of deleterious catecholamine oxidation to reactive intermediates and prevention of ROS-mediated cardiotoxicity.


Assuntos
Compostos de Boro/farmacologia , Cardiotônicos/farmacologia , Catecolaminas/antagonistas & inibidores , Catecolaminas/toxicidade , Quelantes de Ferro/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Semicarbazonas/farmacologia , Animais , Biocatálise , Ácidos Borônicos/farmacologia , Linhagem Celular , Epinefrina/antagonistas & inibidores , Epinefrina/toxicidade , Glutationa/metabolismo , Humanos , Radical Hidroxila/metabolismo , Ferro/química , Isoproterenol/antagonistas & inibidores , Isoproterenol/toxicidade , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Pró-Fármacos/farmacologia , Ratos
13.
Toxicology ; 350-352: 15-24, 2016 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-27046792

RESUMO

Free cellular iron catalyzes the formation of toxic hydroxyl radicals and therefore chelation of iron could be a promising therapeutic approach in pathological states associated with oxidative stress. Salicylaldehyde isonicotinoyl hydrazone (SIH) is a strong intracellular iron chelator with well documented potential to protect against oxidative damage both in vitro and in vivo. Due to the short biological half-life of SIH and risk of toxicity due to iron depletion, boronate prochelator BSIH has been designed. BSIH cannot bind iron until it is activated by certain reactive oxygen species to active chelator SIH. The aim of this study was to examine the toxicity and cytoprotective potential of BSIH, SIH, and their decomposition products against hydrogen peroxide-induced injury of H9c2 cardiomyoblast cells. Using HPLC, we observed that salicylaldehyde was the main decomposition products of SIH and BSIH, although a small amount of salicylic acid was also detected. In the case of BSIH, the concentration of formed salicylaldehyde consistently exceeded that of SIH. Isoniazid and salicylic acid were not toxic nor did they provide any antioxidant protective effect in H9c2 cells. In contrast, salicylaldehyde was able to chelate intracellular iron and significantly preserve cellular viability and mitochondrial inner membrane potential induced by hydrogen peroxide. However it was consistently less effective than SIH. The inherent toxicities of salicylaldehyde and SIH were similar. Hence, although SIH - the active chelating agent formed following the BSIH activation - undergoes rapid hydrolysis, its principal decomposition product salicylaldehyde accounts markedly for both cytoprotective and toxic properties.


Assuntos
Aldeídos/farmacologia , Ácidos Borônicos/farmacologia , Hidrazonas/farmacologia , Quelantes de Ferro/farmacologia , Ácidos Isonicotínicos/farmacologia , Mioblastos Cardíacos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Aldeídos/toxicidade , Animais , Ácidos Borônicos/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Meia-Vida , Hidrazonas/toxicidade , Peróxido de Hidrogênio/toxicidade , Ferro/metabolismo , Quelantes de Ferro/toxicidade , Ácidos Isonicotínicos/toxicidade , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mioblastos Cardíacos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos
14.
Exp Cell Res ; 339(2): 174-86, 2015 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-26515555

RESUMO

Due to their cardiac origin, H9c2 cells rank among the most popular cell lines in current cardiovascular research, yet molecular phenotype remains elusive. Hence, in this study we used proteomic approach to describe molecular phenotype of H9c2 cells in their undifferentiated (i.e., most frequently used) state, and its functional response to cardiotoxic drug doxorubicin. Of 1671 proteins identified by iTRAQ IEF/LC-MSMS analysis, only 12 proteins were characteristic for striated muscle cells and none was cardiac phenotype-specific. Targeted LC-SRM and western blot analyses confirmed that undifferentiated H9c2 cells are phenotypically considerably different to both primary neonatal cardiomyocytes and adult myocardium. These cells lack proteins essential for formation of striated muscle myofibrils or they express only minor amounts thereof. They also fail to express many proteins important for metabolism of muscle cells. The challenge with clinically relevant concentrations of doxorubicin did not induce a proteomic signature that has been previously noted in primary cardiomyocytes or adult hearts. Instead, several alterations previously described in other cells of mesodermal origin, such as fibroblasts, were observed (e.g., severe down-regulation of collagen synthesis pathway). In conclusion, the molecular phenotype of H9c2 cells resembles very immature myogenic cells with skeletal muscle commitment upon differentiation and thus, translatability of findings obtained in these cells deserves caution.


Assuntos
Doxorrubicina/toxicidade , Miocárdio/citologia , Proteoma/análise , Animais , Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Miocárdio/metabolismo , Fenótipo , Ratos , Relação Estrutura-Atividade
15.
PLoS One ; 10(10): e0139929, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26460540

RESUMO

Cancer cells have a high iron requirement and many experimental studies, as well as clinical trials, have demonstrated that iron chelators are potential anti-cancer agents. The ligand, 2-benzoylpyridine 4-ethyl-3-thiosemicarbazone (Bp4eT), demonstrates both potent anti-neoplastic and anti-retroviral properties. In this study, Bp4eT and its recently identified amidrazone and semicarbazone metabolites were examined and compared with respect to their anti-proliferative activity towards cancer cells (HL-60 human promyelocytic leukemia, MCF-7 human breast adenocarcinoma, HCT116 human colon carcinoma and A549 human lung adenocarcinoma), non-cancerous cells (H9c2 neonatal rat-derived cardiomyoblasts and 3T3 mouse embryo fibroblasts) and their interaction with intracellular iron pools. Bp4eT was demonstrated to be a highly potent and selective anti-neoplastic agent that induces S phase cell cycle arrest, mitochondrial depolarization and apoptosis in MCF-7 cells. Both semicarbazone and amidrazone metabolites showed at least a 300-fold decrease in cytotoxic activity than Bp4eT towards both cancer and normal cell lines. The metabolites also lost the ability to: (1) promote the redox cycling of iron; (2) bind and mobilize iron from labile intracellular pools; and (3) prevent 59Fe uptake from 59Fe-labeled transferrin by MCF-7 cells. Hence, this study demonstrates that the highly active ligand, Bp4eT, is metabolized to non-toxic and pharmacologically inactive analogs, which most likely contribute to its favorable pharmacological profile. These findings are important for the further development of this drug candidate and contribute to the understanding of the structure-activity relationships of these agents.


Assuntos
Antineoplásicos/farmacologia , Quelantes de Ferro/farmacologia , Redes e Vias Metabólicas/efeitos dos fármacos , Tiossemicarbazonas/farmacologia , Antineoplásicos/química , Morte Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Ferro/química , Ferro/metabolismo , Quelantes de Ferro/química , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Oxirredução/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Semicarbazonas/química , Semicarbazonas/metabolismo , Semicarbazonas/farmacologia , Semicarbazonas/toxicidade , Tiossemicarbazonas/química , Tiossemicarbazonas/metabolismo , Tiossemicarbazonas/toxicidade
16.
PLoS One ; 9(11): e112059, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25393531

RESUMO

Salicylaldehyde isonicotinoyl hydrazone (SIH) is a lipophilic, tridentate iron chelator with marked anti-oxidant and modest cytotoxic activity against neoplastic cells. However, it has poor stability in an aqueous environment due to the rapid hydrolysis of its hydrazone bond. In this study, we synthesized a series of new SIH analogs (based on previously described aromatic ketones with improved hydrolytic stability). Their structure-activity relationships were assessed with respect to their stability in plasma, iron chelation efficacy, redox effects and cytotoxic activity against MCF-7 breast adenocarcinoma cells. Furthermore, studies assessed the cytotoxicity of these chelators and their ability to afford protection against hydrogen peroxide-induced oxidative injury in H9c2 cardiomyoblasts. The ligands with a reduced hydrazone bond, or the presence of bulky alkyl substituents near the hydrazone bond, showed severely limited biological activity. The introduction of a bromine substituent increased ligand-induced cytotoxicity to both cancer cells and H9c2 cardiomyoblasts. A similar effect was observed when the phenolic ring was exchanged with pyridine (i.e., changing the ligating site from O, N, O to N, N, O), which led to pro-oxidative effects. In contrast, compounds with long, flexible alkyl chains adjacent to the hydrazone bond exhibited specific cytotoxic effects against MCF-7 breast adenocarcinoma cells and low toxicity against H9c2 cardiomyoblasts. Hence, this study highlights important structure-activity relationships and provides insight into the further development of aroylhydrazone iron chelators with more potent and selective anti-neoplastic effects.


Assuntos
Aldeídos/química , Aldeídos/farmacologia , Antineoplásicos/toxicidade , Antioxidantes/farmacologia , Hidrazonas/química , Hidrazonas/farmacologia , Quelantes de Ferro/farmacologia , Aldeídos/toxicidade , Antineoplásicos/química , Antioxidantes/química , Linhagem Celular , Humanos , Hidrazonas/toxicidade , Peróxido de Hidrogênio/toxicidade , Quelantes de Ferro/química , Células MCF-7 , Mioblastos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Relação Estrutura-Atividade
17.
Free Radic Biol Med ; 74: 210-21, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24992833

RESUMO

Oxidative stress is a common denominator of numerous cardiovascular disorders. Free cellular iron catalyzes the formation of highly toxic hydroxyl radicals, and iron chelation may thus be an effective therapeutic approach. However, using classical iron chelators in diseases without iron overload poses risks that necessitate more advanced approaches, such as prochelators that are activated to chelate iron only under disease-specific oxidative stress conditions. In this study, three cell-membrane-permeable iron chelators (clinically used deferasirox and experimental SIH and HAPI) and five boronate-masked prochelator analogs were evaluated for their ability to protect cardiac cells against oxidative injury induced by hydrogen peroxide. Whereas the deferasirox-derived agents TIP and TRA-IMM displayed negligible protection and even considerable toxicity, the aroylhydrazone prochelators BHAPI and BSIH-PD provided significant cytoprotection and displayed lower toxicity after prolonged cellular exposure compared to their parent chelators HAPI and SIH, respectively. Overall, the most favorable properties in terms of protective efficiency and low inherent cytotoxicity were observed with the aroylhydrazone prochelator BSIH. BSIH efficiently protected both H9c2 rat cardiomyoblast-derived cells and isolated primary rat cardiomyocytes against hydrogen peroxide-induced mitochondrial and lysosomal dysregulation and cell death. At the same time, BSIH was nontoxic at concentrations up to its solubility limit (600 µM) and in 72-h incubation. Hence, BSIH merits further investigation for prevention and/or treatment of cardiovascular disorders associated with a known (or presumed) component of oxidative stress.


Assuntos
Citoproteção , Quelantes de Ferro/farmacologia , Mitocôndrias Cardíacas/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Aldeídos/química , Aldeídos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Benzoatos/química , Benzoatos/farmacologia , Compostos de Boro/química , Compostos de Boro/farmacologia , Ácidos Borônicos/química , Ácidos Borônicos/farmacologia , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Deferasirox , Hidrazonas/química , Hidrazonas/farmacologia , Peróxido de Hidrogênio/metabolismo , Ferro/química , Ferro/metabolismo , Quelantes de Ferro/química , Ácidos Isonicotínicos/química , Ácidos Isonicotínicos/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias Cardíacas/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Ratos , Ratos Wistar , Semicarbazonas/química , Semicarbazonas/farmacologia , Triazóis/química , Triazóis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...