38 results on '"Simůnek T"'
Search Results
2. Dexrazoxane-afforded protection against chronic anthracycline cardiotoxicity in vivo: effective rescue of cardiomyocytes from apoptotic cell death.
- Author
-
Popelová, O., Štěrba, M., Hašková, P., Šimůnek, T., Hroch, M., Gunčová, I., Nachtigal, P., Adamcová, M., Geršl, V., Mazurová, Y., Popelová, O, Sterba, M, Hasková, P, Simůnek, T, Guncová, I, Adamcová, M, Gersl, V, and Mazurová, Y
- Subjects
ANTHRACYCLINES ,APOPTOSIS ,CELL death ,HYDROLYSIS ,HEART cells ,PREVENTION of heart diseases ,RESEARCH ,HETEROCYCLIC compounds ,ANIMAL experimentation ,RESEARCH methodology ,RABBITS ,MEDICAL cooperation ,EVALUATION research ,COMPARATIVE studies ,CELLS ,CARDIOTONIC agents ,HEART diseases ,PHARMACODYNAMICS ,CHEMICAL inhibitors - Abstract
Background: Dexrazoxane (DEX, ICRF-187) is the only clinically approved cardioprotectant against anthracycline cardiotoxicity. It has been traditionally postulated to undergo hydrolysis to iron-chelating agent ADR-925 and to prevent anthracycline-induced oxidative stress, progressive cardiomyocyte degeneration and subsequent non-programmed cell death. However, the additional capability of DEX to protect cardiomyocytes from apoptosis has remained unsubstantiated under clinically relevant in vivo conditions.Methods: Chronic anthracycline cardiotoxicity was induced in rabbits by repeated daunorubicin (DAU) administrations (3 mg kg(-1) weekly for 10 weeks). Cardiomyocyte apoptosis was evaluated using TUNEL (terminal deoxynucleotidyl transferase biotin-dUTP nick end labelling) assay and activities of caspases 3/7, 8, 9 and 12. Lipoperoxidation was assayed using HPLC determination of myocardial malondialdehyde and 4-hydroxynonenal immunodetection.Results: Dexrazoxane (60 mg kg(-1)) co-treatment was capable of overcoming DAU-induced mortality, left ventricular dysfunction, profound structural damage of the myocardium and release of cardiac troponin T and I to circulation. Moreover, for the first time, it has been shown that DEX affords significant and nearly complete cardioprotection against anthracycline-induced apoptosis in vivo and effectively suppresses the complex apoptotic signalling triggered by DAU. In individual animals, the severity of apoptotic parameters significantly correlated with cardiac function. However, this effective cardioprotection occurred without a significant decrease in anthracycline-induced lipoperoxidation.Conclusion: This study identifies inhibition of apoptosis as an important target for effective cardioprotection against chronic anthracycline cardiotoxicity and suggests that lipoperoxidation-independent mechanisms are involved in the cardioprotective action of DEX. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
3. Anthracycline toxicity to cardiomyocytes or cancer cells is differently affected by iron chelation with salicylaldehyde isonicotinoyl hydrazone.
- Author
-
Šimůnek, T., Štěrba, M., Popelová, O., Kaiserová, H., Adamcová, M., Hroch, M., Hašková, P., Poňka, P., Geršl, V., Simůnek, T, Sterba, M, Popelová, O, Kaiserová, H, Adamcová, M, Hasková, P, Ponka, P, and Gersl, V
- Subjects
ANTINEOPLASTIC agents ,ANTHRACYCLINES ,CELL lines ,TOXICITY testing ,HEART cells ,PEROXIDATION ,MEDICAL research ,PHARMACEUTICAL research ,CELL metabolism ,ALDEHYDES ,ANIMAL experimentation ,ANIMAL populations ,ANTINEOPLASTIC antibiotics ,CELL physiology ,CELLS ,COMPARATIVE studies ,DAUNOMYCIN ,DOSE-effect relationship in pharmacology ,RESEARCH methodology ,MEDICAL cooperation ,LIPID peroxidation (Biology) ,ORGANIC compounds ,PHARMACOLOGY ,RATS ,RESEARCH ,RESEARCH funding ,TIME ,MALONDIALDEHYDE ,OXIDATIVE stress ,EVALUATION research ,ACUTE promyelocytic leukemia ,CHELATING agents ,PHARMACODYNAMICS - Abstract
Background and Purpose: The clinical utility of anthracycline antineoplastic drugs is limited by the risk of cardiotoxicity, which has been traditionally attributed to iron-mediated production of reactive oxygen species (ROS).Experimental Approach: The aims of this study were to examine the strongly lipophilic iron chelator, salicylaldehyde isonicotinoyl hydrazone (SIH), for its ability to protect rat isolated cardiomyocytes against the toxicity of daunorubicin (DAU) and to investigate the effects of SIH on DAU-induced inhibition of proliferation in a leukaemic cell line. Cell toxicity was measured by release of lactate dehydrogenase and staining with Hoechst 33342 or propidium iodide and lipid peroxidation by malonaldehyde formation.Key Results: SIH fully protected cardiomyocytes against model oxidative injury induced by hydrogen peroxide exposure. SIH also significantly but only partially and with no apparent dose-dependency, reduced DAU-induced cardiomyocyte death. However, the observed protection was not accompanied by decreased lipid peroxidation. In the HL-60 acute promyelocytic leukaemia cell line, SIH did not blunt the antiproliferative efficacy of DAU. Instead, at concentrations that reduced DAU toxicity to cardiomyocytes, SIH enhanced the tumoricidal action of DAU.Conclusions and Implications: This study demonstrates that iron is most likely involved in anthracycline cardiotoxicity and that iron chelation has protective potential, but apparently through mechanism(s) other than by inhibition of ROS-induced injury. In addition to cardioprotection, iron chelation may have considerable potential to improve the therapeutic action of anthracyclines by enhancing their anticancer efficiency and this potential warrants further investigation. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
4. Exploring the anti-cancer activity of novel thiosemicarbazones generated through the combination of retro-fragments: dissection of critical structure-activity relationships.
- Author
-
Serda M, Kalinowski DS, Rasko N, Potůčková E, Mrozek-Wilczkiewicz A, Musiol R, Małecki JG, Sajewicz M, Ratuszna A, Muchowicz A, Gołąb J, Simůnek T, Richardson DR, and Polanski J
- Subjects
- Antineoplastic Agents chemical synthesis, Ascorbic Acid metabolism, Biological Transport drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Humans, Iron chemistry, Oxidation-Reduction drug effects, Structure-Activity Relationship, Thiosemicarbazones chemical synthesis, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Drug Design, Thiosemicarbazones chemistry, Thiosemicarbazones pharmacology
- Abstract
Thiosemicarbazones (TSCs) are an interesting class of ligands that show a diverse range of biological activity, including anti-fungal, anti-viral and anti-cancer effects. Our previous studies have demonstrated the potent in vivo anti-tumor activity of novel TSCs and their ability to overcome resistance to clinically used chemotherapeutics. In the current study, 35 novel TSCs of 6 different classes were designed using a combination of retro-fragments that appear in other TSCs. Additionally, di-substitution at the terminal N4 atom, which was previously identified to be critical for potent anti-cancer activity, was preserved through the incorporation of an N4-based piperazine or morpholine ring. The anti-proliferative activity of the novel TSCs were examined in a variety of cancer and normal cell-types. In particular, compounds 1d and 3c demonstrated the greatest promise as anti-cancer agents with potent and selective anti-proliferative activity. Structure-activity relationship studies revealed that the chelators that utilized "soft" donor atoms, such as nitrogen and sulfur, resulted in potent anti-cancer activity. Indeed, the N,N,S donor atom set was crucial for the formation of redox active iron complexes that were able to mediate the oxidation of ascorbate. This further highlights the important role of reactive oxygen species generation in mediating potent anti-cancer activity. Significantly, this study identified the potent and selective anti-cancer activity of 1d and 3c that warrants further examination.
- Published
- 2014
- Full Text
- View/download PDF
5. Comparison of various iron chelators and prochelators as protective agents against cardiomyocyte oxidative injury.
- Author
-
Jansová H, Macháček M, Wang Q, Hašková P, Jirkovská A, Potůčková E, Kielar F, Franz KJ, and Simůnek T
- Subjects
- Aldehydes chemistry, Aldehydes pharmacology, Animals, Apoptosis drug effects, Benzoates chemistry, Benzoates pharmacology, Boron Compounds chemistry, Boron Compounds pharmacology, Boronic Acids chemistry, Boronic Acids pharmacology, Cell Line, Cell Membrane Permeability drug effects, Deferasirox, Hydrazones chemistry, Hydrazones pharmacology, Hydrogen Peroxide metabolism, Iron chemistry, Iron metabolism, Iron Chelating Agents chemistry, Isonicotinic Acids chemistry, Isonicotinic Acids pharmacology, Membrane Potential, Mitochondrial drug effects, Mitochondria, Heart drug effects, Myocytes, Cardiac physiology, Rats, Rats, Wistar, Semicarbazones chemistry, Semicarbazones pharmacology, Triazoles chemistry, Triazoles pharmacology, Cytoprotection, Iron Chelating Agents pharmacology, Mitochondria, Heart physiology, Myocytes, Cardiac drug effects, Oxidative Stress drug effects
- Abstract
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., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
6. Oxidative stress, redox signaling, and metal chelation in anthracycline cardiotoxicity and pharmacological cardioprotection.
- Author
-
Stěrba M, Popelová O, Vávrová A, Jirkovský E, Kovaříková P, Geršl V, and Simůnek T
- Subjects
- Anthracyclines chemistry, Anthracyclines pharmacology, Antineoplastic Agents adverse effects, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Antioxidants chemistry, Antioxidants pharmacology, Cardiotonic Agents adverse effects, Cardiotonic Agents chemistry, Cardiotonic Agents pharmacology, Chelating Agents adverse effects, Chelating Agents chemistry, Humans, Oxidation-Reduction, Razoxane adverse effects, Razoxane chemistry, Razoxane pharmacology, Reactive Oxygen Species metabolism, Anthracyclines adverse effects, Chelating Agents pharmacology, Heart drug effects, Metals adverse effects, Myocardium metabolism, Oxidative Stress, Signal Transduction
- Abstract
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.
- Published
- 2013
- Full Text
- View/download PDF
7. Amino acid derivatives as transdermal permeation enhancers.
- Author
-
Janůšová B, Skolová B, Tükörová K, Wojnarová L, Simůnek T, Mladěnka P, Filipský T, Ríha M, Roh J, Palát K, Hrabálek A, and Vávrová K
- Subjects
- 3T3 Cells, Administration, Cutaneous, Amino Acids metabolism, Amino Acids toxicity, Animals, Cell Line, Drug Stability, Humans, Keratinocytes drug effects, Keratinocytes metabolism, Mice, Plasma metabolism, Proline analogs & derivatives, Proline metabolism, Proline pharmacology, Proline toxicity, Rats, Skin metabolism, Swine, Amino Acids chemistry, Amino Acids pharmacology, Permeability drug effects, Skin drug effects, Skin Absorption drug effects
- Abstract
Transdermal permeation enhancers are compounds that temporarily decrease skin barrier properties to promote drug flux. In this study, we investigated enhancers with amino acids (proline, sarcosine, alanine, β-alanine, and glycine) attached to hydrophobic chain(s) via a biodegradable ester link. The double-chain lipid-like substances displayed no enhancing effect, whereas single-chain substances significantly increased skin permeability. The proline derivative l-Pro2 reached enhancement ratios of up to 40 at 1% concentration, which is higher than that of the well-established and standard enhancers Azone, DDAIP, DDAK, and Transkarbam 12. No stereoselectivity was observed. l-Pro2 acted synergistically with propylene glycol. Infrared studies revealed that l-Pro2 forms a separate liquid ordered phase in the stratum corneum lipids and has no significant effect on proteins. l-Pro2 action was at least partially reversible as measured by skin electrical impedance. Toxicity in keratinocyte (HaCaT) and fibroblast (3T3) cell lines showed IC(50) values ranging from tens to hundreds of μM, which is comparable with standard enhancers. Furthermore, l-Pro2 was rapidly decomposed in plasma. In vivo transdermal absorption studies in rats confirmed the enhancing activity of l-Pro2 and suggested its negligible skin toxicity and minimal effect on transepidermal water loss. These properties make l-Pro2 a promising candidate for potential clinical use., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
8. Chronic anthracycline cardiotoxicity: molecular and functional analysis with focus on nuclear factor erythroid 2-related factor 2 and mitochondrial biogenesis pathways.
- Author
-
Jirkovsky E, Popelová O, Kriváková-Stanková P, Vávrová A, Hroch M, Hasková P, Brcáková-Dolezelová E, Micuda S, Adamcová M, Simůnek T, Cervinková Z, Gersl V, and Sterba M
- Subjects
- Animals, Cell Nucleus drug effects, Cell Nucleus metabolism, Daunorubicin pharmacology, Echocardiography, Glutathione metabolism, Heart Function Tests, Heart Ventricles drug effects, Heart Ventricles metabolism, Lipid Peroxidation drug effects, Male, Mitochondria, Heart drug effects, Myocardium pathology, Oxidative Stress drug effects, Rabbits, Real-Time Polymerase Chain Reaction, Survival, Transcription Factors metabolism, Troponin T metabolism, Anthracyclines toxicity, Antibiotics, Antineoplastic toxicity, Heart Diseases chemically induced, Heart Diseases metabolism, Mitochondria, Heart metabolism, NF-E2-Related Factor 2 biosynthesis
- Abstract
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.
- Published
- 2012
- Full Text
- View/download PDF
9. Methyl and ethyl ketone analogs of salicylaldehyde isonicotinoyl hydrazone: novel iron chelators with selective antiproliferative action.
- Author
-
Macková E, Hrušková K, Bendová P, Vávrová A, Jansová H, Hašková P, Kovaříková P, Vávrová K, and Simůnek T
- Subjects
- Aldehydes pharmacology, Antineoplastic Agents chemistry, Apoptosis drug effects, Ascorbic Acid metabolism, Caspases metabolism, Cell Cycle drug effects, Cell Line, Tumor, Deferoxamine pharmacology, Drug Screening Assays, Antitumor, HL-60 Cells, Humans, Hydrazones pharmacology, Ketones chemistry, Membrane Potential, Mitochondrial drug effects, Oxidation-Reduction, Aldehydes chemistry, Antineoplastic Agents pharmacology, Hydrazones chemistry, Iron Chelating Agents chemistry, Iron Chelating Agents pharmacology
- Abstract
Salicylaldehyde isonicotinoyl hydrazone (SIH) is a lipophilic, orally-active tridentate iron chelator providing both effective protection against various types of oxidative stress-induced cellular injury and anticancer action. However, the major limitation of SIH is represented by its labile hydrazone bond that makes it prone to plasma hydrolysis. Recently, nine new SIH analogues derived from aromatic ketones with improved hydrolytic stability were developed. Here we analyzed their antiproliferative potential in MCF-7 breast adenocarcinoma and HL-60 promyelocytic leukemia cell lines. Seven of the tested substances showed greater selectivity than the parent agent SIH towards the latter cancer cell lines compared to non-cancerous H9c2 cardiomyoblast-derived cells. The tested chelators induced a dose-dependent dissipation of the inner mitochondrial membrane potential, an induction of apoptosis as evidenced by Annexin V positivity or significant increases of activities of caspases 3, 7, 8 and 9 and cell cycle arrest. With the exception of nitro group-bearing NHAPI, the studies of iron complexes of the chelators confirmed the crucial role of iron in the mechanism of their antiproliferative action. Finally, all the assayed chelators inhibited the oxidation of ascorbate by iron ions indicating lack of redox activity of the chelator-iron complexes. In conclusion, this study identified several important design criteria for improvement of the antiproliferative selectivity of the aroylhydrazone iron chelators. Several of the novel compounds--in particular the ethylketone-derived HPPI, NHAPI and acetyl-substituted A2,4DHAPI--merit deeper investigation as promising potent and selective anticancer agents., (Copyright © 2012 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
10. Comparison of various iron chelators used in clinical practice as protecting agents against catecholamine-induced oxidative injury and cardiotoxicity.
- Author
-
Hašková P, Koubková L, Vávrová A, Macková E, Hrušková K, Kovaříková P, Vávrová K, and Simůnek T
- Subjects
- Animals, Cardiotoxins antagonists & inhibitors, Catecholamines antagonists & inhibitors, Cell Line, Myocytes, Cardiac metabolism, Oxidation-Reduction drug effects, Oxidative Stress physiology, Rats, Cardiotonic Agents pharmacology, Cardiotoxins toxicity, Catecholamines toxicity, Iron Chelating Agents pharmacology, Myocytes, Cardiac drug effects, Oxidative Stress drug effects
- Abstract
Catecholamines are stress hormones and sympathetic neurotransmitters essential for control of cardiac function and metabolism. However, pathologically increased catecholamine levels may be cardiotoxic by mechanism that includes iron-catalyzed formation of reactive oxygen species. In this study, five iron chelators used in clinical practice were examined for their potential to protect cardiomyoblast-derived cell line H9c2 from the oxidative stress and toxicity induced by catecholamines epinephrine and isoprenaline and their oxidation products. Hydroxamate iron chelator desferrioxamine (DFO) significantly reduced oxidation of catecholamines to more toxic products and abolished redox activity of the catecholamine-iron complex at pH 7.4. However, due to its hydrophilicity and large molecule, DFO was able to protects cells only at very high and clinically unachievable concentrations. Two newer chelators, deferiprone (L1) and deferasirox (ICL670A), showed much better protective potential and were effective at one or two orders of magnitude lower concentrations as compared to DFO that were within their clinically relevant plasma levels. Ethylenediaminetetraacetic acid (EDTA), dexrazoxane (ICRF-187, clinically approved cardioprotective agent against anthracycline-induced cardiotoxicity) as well as selected beta adrenoreceptor antagonists and calcium channel blockers exerted no effect. Hence, results of the present study indicate that small, lipophilic and iron-specific chelators L1 and ICL670A can provide significant protection against the oxidative stress and cardiomyocyte damage exerted by catecholamines and/or their reactive oxidation intermediates. This potential new application of the clinically approved drugs L1 and ICL670A warrants further investigation, preferably using more complex in vivo animal models., (Copyright © 2011 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
11. In vivo and in vitro assessment of the role of glutathione antioxidant system in anthracycline-induced cardiotoxicity.
- Author
-
Vávrová A, Popelová O, Stěrba M, Jirkovský E, Hašková P, Mertlíková-Kaiserová H, Geršl V, and Simůnek T
- Subjects
- Animals, Antibiotics, Antineoplastic, Buthionine Sulfoximine metabolism, Cell Line, Dose-Response Relationship, Drug, Glutathione Peroxidase metabolism, Glutathione Reductase metabolism, Heart drug effects, Hydrogen Peroxide toxicity, Male, Models, Animal, Oxidative Stress drug effects, Pyrrolidonecarboxylic Acid metabolism, Rabbits, Rats, Thiazolidines metabolism, Antioxidants pharmacology, Daunorubicin adverse effects, Glutathione pharmacology, Heart Diseases chemically induced
- Abstract
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.
- Published
- 2011
- Full Text
- View/download PDF
12. Proteomic insights into chronic anthracycline cardiotoxicity.
- Author
-
Stěrba M, Popelová O, Lenčo J, Fučíková A, Brčáková E, Mazurová Y, Jirkovský E, Simůnek T, Adamcová M, Mičuda S, Stulík J, and Geršl V
- Subjects
- Animals, Blotting, Western, Daunorubicin toxicity, Echocardiography, Electrophoresis, Gel, Two-Dimensional, Extracellular Matrix drug effects, Extracellular Matrix metabolism, Heart Ventricles drug effects, Immunohistochemistry, Malondialdehyde metabolism, Mitochondrial Proteins metabolism, Proteomics, Rabbits, Reverse Transcriptase Polymerase Chain Reaction, Troponin I metabolism, Vimentin metabolism, Anthracyclines toxicity, Heart Failure chemically induced, Heart Failure metabolism, Heart Ventricles metabolism, Myocardium metabolism
- Abstract
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., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
13. Iron chelation with salicylaldehyde isonicotinoyl hydrazone protects against catecholamine autoxidation and cardiotoxicity.
- Author
-
Hašková P, Kovaříková P, Koubková L, Vávrová A, Macková E, and Simůnek T
- Subjects
- Animals, Ascorbic Acid chemistry, Binding, Competitive, Caspases metabolism, Cell Line, Cell Survival drug effects, Cytoprotection, Enzyme Assays, Myoblasts, Cardiac cytology, Myoblasts, Cardiac metabolism, Oxidation-Reduction, Oxidative Stress, Rats, Reactive Oxygen Species metabolism, Aldehydes pharmacology, Catecholamines metabolism, Coordination Complexes pharmacology, Hydrazones pharmacology, Iron metabolism, Iron Chelating Agents pharmacology, Myoblasts, Cardiac drug effects
- Abstract
Elevated catecholamine levels are known to induce damage of the cardiac tissue. This catecholamine cardiotoxicity may stem from their ability to undergo oxidative conversion to aminochromes and concomitant production of reactive oxygen species (ROS), which damage cardiomyocytes via the iron-catalyzed Fenton-type reaction. This suggests the possibility of cardioprotection by iron chelation. Our in vitro experiments have demonstrated a spontaneous decrease in the concentration of the catecholamines epinephrine and isoprenaline during their 24-h preincubation in buffered solution as well as their gradual conversion to oxidation products. These changes were significantly augmented by addition of iron ions and reduced by the iron-chelating agent salicylaldehyde isonicotinoyl hydrazone (SIH). Oxidized catecholamines were shown to form complexes with iron that had significant redox activity, which could be suppressed by SIH. Experiments using the H9c2 cardiomyoblast cell line revealed higher cytotoxicity of oxidized catecholamines than of the parent compounds, apparently through the induction of caspase-independent cell death, whereas co-incubation of cells with SIH was able to significantly preserve cell viability. A significant increase in intracellular ROS formation was observed after the incubation of cells with catecholamine oxidation products; this could be significantly reduced by SIH. In contrast, parent catecholamines did not increase, but rather decreased, cellular ROS production. Hence, our results demonstrate an important role for redox-active iron in catecholamine autoxidation and subsequent toxicity. The iron chelator SIH has shown considerable potential to protect cardiac cells by both inhibition of deleterious catecholamine oxidation to reactive intermediates and prevention of ROS-mediated cardiotoxicity., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
14. Direct administration of rutin does not protect against catecholamine cardiotoxicity.
- Author
-
Mladenka P, Zatloukalová L, Simůnek T, Bobrovová Z, Semecký V, Nachtigal P, Hasková P, Macková E, Vávrová J, Holecková M, Palicka V, and Hrdina R
- Subjects
- Adrenergic beta-Agonists pharmacology, Animals, Cardiac Output drug effects, Chelating Agents pharmacology, Data Interpretation, Statistical, Free Radical Scavengers metabolism, Glutathione metabolism, Heart Diseases pathology, Heart Function Tests, Isoproterenol pharmacology, Male, Myocardium pathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Organ Size drug effects, Rats, Rats, Wistar, Reactive Oxygen Species metabolism, Vascular Resistance drug effects, Catecholamines antagonists & inhibitors, Catecholamines toxicity, Heart Diseases chemically induced, Heart Diseases prevention & control, Rutin pharmacology
- Abstract
High levels of catecholamines are cardiotoxic and may trigger acute myocardial infarction (AMI). Similarly, the synthetic catecholamine isoprenaline (ISO) evokes a pathological state similar to AMI. During AMI there is a marked increase of free iron and copper which are crucial catalysts of reactive oxygen species formation. Rutin, a natural flavonoid glycoside possessing free radical scavenging and iron/copper chelating activity, may therefore be potentially useful in reduction of catecholamine cardiotoxicity as was previously demonstrated after its long-term peroral administration. Male Wistar:Han rats received rutin (46 or 11.5 mg kg(-1) i.v.) alone or with necrogenic dose of ISO (100 mg kg(-1) s.c.). Haemodynamic parameters were measured 24h after drug application together with analysis of blood, myocardial content of elements and histological examination. Results were confirmed by cytotoxicity studies using cardiomyoblast cell line H9c2. Rutin in a dose of 46 mg kg(-1) aggravated ISO-cardiotoxicity while the dose of 11 mg kg(-1) had no effect. These unexpected results were in agreement with in vitro experiments, where co-incubation with larger concentrations of rutin significantly augmented ISO cytotoxicity. Our results, in contrast to previous studies in the literature, suggest that the reported positive effects of peroral administration of rutin were unlikely to have been mediated by rutin per se but probably by its metabolite(s) or by some other, at this moment, unknown adaptive mechanism(s), which merit further investigation.
- Published
- 2009
- Full Text
- View/download PDF
15. Anthracycline-induced cardiotoxicity: overview of studies examining the roles of oxidative stress and free cellular iron.
- Author
-
Simůnek T, Stérba M, Popelová O, Adamcová M, Hrdina R, and Gersl V
- Subjects
- Animals, Anthracyclines pharmacology, Antibiotics, Antineoplastic pharmacology, Clinical Trials as Topic, Heart Diseases physiopathology, Humans, Iron metabolism, Oxidative Stress drug effects, Reactive Oxygen Species metabolism, Risk Factors, Anthracyclines adverse effects, Antibiotics, Antineoplastic adverse effects, Heart Diseases chemically induced
- Abstract
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.
- Published
- 2009
- Full Text
- View/download PDF
16. The novel iron chelator, 2-pyridylcarboxaldehyde 2-thiophenecarboxyl hydrazone, reduces catecholamine-mediated myocardial toxicity.
- Author
-
Mladĕnka P, Kalinowski DS, Haskova P, Bobrovová Z, Hrdina R, Simůnek T, Nachtigal P, Semecký V, Vávrová J, Holeckova M, Palicka V, Mazurová Y, Jansson PJ, and Richardson DR
- Subjects
- Animals, Catecholamines antagonists & inhibitors, Catecholamines metabolism, Catecholamines toxicity, Cell Line, Deferoxamine administration & dosage, Iron metabolism, Isoproterenol antagonists & inhibitors, Isoproterenol metabolism, Male, Myocytes, Cardiac metabolism, Oxidation-Reduction, Rats, Rats, Wistar, Iron Chelating Agents pharmacology, Isoproterenol toxicity, Myocytes, Cardiac drug effects, Thiophenes pharmacology
- Abstract
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.
- Published
- 2009
- Full Text
- View/download PDF
17. Deferiprone does not protect against chronic anthracycline cardiotoxicity in vivo.
- Author
-
Popelová O, Sterba M, Simůnek T, Mazurová Y, Guncová I, Hroch M, Adamcová M, and Gersl V
- Subjects
- Animals, Anthracyclines antagonists & inhibitors, Antineoplastic Agents therapeutic use, Antineoplastic Agents toxicity, Cardiotonic Agents therapeutic use, Cardiotonic Agents toxicity, Cell Proliferation drug effects, Daunorubicin antagonists & inhibitors, Deferiprone, HL-60 Cells, Heart Diseases mortality, Heart Diseases pathology, Humans, Male, Rabbits, Anthracyclines administration & dosage, Anthracyclines toxicity, Heart Diseases chemically induced, Pyridones therapeutic use
- Abstract
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.
- Published
- 2008
- Full Text
- View/download PDF
18. Flavonoids as protectors against doxorubicin cardiotoxicity: role of iron chelation, antioxidant activity and inhibition of carbonyl reductase.
- Author
-
Kaiserová H, Simůnek T, van der Vijgh WJ, Bast A, and Kvasnicková E
- Subjects
- Adolescent, Adult, Animals, Animals, Newborn, Antibiotics, Antineoplastic adverse effects, Cells, Cultured, Female, Heart drug effects, Humans, Iron metabolism, Male, Middle Aged, Models, Biological, Rats, Rats, Wistar, Alcohol Oxidoreductases antagonists & inhibitors, Antioxidants pharmacology, Cardiotonic Agents pharmacology, Doxorubicin adverse effects, Flavonoids pharmacology, Iron Chelating Agents pharmacology
- Abstract
Anthracycline antibiotics (e.g. doxorubicin and daunorubicin) are among the most effective and widely used anticancer drugs. Unfortunately, their clinical use is limited by the dose-dependent cardiotoxicity. Flavonoids represent a potentially attractive class of compounds to mitigate the anthracycline cardiotoxicity due to their iron-chelating, antioxidant and carbonyl reductase-inhibitory effects. The relative contribution of various characteristics of the flavonoids to their cardioprotective activity is, however, not known. A series of ten flavonoids including quercetin, quercitrin, 7-monohydroxyethylrutoside (monoHER) and seven original synthetic compounds were employed to examine the relationships between their inhibitory effects on carbonyl reduction, iron-chelation and antioxidant properties with respect to their protective potential against doxorubicin-induced cardiotoxicity. Cardioprotection was investigated in the neonatal rat ventricular cardiomyocytes whereas the H9c2 cardiomyoblast cells were used for cytotoxicity testing. Iron chelation was examined via the calcein assay and antioxidant effects and site-specific scavenging were quantified by means of inhibition of lipid peroxidation and hydroxyl radical scavenging activity, respectively. Inhibition of carbonyl reductases was assessed in cytosol from human liver. None of the flavonoids tested had better cardioprotective action than the reference cardioprotector, monoHER. However, a newly synthesized quaternary ammonium analog with comparable cardioprotective effects has been identified. No direct correlation between the iron-chelating and/or antioxidant effect and cardioprotective potential has been found. A major role of carbonyl reductase inhibition seems unlikely, as the best two cardioprotectors of the series are only weak reductase inhibitors.
- Published
- 2007
- Full Text
- View/download PDF
19. Iron chelation-afforded cardioprotection against chronic anthracycline cardiotoxicity: a study of salicylaldehyde isonicotinoyl hydrazone (SIH).
- Author
-
Sterba M, Popelová O, Simůnek T, Mazurová Y, Potácová A, Adamcová M, Guncová I, Kaiserová H, Palicka V, Ponka P, and Gersl V
- Subjects
- Animals, Cells, Cultured, Daunorubicin, Heart Diseases chemically induced, Heart Diseases pathology, Heart Diseases physiopathology, Heart Ventricles pathology, Heart Ventricles physiopathology, Male, Myocardial Contraction drug effects, Myocytes, Cardiac drug effects, Myocytes, Cardiac physiology, Rabbits, Rats, Troponin T blood, Aldehydes pharmacology, Heart Diseases prevention & control, Hydrazones pharmacology, Iron Chelating Agents pharmacology
- Abstract
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.
- Published
- 2007
- Full Text
- View/download PDF
20. A pilot study of matrix metalloproteinases on the model of daunorubicin-induced cardiomyopathy in rabbits.
- Author
-
Potácová A, Adamcová M, Sterba M, Popelová O, Simůnek T, Mazurová Y, Guncová I, and Gersl V
- Subjects
- Animals, Cardiomyopathies chemically induced, Cardiomyopathies metabolism, Cardiomyopathies physiopathology, Collagen metabolism, Rabbits, Troponin T metabolism, Ventricular Remodeling, Cardiomyopathies enzymology, Daunorubicin toxicity, Matrix Metalloproteinases metabolism
- Abstract
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.
- Published
- 2007
21. Early detection of anthracycline cardiotoxicity in a rabbit model: left ventricle filling pattern versus troponin T determination.
- Author
-
Sterba M, Simůnek T, Popelová O, Potácová A, Adamcová M, Mazurová Y, Holecková M, and Gersl V
- Subjects
- Anesthetics administration & dosage, Animals, Antibiotics, Antineoplastic, Biomarkers blood, Blood Pressure, Calcium metabolism, Collagen metabolism, Daunorubicin, Disease Models, Animal, Echocardiography, Doppler, Heart Diseases blood, Heart Diseases chemically induced, Heart Diseases pathology, Heart Diseases physiopathology, Heart Rate, Male, Myocardial Contraction, Myocardium metabolism, Myocardium pathology, Pilot Projects, Rabbits, Time Factors, Ventricular Pressure, Xylazine administration & dosage, Heart Diseases diagnosis, Troponin T blood, Ventricular Function, Left
- Abstract
Anthracycline cardiotoxicity represents a serious risk of anticancer chemotherapy. The aim of the present pilot study was to compare the potential of both the left ventricular (LV) filling pattern evaluation and cardiac troponin T (cTnT) plasma levels determination for the early detection of daunorubicin-induced cardiotoxicity in rabbits. The echocardiographic measurements of transmitral LV inflow as well as cTnT determinations were performed weekly for 10 weeks in daunorubicin (3 mg/kg weekly) and control groups (n=5, each). Surprisingly, no significant changes in LV-filling pattern were observed through the study, most likely due to the xylazine-containing anesthesia, necessary for appropriate resolving of the E and A waves. In contrast to the echographic measurement, the dP/dt(min) index obtained invasively at the end of the study revealed a significant impairment in LV relaxation, which was further supported by observed disturbances in myocardial collagen content and calcium homeostasis. However, at the same time cTnT plasma levels were progressively rising in the daunorubicin-treated animals from the fifth week (0.024+/-0.008 microg/l) until the end of the experiment (0.186+/-0.055 microg/l). Therefore, in contrast to complicated non-invasive evaluation of diastolic function, cTnT is shown to be an early and sensitive marker of anthracycline-induced cardiotoxicity in the rabbit model.
- Published
- 2007
- Full Text
- View/download PDF
22. Iron is not involved in oxidative stress-mediated cytotoxicity of doxorubicin and bleomycin.
- Author
-
Kaiserová H, den Hartog GJ, Simůnek T, Schröterová L, Kvasnicková E, and Bast A
- Subjects
- Aldehydes pharmacology, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Deferoxamine pharmacology, Electron Spin Resonance Spectroscopy, Free Radicals chemistry, Humans, Hydrazones pharmacology, Hydrogen Peroxide chemistry, Iron chemistry, Iron Chelating Agents chemistry, Iron Compounds chemistry, Isoniazid analogs & derivatives, Isoniazid pharmacology, Lipid Peroxidation drug effects, Lung Neoplasms pathology, Pyridoxal analogs & derivatives, Pyridoxal pharmacology, Razoxane pharmacology, Time Factors, Antibiotics, Antineoplastic toxicity, Bleomycin toxicity, Doxorubicin toxicity, Iron Chelating Agents pharmacology, Iron Compounds metabolism, Lung Neoplasms metabolism, Oxidative Stress drug effects
- Abstract
Background and Purpose: The anticancer drugs doxorubicin and bleomycin are well-known for their oxidative stress-mediated side effects in heart and lung, respectively. It is frequently suggested that iron is involved in doxorubicin and bleomycin toxicity. We set out to elucidate whether iron chelation prevents the oxidative stress-mediated toxicity of doxorubicin and bleomycin and whether it affects their antiproliferative/proapoptotic effects., Experimental Approach: Cell culture experiments were performed in A549 cells. Formation of hydroxyl radicals was measured in vitro by electron paramagnetic resonance (EPR). We investigated interactions between five iron chelators and the oxidative stress-inducing agents (doxorubicin, bleomycin and H(2)O(2)) by quantifying oxidative stress and cellular damage as TBARS formation, glutathione (GSH) consumption and lactic dehydrogenase (LDH) leakage. The antitumour/proapoptotic effects of doxorubicin and bleomycin were assessed by cell proliferation and caspase-3 activity assay., Key Results: All the tested chelators, except for monohydroxyethylrutoside (monoHER), prevented hydroxyl radical formation induced by H(2)O(2)/Fe(2+) in EPR studies. However, only salicylaldehyde isonicotinoyl hydrazone and deferoxamine protected intact A549 cells against H(2)O(2)/Fe(2+). Conversely, the chelators that decreased doxorubicin and bleomycin-induced oxidative stress and cellular damage (dexrazoxane, monoHER) were not able to protect against H(2)O(2)/Fe(2+)., Conclusions and Implications: We have shown that the ability to chelate iron as such is not the sole determinant of a compound protecting against doxorubicin or bleomycin-induced cytotoxicity. Our data challenge the putative role of iron and hydroxyl radicals in the oxidative stress-mediated cytotoxicity of doxorubicin and bleomycin and have implications for the development of new compounds to protects against this toxicity.
- Published
- 2006
- Full Text
- View/download PDF
23. Myocardial regulatory proteins and heart failure.
- Author
-
Adamcová M, Stĕrba M, Simůnek T, Potácová A, Popelová O, and Gersl V
- Subjects
- Humans, Cardiac Output, Low physiopathology, Troponin I physiology, Troponin T physiology
- Abstract
Cardiac troponin T (cTnT) and cardiac troponin I (cTnI) are considered to be the most specific and sensitive biochemical markers of myocardial damage. Troponins have been studied in a wide range of clinical settings, including heart failure; however, there are few data on the role of regulatory proteins in the pathogenesis of heart failure, although a few interesting hypotheses have been proposed. A considerable body of evidence favours the view that alteration of the myocardial thin filament is the primary event leading to defective contractility of the failing myocardium, while the changes in Ca(2+) handling are a compensatory response. A better understanding of the role of regulatory proteins under different physiological and pathological conditions could lead to new therapeutic approaches in heart failure. Recently, calcium sensitisation has been proposed as a novel method by which cardiac performance may be enhanced via an increase in the affinity of troponin C for calcium but without affecting intracellular calcium concentration. To date, the only calcium sensitizer used in clinical practice is levosimendan.
- Published
- 2006
- Full Text
- View/download PDF
24. The role of reactive oxygen and nitrogen species in cellular iron metabolism.
- Author
-
Mladenka P, Simůnek T, Hübl M, and Hrdina R
- Subjects
- Animals, Humans, Iron metabolism, Iron-Sulfur Proteins metabolism, Reactive Nitrogen Species metabolism, Reactive Oxygen Species metabolism
- Abstract
The catalytic role of iron in the Haber-Weiss chemistry, which results in propagation of damaging reactive oxygen species (ROS), is well established. In this review, we attempt to summarize the recent evidence showing the reverse: That reactive oxygen and nitrogen species can significantly affect iron metabolism. Their interaction with iron-regulatory proteins (IRPs) seems to be one of the essential mechanisms of influencing iron homeostasis. Iron depletion is known to provoke normal iron uptake via IRPs, superoxide and hydrogen peroxide are supposed to cause unnecessary iron uptake by similar mechanism. Furthermore, ROS are able to release iron from iron-containing molecules. On the contrary, nitric oxide (NO) appears to be involved in cellular defense against the iron-mediated ROS generation probably mainly by inducing iron removal from cells. In addition, NO may attenuate the effect of superoxide by mutual reaction, although the reaction product-peroxynitrite-is capable to produce highly reactive hydroxyl radicals.
- Published
- 2006
- Full Text
- View/download PDF
25. Safety and tolerability of repeated administration of pyridoxal 2-chlorobenzoyl hydrazone in rabbits.
- Author
-
Sterba M, Simůnek T, Mazurová Y, Adamcová M, Popelová O, Kaplanova J, Ponka P, and Gersl V
- Subjects
- Animals, Blood Cell Count, Body Weight drug effects, Dose-Response Relationship, Drug, Enzymes blood, Hydrazones administration & dosage, Hydrazones pharmacokinetics, Iron Chelating Agents administration & dosage, Male, Microscopy, Electron, Scanning, Pyridoxal administration & dosage, Pyridoxal pharmacokinetics, Pyridoxal toxicity, Rabbits, Time Factors, Tissue Distribution, Troponin T blood, Hydrazones toxicity, Iron Chelating Agents toxicity, Pyridoxal analogs & derivatives
- Abstract
Recently, pyridoxal 2-chlorobenzoyl hydrazone (o-108) has been identified as an effective iron chelator [Link et al., Blood 2003; 101: 4172-79]. Since chronic treatment would be necessary in its potential indications, in the present study, the safety and tolerability of this agent after repeated administration was determined. Three doses of o-108 (25, 50, 100 mg/kg, in 10% Cremophor EL) were administered intraperitoneally, once weekly, for 10 weeks to three groups (n=5 each) of Chinchilla male rabbits. The effects on biochemical, haematological and cardiovascular parameters were examined during the experiment; histopathological examination was performed at the end of the experiment. Results were compared with control (saline 2 mL/kg, n=11) and vehicle groups (10% Cremophor EL, 2 mL/kg, n=12). No premature deaths occurred; the well-being of animals was evidenced by their body weight gain, although lower gain was observed with the highest dose (100 mg/kg). Significant elevations of cardiac troponin T plasma concentrations were observed with the highest dose of o-108, but no abnormalities were found in the cardiovascular function and only minor and inconsistent changes in haematological and biochemical parameters were observed. Histopathological examinations of selected organs revealed only weak and reversible changes through all studied groups. Thus, the data from this study suggest that o-108 remains a promising drug from the standpoint of the possibility of its repeated administration and warrants further investigation.
- Published
- 2005
- Full Text
- View/download PDF
26. SIH--a novel lipophilic iron chelator--protects H9c2 cardiomyoblasts from oxidative stress-induced mitochondrial injury and cell death.
- Author
-
Simůnek T, Boer C, Bouwman RA, Vlasblom R, Versteilen AM, Sterba M, Gersl V, Hrdina R, Ponka P, de Lange JJ, Paulus WJ, and Musters RJ
- Subjects
- Aldehydes chemistry, Animals, Cell Death drug effects, Cell Line, Cell Shape, Cell Survival drug effects, Hydrazones chemistry, Hydrogen Peroxide pharmacology, Iron Chelating Agents chemistry, Membrane Potentials drug effects, Mitochondria metabolism, Mitochondria pathology, Myoblasts, Cardiac metabolism, Rats, Aldehydes pharmacology, Cytoprotection drug effects, Hydrazones pharmacology, Iron Chelating Agents pharmacology, Mitochondria drug effects, Myoblasts, Cardiac cytology, Myoblasts, Cardiac drug effects, Oxidative Stress
- Abstract
Recent evidence suggests that oxidative stress is a common denominator in many aspects of cardiovascular pathogenesis. Free cellular iron plays a crucial catalytic role in the formation of highly toxic hydroxyl radicals, and thereby it may aggravate the contribution of oxidative stress to cardiovascular disease. Therefore, iron chelation may be an effective therapeutic approach, but the progress in this area is hindered by the lack of effective agents. In this study, using the rat heart myoblast-derived cell line H9c2, we aimed to investigate whether the novel lipophilic iron chelator salicylaldehyde isonicotinoyl hydrazone (SIH) protects the cells against hydrogen peroxide (H2O2)-induced cytotoxicity. Exposure of cells to 100 micromol/l H2O2 has within 4 h induced a complete dissipation of their mitochondrial membrane potential (DeltaPsim). Co-treatment with SIH dose-dependently reduced (EC50=0.8 micromol/l) or even completely abolished (3 micromol/l) this collapse. Furthermore, the latter SIH concentration was capable to fully prevent alterations in cell morphology, and inhibited both apoptosis (annexin-V staining, nuclear chromatin shrinkage, TUNEL positivity) and necrosis (propidium iodide staining), even 24 h after the H2O2 exposure. In comparison, deferoxamin (a commercially available hydrophilic iron chelator used in clinical practice and most previous studies) was cytoprotective only at three-order higher and clinically unachievable concentrations (EC50=1300 micromol/l). Thus, in this study, we present iron chelation as a very powerful tool by which oxidative stress-induced myocardial damage can be prevented.
- Published
- 2005
- Full Text
- View/download PDF
27. Myocardial content of selected elements in experimental anthracycline-induced cardiomyopathy in rabbits.
- Author
-
Simůnek T, Sterba M, Holecková M, Kaplanová J, Klimtová I, Adamcová M, Gersl V, and Hrdina R
- Subjects
- Animals, Antibiotics, Antineoplastic toxicity, Cardiomyopathies metabolism, Disease Models, Animal, Iron metabolism, Magnesium metabolism, Male, Potassium metabolism, Rabbits, Razoxane toxicity, Selenium metabolism, Anthracyclines toxicity, Calcium metabolism, Cardiomyopathies chemically induced, Myocardium metabolism, Trace Elements metabolism
- Abstract
Cardiotoxicity represents the main drawback of clinical usefulness of anthracycline antineoplastic drugs. In this study, a content of selected elements (Ca, Mg, K, Se, Fe) in the post-mortem removed samples of the myocardial tissue was studied in three groups of rabbits: 1) control group (i.v. saline; n = 10); 2) daunorubicin-receiving animals (DAU; 3 mg/kg, i.v; n = 11); 3) animals receiving cardioprotective iron-chelating agent dexrazoxane (DEX; 60 mg/kg, i.p.; n = 5) prior to DAU. Drugs were administered once weekly for 10 weeks. 5-7 days after the last administration, cardiac left ventricular contractility (dP/dtmax) was significantly decreased in DAU-treated animals (745 +/- 69 versus 1245 +/- 86 kPa/s in the control group; P < 0.05), while in the DEX + DAU group it was insignificantly increased (1411 +/- 77 kPa/s). Of the myocardial elements' content studied, a significant increase in total Ca against control (16.2 +/- 2.4 versus 10.6 +/- 0.9 microg/g of dry tissue; P < 0.05) was determined in the DAU-group, which was accompanied with significant decreases in Mg and K. In the heart tissue of DEX-pretreated animals, no significant changes of elements' content were found as compared to controls, while the Ca content was in these animals significantly lower than in the DAU group (9.1 +/- 0.4 versus 16.2 +/- 2.4 microg/g; P < 0.05). Hence, in this study we show that systolic heart failure induced by chronic DAU administration is primarily accompanied by persistent calcium overload of cardiac tissue and the protective action of DEX is associated with the restoration of normal myocardial Ca content.
- Published
- 2005
- Full Text
- View/download PDF
28. The fate of iron in the organism and its regulatory pathways.
- Author
-
Mladenka P, Hrdina R, Hübl M, and Simůnek T
- Subjects
- Absorption, Animals, Humans, Intestine, Small metabolism, Iron pharmacokinetics, Iron-Regulatory Proteins metabolism, Iron metabolism
- Abstract
Iron is an essential element involved in many life-necessary processes. Interestingly, in mammals there is no active excretion mechanism for iron. Therefore iron kinetics has to be meticulously regulated. The most important step for regulation of iron kinetics is absorption. The absorption takes place in small intestine and it is implicated that it requires several proteins. Iron is then released from enterocytes into the circulation and delivered to the cells. Iron movement inside the cell is only partially elucidated and its traffic to mitochondia is not known. Surprisingly, the regulation of various proteins related to iron kinetics and energy metabolism at the molecular level is better described. On contrary, the complex control of iron absorption cannot be fully explicated with present knowledge.
- Published
- 2005
29. Rabbit model for in vivo study of anthracycline-induced heart failure and for the evaluation of protective agents.
- Author
-
Simůnek T, Klimtová I, Kaplanová J, Mazurová Y, Adamcová M, Sterba M, Hrdina R, and Gersl V
- Subjects
- Animals, Anthracyclines administration & dosage, Antibiotics, Antineoplastic administration & dosage, Antibiotics, Antineoplastic adverse effects, Biomarkers blood, Body Weight drug effects, Cardiovascular Agents administration & dosage, Daunorubicin administration & dosage, Daunorubicin adverse effects, Disease Models, Animal, Dose-Response Relationship, Drug, Echocardiography, Erythrocyte Indices drug effects, Heart Failure mortality, Heart Failure physiopathology, Heart Rate drug effects, Heart Ventricles diagnostic imaging, Heart Ventricles drug effects, Heart Ventricles pathology, Male, Models, Cardiovascular, Myocardium cytology, Myocardium pathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac pathology, Rabbits, Razoxane administration & dosage, Razoxane therapeutic use, Stroke Volume drug effects, Time Factors, Anthracyclines adverse effects, Cardiovascular Agents therapeutic use, Heart Failure chemically induced, Heart Failure prevention & control
- Abstract
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.
- Published
- 2004
- Full Text
- View/download PDF
30. Cardiac troponin T as an indicator of reduced left ventricular contractility in experimental anthracycline-induced cardiomyopathy.
- Author
-
Simůnek T, Klimtová I, Adamcová M, Gersl V, Hrdina R, Sterba M, Kaplanová J, and Mazurová Y
- Subjects
- Animals, Biomarkers, Chinchilla, Daunorubicin toxicity, Male, Myocardial Contraction drug effects, Rabbits, Ventricular Function, Left drug effects, Antibiotics, Antineoplastic toxicity, Cardiomyopathies chemically induced, Cardiomyopathies metabolism, Myocardium metabolism, Troponin T metabolism, Ventricular Dysfunction, Left chemically induced, Ventricular Dysfunction, Left metabolism
- Abstract
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.
- Published
- 2003
- Full Text
- View/download PDF
31. Protein profiling in daunorubicin-induced cardiomyopathy.
- Author
-
Adamcová M, Pelouch V, Gersl V, Kaplanová J, Mazurová Y, Simůnek T, Klimtová I, and Hrdina R
- Subjects
- Animals, Antibiotics, Antineoplastic toxicity, Cardiomyopathies drug therapy, Cardiomyopathies pathology, Cardiotonic Agents administration & dosage, Heart Ventricles pathology, Male, Rabbits, Cardiomyopathies chemically induced, Cardiomyopathies physiopathology, Daunorubicin toxicity, Heart Ventricles drug effects, Heart Ventricles physiopathology, Proteins metabolism, Razoxane administration & dosage
- Abstract
The aim of this paper was to study the protein remodelling of the left ventricle following repeated administration of either daunorubicin (DNR) or DNR in combination with the cardioprotective agent dexrazoxane (DXZ). The experiment was carried out on three groups of Chinchilla male rabbits: 1. DNR (3 mg/kg i.v.), 2. DNR (3 mg/kg i.v.) + DXZ (60 mg/kg i.p.), and 3. the control group (saline 1 ml/kg i.v. in the same schedule). The drugs were given once weekly, max. 10 administrations. Protein fractions were isolated by stepwise extraction from the samples of the left ventricle. In the DNR-group, the concentrations of both, metabolic and contractile proteins were significantly reduced, while the amount of collagen was significantly higher in comparison with the control group. In the group treated with DNR and DXZ, the concentrations of individual protein fractions (except metabolic proteins) were comparable to those of the control group, which confirms a significant cardioprotective effect of DXZ. The changes of protein profiling corresponded to functional examination of both cardiac parameters (EF, dP/dt(max), PEP: LVET index) and histological examination. These data should be used in further studies dealing with evaluation of cardiotoxic and, possibly, cardioprotective effects of new drugs.
- Published
- 2003
32. Cardiac troponins following repeated administration of an iron chelator--salicylaldehyde isonicotinoyl hydrazone (SIH)--in rabbits.
- Author
-
Adamcová M, Sterba M, Klimtová I, Simůnek T, Hrdina R, Gersl V, and Ponka P
- Subjects
- Animals, Biomarkers blood, Heart drug effects, Male, Rabbits, Aldehydes toxicity, Hydrazones toxicity, Iron Chelating Agents toxicity, Myocardium metabolism, Troponin I metabolism, Troponin T metabolism
- Abstract
Both cardiac troponin T (cTnT) and cardiac troponin I (cTnI) are considered to be reliable biomarkers with sufficient sensitivity and specificity for cardiac injury in the majority of laboratory animals. The aim of our study was to compare the diagnostic performance of cTnT and cTnI in three groups of rabbits: 1) control (saline 1 ml/kg i.v.); 2) Salicylaldehyde Isonicotinoyl Hydrazone--SIH (50 mg/kg, once weekly, i.p.; partially dissolved in 10% Cremophor solution); 3) 10% Cremophor solution in water (2 ml/kg i.v.). The drugs were given once a week, 10 administrations. The concentration of cTnT was measured using Elecsys Troponin T STAT Immunoassay (Roche). The concentration of cTnI was measured using AxSYM Troponin I (Abbott). The linear regression model was applied to see if there is a dependence between cTnT and cTnI. The coefficient of determination was not acceptable in all groups. The highest value of R2 was found in the control group (R2 = 0.424). We may conclude that in rabbits meaningful dependence between cTnT and cTnI was not found. According to our long-term experiences cTnT seems to be more suitable cardiomarker in rabbits in comparison with cTnI where the data are characterized by the large scatter.
- Published
- 2003
33. A study of potential toxic effects after repeated 10-week administration of a new iron chelator--salicylaldehyde isonicotinoyl hydrazone (SIH) to rabbits.
- Author
-
Klimtová I, Simůnek T, Mazurová Y, Kaplanová J, Sterba M, Hrdina R, Gersl V, Adamcová M, and Ponka P
- Subjects
- Animals, Male, Rabbits, Aldehydes toxicity, Hydrazones toxicity, Iron Chelating Agents toxicity
- Abstract
Salicylaldehyde Isonicotinoyl Hydrazone (SIH)--a Pyridoxal Isonicotinoyl Hydrazone (PIH) analogue--is an effective iron chelator with antioxidant and antimalarial effects, as documented in numerous in vitro studies. However, no toxicological data obtained from in vivo studies have been made available yet. In this study, the potential toxic effects of repeated administration of SIH (50 mg/kg, once weekly, 10 weeks, i.p.), partially dissolved in a 10% Cremophor solution, on various biochemical, haematological, and cardiovascular parameters and on morphology of selected tissues were investigated in rabbits. The obtained values were compared with data from the control (saline, 1 ml/kg, i.v.) and the Cremophor (10% Cremophor solution, 2 ml/kg, i.p.) groups. In this study, SIH did not induced marked signs of toxicity: No premature deaths occurred, the body weight increase was comparable with the control and Cremophor groups. Only few and mild changes in some biochemical and haematological parameters could be determined, most of them were noticed also in the control or Cremophor groups. The morphological changes in the kidney were mild and did not manifest in the biochemical examination. The cardiac function was also not affected markedly--the values of left ventricular ejection fraction and systolic time interval did not differ from the values of control group. Only an increased left ventricular contractility (dP/dtmax) was noticed in the SIH group at the end of the experiment as compared to the controls (13,354+/-1191 vs. 9339+/-647 mmHg/s, resp.). These results seem to be promising from the standpoint of possible clinical use of SIH.
- Published
- 2003
34. Comparative study of chronic toxic effects of daunorubicin and doxorubicin in rabbits.
- Author
-
Klimtová I, Simůnek T, Mazurová Y, Hrdina R, Gersl V, and Adamcová M
- Subjects
- Animals, Body Weight drug effects, Clinical Chemistry Tests, Disease Models, Animal, Heart drug effects, Heart Failure chemically induced, Heart Failure pathology, Heart Ventricles drug effects, Heart Ventricles pathology, Heart Ventricles physiopathology, Hemodynamics drug effects, Kidney drug effects, Kidney pathology, Male, Myocardium pathology, Rabbits, Toxicity Tests, Chronic, Ventricular Dysfunction, Left chemically induced, Ventricular Dysfunction, Left pathology, Antibiotics, Antineoplastic toxicity, Daunorubicin toxicity, Doxorubicin toxicity
- Abstract
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.
- Published
- 2002
- Full Text
- View/download PDF
35. Cardiac troponin T following repeated administration of pyridoxal isonicotinoyl hydrazone in rabbits.
- Author
-
Adamcová M, Machácková J, Gersl V, Pelouch V, Simůnek T, Klimtová I, Hrdina R, and Ponka P
- Subjects
- Animals, Cell Fractionation methods, Cytosol metabolism, Iron Overload drug therapy, Male, Myofibrils metabolism, Rabbits, Iron Chelating Agents toxicity, Isoniazid analogs & derivatives, Isoniazid toxicity, Myocardium metabolism, Pyridoxal analogs & derivatives, Pyridoxal toxicity, Troponin T blood
- Abstract
Pyridoxal isonicotinoyl hydrazone (PIH) is a new tridentate Fe-chelating agent that should be very promising in many pathological states resulting from both an iron-overload and formation of free radicals. The aim of our study was to investigate the effect of PIH on the cardiovascular system focusing to the regulatory protein -- cardiac troponin T (cTnT). The study was carried out in two groups of Chinchilla male rabbits: 1) PIH (50 mg/kg dissolved in 10 % Cremophor i.p., once a week, 10 administrations, n=8) and 2) Cremophor (2 ml/kg i.p. in the same schedule, n=7). Plasma concentrations of cTnT (as a marker of myocardial damage) were measured using a commercial kit (Roche). cTnT was within the physiological range (i.e. < 0.1 microg/l) during the whole experiment in the Cremophor group. In the PIH group, the cTnT levels were not significantly increased when compared with the control group or with the initial values (except with those before the 5th administration). Furthermore, we analyzed the cytosolic and myofibrillar fraction of cTnT in the left ventricular myocardium. Using SDS-PAGE and Western blot we resolved three isoforms. The profiling of TnT did not differ significantly between the PIH-treated group and the Cremophor-treated group. Our data concerning cTnT support the opinion that the possible cardiotoxicity of PIH is very low.
- Published
- 2002
36. Effect of sodium 2,3-dimercaptopropane-1-sulphonate (DMPS) on chronic daunorubicin toxicity in rabbits: comparison with dexrazoxane.
- Author
-
Hrdina R, Gersl V, Klimtová I, Simůnek T, Mazurová Y, Machácková J, and Adamcová M
- Subjects
- Animals, Calcium analysis, Iron analysis, Male, Myocardium chemistry, Myocardium pathology, Rabbits, Antibiotics, Antineoplastic toxicity, Chelating Agents pharmacology, Daunorubicin toxicity, Heart drug effects, Razoxane pharmacology, Unithiol pharmacology
- Abstract
A possible protective action of DMPS (a dithiol chelating agent) against chronic daunorubicin toxicity in rabbits in comparison with dexrazoxane was investigated. The rabbits were divided into five groups: control (saline, 1 ml/kg i.v.), daunorubicin (3 mg/kg i.v.), DMPS (50 mg/kg i.v.); the remaining two groups were pre-treated either with dexrazoxane (60 mg/kg i.p.) or DMPS (50 mg/kg i.v.) 30 min before administration of daunorubicin (3 mg/kg i.v.). Drugs were given once a week for 10 weeks. Routine biochemical parameters were determined in weeks 1, 5 and 11. In the 11th week, invasive haemodynamic parameters were measured, then the rabbits underwent autopsy, cardiac tissue was examined by light microscopy and scored semiquantitatively. The contents of calcium, potassium, magnesium, iron and selenium were measured in the left heart ventricle. DMPS administered alone was well tolerated and did not cause any major signs of toxicity. It decreased the cardiac content of calcium, but did not affect the iron concentration. In contrast to dexrazoxane, DMPS pre-treatment did not prevent the decline in body weight in weeks 8-11 caused by daunorubicin, actually worsened mortality (26.7% vs 40.0%), did not ameliorate daunorubicin-induced nephrotic syndrome, and did not prevent the occurrence of the severe myocardial lesions. Unlike dexrazoxane, a lack of protective effect of DMPS against chronic daunorubicin toxicity in rabbits was demonstrated. The underlying cause may consist in the fact that DMPS does not efficiently chelate tissue iron and thus may not prevent the formation of oxygen free radicals.
- Published
- 2002
37. Troponins in experimental studies.
- Author
-
Adamcová M, Gersl V, Machácková J, Hrdina R, Klimtová I, Simůnek T, Vávrová J, and Bukac J
- Subjects
- Animals, Biomarkers blood, Cardiomyopathies diagnosis, Heart drug effects, Male, Protective Agents administration & dosage, Rabbits, Razoxane administration & dosage, Antibiotics, Antineoplastic toxicity, Cardiomyopathies chemically induced, Daunorubicin toxicity, Troponin T blood
- Abstract
The aim of our study was to compare the diagnostic performance of cardiac troponin T (cTnT) and cardiac troponin I (cTnI) in three groups of rabbits: 1) control (saline 1 ml/kg i.v.); 2) daunorubicin (3 mg/kg i.v.); 3) daunorubicin (3 mg/kg i.v.) + dexrazoxane (60 mg/kg i.p.). The drugs were given once a week, 10 administrations. The concentration of cTnT was measured using Elecsys Troponin T STAT Immunoassay (Roche). The concentration of cTnI was measured using AxSYM Troponin I (Abbott). The linear regression model was applied to see if there is a dependence between cTnT and cTnI. The coefficient of determination (R2 = 0.79) was acceptable only in the control group. In the remaining cases (i.e. in the daunorubicin group and in the daunorubicin + dexrazoxane treated group) R2 was too small (0.53, and 0.06). We may conclude that in rabbits after repeated administration of cardiotoxic or cardioprotective drugs meaningful dependence between cTnT and cTnI was not found. The choice of the most suitable cardiomarker in laboratory animals deserves further studies.
- Published
- 2002
38. Anthracycline-induced cardiotoxicity.
- Author
-
Hrdina R, Gersl V, Klimtová I, Simůnek T, Machácková J, and Adamcová M
- Subjects
- Animals, Antibiotics, Antineoplastic metabolism, Drug Monitoring, Heart Diseases chemically induced, Heart Diseases prevention & control, Humans, Antibiotics, Antineoplastic adverse effects, Heart drug effects
- Abstract
Anthracycline antibiotics are among the most effective and widely used antineoplastic drugs. Their usefulness is limited by a cumulative dose-related cardiotoxicity, whose precise mechanisms are not clear as yet. The principal role is possibly exerted by free oxygen radicals generated by "redox-cycling" of anthracycline molecule and/or by the formation of anthracycline-ferric ion complexes. The iron catalyzes the hydroxyl radical production via Haber-Weiss reaction. The selective toxicity of ANT against cardiomyocytes results from high accumulation of ANT in cardiac tissue, appreciable production of oxygen radicals by mitochondria and relatively poor antioxidant defense systems. Other additional mechanisms of the anthracycline cardiotoxicity have been proposed--calcium overload, histamine release and impairment in autonomic regulation of heart function. The currently used methods for an early identification of anthracycline cardiotoxicity comprise ECG measurement, biochemical markers, functional measurement and morphologic examination. Among a plenty of studied cardioprotective agents only dexrazoxane (ICRF-187) has been approved for clinical use. Its protective effect likely consists in intracellular chelating of iron. However, in high doses dexrazoxane itself may cause myelotoxicity. This fact encourages investigation of new cardioprotectants with lower toxicity. Orally active iron chelators and flavonoids attract more attention. Modification of dosage schedule and synthesis of new anthracycline analogues may represent alternative approaches to mitigate anthracycline cardiotoxicity while preserving antitumour activity.
- Published
- 2000
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.