Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Folia Microbiol (Praha) ; 67(1): 55-61, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34499312

ABSTRACT

Identification of filamentous fungi based on morphological features is the most available approach used in clinical mycology laboratories. However, MALDI-TOF mass spectrometry is currently invaluable for identification of microorganisms because of its rapidity, simplicity, and accuracy. This study aimed to find the optimal way of identifying filamentous fungi using MALDI-TOF MS.The sample comprised 193 isolates of filamentous fungi. The identification started with morphological assessment. Then isolates were identified using MALDI-TOF MS, both directly from culture and following culture in liquid media with extraction. Subsequently, identification of 20 selected isolates was compared by sequencing of the benA gene, ITS1-5,8-ITS2, and D1-D2 LSU regions.Based on morphological criteria, 17 genera of fungi were identified. With MALDI-TOF MS performed directly from culture, nine isolates were identified to the genus level and 184 to the species level, with a total of 75 species being noted. With the MALDI-TOF MS extraction method, 190 isolates were identified to the species level, with 43 species being noted. The rates of agreement between identification using morphology and the MALDI-TOF MS direct method were 58.55% at the genus level and 22.24% at the species level. The rates of agreement between identification using morphology and the MALDI-TOF MS extraction method were 84.97% at the genus level and 46.11% at the species level. Using sequencing, 87.5% agreement was found for identification with the MALDI-TOF MS extraction method, as compared with only 43.75% for the direct method.The results suggest that the optimal approach to identification of filamentous fungi is a combination of morphological features and MALDI-TOF MS using the extraction method.


Subject(s)
Arthrodermataceae , Arthrodermataceae/genetics , Culture Media , Diagnostic Tests, Routine , Fungi/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
2.
Front Cell Dev Biol ; 9: 702986, 2021.
Article in English | MEDLINE | ID: mdl-34381783

ABSTRACT

Cephalochordates (amphioxi or lancelets) are representatives of the most basally divergent group of the chordate phylum. Studies of amphioxus development and anatomy hence provide a key insight into vertebrate evolution. More widespread use of amphioxus in the evo-devo field would be greatly facilitated by expanding the methodological toolbox available in this model system. For example, evo-devo research on amphioxus requires deep understanding of animal anatomy. Although conventional confocal microscopy can visualize transparent amphioxus embryos and early larvae, the imaging of later developmental stages is problematic because of the size and opaqueness of the animal. Here, we show that light sheet microscopy combined with tissue clearing methods enables exploration of large amphioxus specimens while keeping the surface and the internal structures intact. We took advantage of the phenomenon of autofluorescence of amphioxus larva to highlight anatomical details. In order to investigate molecular markers at the single-cell level, we performed antibody-based immunodetection of melanopsin and acetylated-α-tubulin to label rhabdomeric photoreceptors and the neuronal scaffold. Our approach that combines light sheet microscopy with the clearing protocol, autofluorescence properties of amphioxus, and antibody immunodetection allows visualizing anatomical structures and even individual cells in the 3D space of the entire animal body.

3.
Front Cell Dev Biol ; 8: 705, 2020.
Article in English | MEDLINE | ID: mdl-32850825

ABSTRACT

The evolution of the vertebrate eye remains so far unresolved. Amphioxus frontal eye pigment cells and photoreceptors were proposed to be homologous to vertebrate photoreceptors and retinal pigmented epithelium, based on ultrastructural morphology and gene expression analysis in B. floridae. Here, we present comparative molecular data using two additional amphioxus species, a closely related B. lanceolatum, and the most divergent A. lucayanum. Taking advantage of a unique set of specific antibodies we characterized photoreceptors and putative interneurons of the frontal eye and investigated its neuronal circuitry. Our results corroborate generally conserved molecular fingerprint among cephalochordate species. Furthermore, we performed pharmacological perturbations and found that the Notch signaling pathway, a key regulator of retina development in vertebrates, is required for correct ratios among frontal eye cell types. In summary, our study provides a valuable insight into cell-type relationships in chordate visual organs and strengthens the previously proposed homology between amphioxus frontal eye and vertebrate eyes.

4.
Toxicology ; 372: 52-63, 2016 Nov 30.
Article in English | MEDLINE | ID: mdl-27816693

ABSTRACT

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


Subject(s)
Anthracyclines/toxicity , Antibiotics, Antineoplastic/toxicity , Cardiotonic Agents/pharmacology , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Molsidomine/pharmacology , Nitric Oxide Donors/pharmacology , Animals , Cardiotoxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Chronic Disease , Daunorubicin/toxicity , Doxorubicin/toxicity , Heart Failure/prevention & control , Lipid Peroxidation/drug effects , Oxidative Stress/drug effects , Rabbits , Reactive Oxygen Species/metabolism , Ventricular Remodeling/drug effects
5.
J Mol Cell Cardiol ; 91: 92-103, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26724189

ABSTRACT

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


Subject(s)
Cardiotonic Agents/pharmacology , Cardiotoxicity/prevention & control , Dexrazoxane/pharmacology , Nitrates/pharmacology , Sodium Nitrite/pharmacology , Animals , Antibiotics, Antineoplastic/adverse effects , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Daunorubicin/adverse effects , Drug Administration Schedule , Infusions, Intravenous , Male , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rabbits
6.
PLoS One ; 9(2): e88754, 2014.
Article in English | MEDLINE | ID: mdl-24586383

ABSTRACT

Recent studies have demonstrated that several chelators possess marked potential as potent anti-neoplastic drugs and as agents that can ameliorate some of the adverse effects associated with standard chemotherapy. Anti-cancer treatment employs combinations of several drugs that have different mechanisms of action. However, data regarding the potential interactions between iron chelators and established chemotherapeutics are lacking. Using estrogen receptor-positive MCF-7 breast cancer cells, we explored the combined anti-proliferative potential of four iron chelators, namely: desferrioxamine (DFO), salicylaldehyde isonicotinoyl hydrazone (SIH), (E)-N'-[1-(2-hydroxy-5-nitrophenyl)ethyliden] isonicotinoyl hydrazone (NHAPI), and di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT), plus six selected anti-neoplastic drugs. These six agents are used for breast cancer treatment and include: paclitaxel, 5-fluorouracil, doxorubicin, methotrexate, tamoxifen and 4-hydroperoxycyclophosphamide (an active metabolite of cyclophosphamide). Our quantitative chelator-drug analyses were designed according to the Chou-Talalay method for drug combination assessment. All combinations of these agents yielded concentration-dependent, anti-proliferative effects. The hydrophilic siderophore, DFO, imposed antagonism when used in combination with all six anti-tumor agents and this antagonistic effect increased with increasing dose. Conversely, synergistic interactions were observed with combinations of the lipophilic chelators, NHAPI or Dp44mT, with doxorubicin and also the combinations of SIH, NHAPI or Dp44mT with tamoxifen. The combination of Dp44mT with anti-neoplastic agents was further enhanced following formation of its redox-active iron and especially copper complexes. The most potent combinations of Dp44mT and NHAPI with tamoxifen were confirmed as synergistic using another estrogen receptor-expressing breast cancer cell line, T47D, but not estrogen receptor-negative MDA-MB-231 cells. Furthermore, the synergy of NHAPI and tamoxifen was confirmed using MCF-7 cells by electrical impedance data, a mitochondrial inner membrane potential assay and cell cycle analyses. This is the first systematic investigation to quantitatively assess interactions between Fe chelators and standard chemotherapies using breast cancer cells. These studies are vital for their future clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Proliferation/drug effects , Iron Chelating Agents/pharmacology , Aldehydes/pharmacology , Cyclophosphamide/analogs & derivatives , Deferoxamine/pharmacology , Doxorubicin , Drug Synergism , Fluorouracil , Humans , Hydrazones/pharmacology , MCF-7 Cells , Methotrexate , Paclitaxel , Tamoxifen , Thiosemicarbazones/pharmacology
7.
PLoS One ; 8(10): e76676, 2013.
Article in English | MEDLINE | ID: mdl-24116135

ABSTRACT

Anthracyclines (such as doxorubicin or daunorubicin) are among the most effective anticancer drugs, but their usefulness is hampered by the risk of irreversible cardiotoxicity. Dexrazoxane (ICRF-187) is the only clinically approved cardioprotective agent against anthracycline cardiotoxicity. Its activity has traditionally been attributed to the iron-chelating effects of its metabolite with subsequent protection from oxidative stress. However, dexrazoxane is also a catalytic inhibitor of topoisomerase II (TOP2). Therefore, we examined whether dexrazoxane and two other TOP2 catalytic inhibitors, namely sobuzoxane (MST-16) and merbarone, protect cardiomyocytes from anthracycline toxicity and assessed their effects on anthracycline antineoplastic efficacy. Dexrazoxane and two other TOP2 inhibitors protected isolated neonatal rat cardiomyocytes against toxicity induced by both doxorubicin and daunorubicin. However, none of the TOP2 inhibitors significantly protected cardiomyocytes in a model of hydrogen peroxide-induced oxidative injury. In contrast, the catalytic inhibitors did not compromise the antiproliferative effects of the anthracyclines in the HL-60 leukemic cell line; instead, synergistic interactions were mostly observed. Additionally, anthracycline-induced caspase activation was differentially modulated by the TOP2 inhibitors in cardiac and cancer cells. Whereas dexrazoxane was upon hydrolysis able to significantly chelate intracellular labile iron ions, no such effect was noted for either sobuzoxane or merbarone. In conclusion, our data indicate that dexrazoxane may protect cardiomyocytes via its catalytic TOP2 inhibitory activity rather than iron-chelation activity. The differential expression and/or regulation of TOP2 isoforms in cardiac and cancer cells by catalytic inhibitors may be responsible for the selective modulation of anthracycline action observed.


Subject(s)
Anthracyclines/pharmacology , Cell Proliferation/drug effects , Myocytes, Cardiac/drug effects , Topoisomerase II Inhibitors/pharmacology , Animals , Animals, Newborn , Biocatalysis/drug effects , Caspases/metabolism , Cell Cycle/drug effects , Cell Survival/drug effects , Cells, Cultured , DNA Topoisomerases, Type II/metabolism , Daunorubicin/pharmacology , Dexrazoxane/pharmacology , Doxorubicin/pharmacology , Drug Interactions , Flow Cytometry , Glutathione/metabolism , Glutathione Disulfide/metabolism , HL-60 Cells , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Piperazines/pharmacology , Rats , Rats, Wistar , Thiobarbiturates/pharmacology
8.
J Pharm Biomed Anal ; 76: 243-51, 2013 Mar 25.
Article in English | MEDLINE | ID: mdl-23339990

ABSTRACT

Dexrazoxane (DEX) is the only clinically used drug effective against anthracycline-induced cardiotoxicity and extravasation injury. However, the mechanism of its cardioprotective action still remains elusive. This paucity of comprehensive data is at least partially caused by the analytical difficulties associated with selective and sensitive simultaneous determination of the parent drug and its putative active metabolite ADR-925 in the relevant biological material. The aim of this study was to develop and validate the first LC-MS/MS method for simultaneous determination of DEX and ADR-925 in the isolated rat neonatal ventricular cardiomyocytes (NVCMs) and the cell culture medium. The analysis was performed on a Synergi Polar-RP column using the gradient profile of the mobile phase composed of 2mM ammonium formate and methanol. Electrospray ionization and ion trap mass analyzer were used as ionization and detection techniques, respectively. NVCMs were precipitated with methanol and the cell culture medium samples were diluted with the same solvent prior the LC-MS/MS analysis. The method was validated within the range of 4-80pmol/10(6) NVCMs and 7-70pmol/10(6) NVCMs for DEX and ADR-925, respectively, and at the concentrations of 8-100µM for both compounds in the culture cell medium. The practical applicability of this method was confirmed by the pilot analysis of NVCMs and the corresponding cell medium samples from relevant in vitro experiment. Hence, the LC-MS/MS method developed in this study represents a modern analytical tool suitable for investigation of DEX bioactivation inside the cardiomyocytes. In addition, the basic utility of the method for the analysis of DEX and ADR-925 in plasma samples was proved in a pilot experiment.


Subject(s)
Chromatography, Liquid/methods , Ethylenediamines/pharmacokinetics , Glycine/analogs & derivatives , Razoxane/pharmacokinetics , Tandem Mass Spectrometry/methods , Animals , Animals, Newborn , Cardiovascular Agents/pharmacokinetics , Cells, Cultured , Glycine/pharmacokinetics , Myocytes, Cardiac/metabolism , Pilot Projects , Rabbits , Rats , Rats, Wistar , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization/methods
9.
Antioxid Redox Signal ; 18(8): 899-929, 2013 Mar 10.
Article in English | MEDLINE | ID: mdl-22794198

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.


Subject(s)
Anthracyclines/adverse effects , Chelating Agents/pharmacology , Heart/drug effects , Metals/adverse effects , Myocardium/metabolism , Oxidative Stress , Signal Transduction , 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
10.
J Pharmacol Exp Ther ; 343(2): 468-78, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22915767

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.


Subject(s)
Anthracyclines/toxicity , Antibiotics, Antineoplastic/toxicity , Heart Diseases/chemically induced , Heart Diseases/metabolism , Mitochondria, Heart/metabolism , NF-E2-Related Factor 2/biosynthesis , 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
11.
Chem Biol Interact ; 197(2-3): 69-79, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22521999

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.


Subject(s)
Aldehydes/chemistry , Antineoplastic Agents/pharmacology , Hydrazones/chemistry , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , 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
12.
Int J Biochem Cell Biol ; 44(6): 834-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22465709

ABSTRACT

DNA topoisomerases II regulate conformational changes in DNA topology. They act on double-stranded DNA, catalyzing its relaxation, decatenation and unknotting. Vertebrate cells express two isoforms of topoisomerase II, which are similar in structure, but different in function and regulation. Whereas the alpha isoform is indispensable for proper cell replication, the functions of the beta isoform as well as reasons for its evolution in vertebrates were long unclear. Unlike topoisomerase II alpha, the beta isoform is predominantly expressed in quiescent cells and has been implicated mainly in the process of gene transcription. Recently, new discoveries point on the role of the topoisomerase II beta in regulation of cellular differentiation and tissue development. Furthermore, contemporary discoveries are raising possibilities for novel therapeutic approaches involving selective targeting of either topoisomerase II isoform in potentiating antitumor and/or reducing adverse effects of topoisomerase II poisons.


Subject(s)
Cell Differentiation/physiology , DNA Topoisomerases, Type II/physiology , DNA-Binding Proteins/physiology , Gene Expression Regulation/physiology , Animals , Humans
13.
Toxicology ; 289(2-3): 122-31, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21864640

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.


Subject(s)
Cardiotonic Agents/pharmacology , Cardiotoxins/toxicity , Catecholamines/toxicity , Iron Chelating Agents/pharmacology , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Animals , Cardiotoxins/antagonists & inhibitors , Catecholamines/antagonists & inhibitors , Cell Line , Myocytes, Cardiac/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/physiology , Rats
14.
Chem Res Toxicol ; 24(3): 290-302, 2011 Mar 21.
Article in English | MEDLINE | ID: mdl-21214215

ABSTRACT

Oxidative stress is known to contribute to a number of cardiovascular pathologies. Free intracellular iron ions participate in the Fenton reaction and therefore substantially contribute to the formation of highly toxic hydroxyl radicals and cellular injury. Earlier work on the intracellular iron chelator salicylaldehyde isonicotinoyl hydrazone (SIH) has demonstrated its considerable promise as an agent to protect the heart against oxidative injury both in vitro and in vivo. However, the major limitation of SIH is represented by its labile hydrazone bond that makes it prone to plasma hydrolysis. Hence, in order to improve the hydrazone bond stability, nine compounds were prepared by a substitution of salicylaldehyde by the respective methyl- and ethylketone with various electron donors or acceptors in the phenyl ring. All the synthesized aroylhydrazones displayed significant iron-chelating activities and eight chelators showed significantly higher stability in rabbit plasma than SIH. Furthermore, some of these chelators were observed to possess higher cytoprotective activities against oxidative injury and/or lower toxicity as compared to SIH. The results of the present study therefore indicate the possible applicability of several of these novel agents in the prevention and/or treatment of cardiovascular disorders with a known (or presumed) role of oxidative stress. In particular, the methylketone HAPI and nitro group-containing NHAPI merit further in vivo investigations.


Subject(s)
Aldehydes/chemistry , Antioxidants/chemistry , Hydrazones/chemistry , Iron Chelating Agents/chemical synthesis , Aldehydes/blood , Aldehydes/pharmacology , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Cell Line , Drug Stability , Hydrazones/blood , Hydrazones/pharmacology , Hydrolysis , Hydroxyl Radical/toxicity , Iron Chelating Agents/metabolism , Iron Chelating Agents/pharmacology , Oxidative Stress , Rabbits , Rats
15.
Free Radic Biol Med ; 50(4): 537-49, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21147217

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.


Subject(s)
Aldehydes/pharmacology , Catecholamines/metabolism , Coordination Complexes/pharmacology , Hydrazones/pharmacology , Iron Chelating Agents/pharmacology , Iron/metabolism , Myoblasts, Cardiac/drug effects , 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
16.
Arch Toxicol ; 85(5): 525-35, 2011 May.
Article in English | MEDLINE | ID: mdl-21046361

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.


Subject(s)
Antioxidants/pharmacology , Daunorubicin/adverse effects , Glutathione/pharmacology , Heart Diseases/chemically induced , 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
17.
Chem Res Toxicol ; 23(6): 1105-14, 2010 Jun 21.
Article in English | MEDLINE | ID: mdl-20521781

ABSTRACT

Iron imbalance plays an important role in oxidative stress associated with numerous pathological conditions. Therefore, iron chelation may be an effective therapeutic approach, but progress in this area is hindered by the lack of effective ligands. Also, the potential favorable effects of chelators against oxidative injury have to be balanced against their own toxicity due to iron depletion and the ability to generate redox-active iron complexes. In this study, we compared selected iron chelators (both drugs used in clinical practice as well as experimental agents) for their efficacy to protect cells against model oxidative injury induced by tert-butyl hydroperoxide (t-BHP). In addition, intracellular chelation efficiency, redox activity, and the cytotoxicity of the chelators and their iron complexes were assayed. Ethylenediaminetetraacetic acid failed to protect cells against t-BHP cytotoxicity, apparently due to the redox activity of the formed iron complex. Hydrophilic desferrioxamine exerted some protection but only at very high clinically unachievable concentrations. The smaller and more lipophilic chelators, deferiprone, deferasirox, and pyridoxal isonicotinoyl hydrazone, were markedly more effective at preventing oxidative injury of cells. The most effective chelator in terms of access to the intracellular labile iron pool was di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone. However, overall, the most favorable properties in terms of protective efficiency against t-BHP and the chelator's own inherent cytotoxicity were observed with salicylaldehyde isonicotinoyl hydrazone. This probably relates to the optimal lipophilicity of this latter agent and its ability to generate iron complexes that do not induce marked redox activity.


Subject(s)
Cytoprotection/drug effects , Iron Chelating Agents/chemistry , Iron Chelating Agents/pharmacology , Oxidative Stress/drug effects , Animals , Cell Line , Cell Survival/drug effects , Myocardium/cytology , tert-Butylhydroperoxide/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...