Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 445
Filter
1.
Toxicol Sci ; 198(2): 288-302, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38290791

ABSTRACT

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


Subject(s)
Anthracyclines , Quinoxalines , Topoisomerase II Inhibitors , Rats , Animals , Rabbits , Topoisomerase II Inhibitors/toxicity , Topoisomerase II Inhibitors/therapeutic use , Anthracyclines/toxicity , Anthracyclines/therapeutic use , Cardiotoxicity , Daunorubicin/toxicity , Daunorubicin/therapeutic use , Doxorubicin/toxicity , Antibiotics, Antineoplastic/toxicity , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type II/therapeutic use , DNA Damage
2.
Sci Rep ; 13(1): 21054, 2023 11 29.
Article in English | MEDLINE | ID: mdl-38030645

ABSTRACT

Liposomal formulations are hypothesized to alleviate anthracycline cardiotoxicity, although this has only been documented clinically for doxorubicin. We developed an in vitro multiparametric model using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) to assess the relative toxicity of anthracyclines across formulations. Proof of concept was established by treating hiPSC-CM with equivalent concentrations of free and liposomal doxorubicin. The study was then repeated with free daunorubicin plus cytarabine and CPX-351, a dual-drug liposomal encapsulation of daunorubicin/cytarabine. hiPSC-CM were treated with free-drug or liposomal formulations for 24 h on Days 1, 3, and 5 at equivalent concentrations ranging from 0 to 1000 ng/mL and assessed on subsequent days. Free-drug treatment resulted in concentration-dependent cumulative cytotoxicity (microscopy), more profound decrease in ATP levels, and significant time- and concentration-dependent decreases in oxygen consumption versus liposomal formulations (p < 0.01). Repeated free-drug exposure also resulted in greater release of biomarkers (cardiac troponin I, FABP3) and lactate dehydrogenase, as well as in a biphasic rhythmicity response (initial increase followed by slowing/quiescence of beating) indicating significant injury, which was not observed after repeated exposure to liposomal formulations. Overall, liposomal formulations were considerably less toxic to hiPSC-CM than their free-drug counterparts. Clinical data will be needed to confirm findings for CPX-351.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Cardiotoxicity , Myocytes, Cardiac , Daunorubicin/toxicity , Cytarabine/toxicity , Anthracyclines , Antibiotics, Antineoplastic/toxicity , Topoisomerase II Inhibitors , Drug Combinations , Liposomes
3.
Toxicol Appl Pharmacol ; 470: 116547, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37178933

ABSTRACT

Daunorubicin (DNR-) induced cardiotoxicity seriously restricts its clinical application. Transient receptor potential cation channel subfamily C member 6 (TRPC6) is involved in multiple cardiovascular physiological and pathophysiological processes. However, the role of TRPC6 anthracycline-induced cardiotoxicity (AIC) remains unclear. Mitochondrial fragmentation greatly promotes AIC. TRPC6-mediated ERK1/2 activation has been shown to favor mitochondrial fission in dentate granule cells. The aim of the present study was to elucidate the effects of TRPC6 on daunorubicin- induced cardiotoxicity and identify the mechanisms associated with mitochondrial dynamics. The sparkling results showed that TRPC6 was upregulated in models in vitro and in vivo. TRPC6 knockdown protected cardiomyocytes from DNR-induced cell apoptosis and death. DNR largely facilitated mitochondrial fission, boosted mitochondrial membrane potential collapse and damaged debilitated mitochondrial respiratory function in H9c2 cells,these effects were accompanied by TRPC6 upregulation. siTRPC6 effectively inhibited these mitochondrial adverse aspects showing a positive unexposed effect on mitochondrial morphology and function. Concomitantly, ERK1/2-DRP1 which is related to mitochondrial fission was significantly activated with amplified phosphorylated forms in DNR-treated H9c2 cells. siTRPC6 effectively suppressed ERK1/2-DPR1 over activation, hinting at a potential correlation between TRPC6 and ERK1/2-DRP1 by which mitochondrial dynamics are possibly modulated in AIC. TRPC6 knockdown also raised the Bcl-2/Bax ratio, which may help to block mitochondrial fragmentation-related functional impairment and apoptotic signaling. These findings suggested an essential role of TRPC6 in AIC by intensifying mitochondrial fission and cell death via ERK1/2-DPR1, which could be a potential therapeutic target for AIC.


Subject(s)
Daunorubicin , Myocytes, Cardiac , TRPC6 Cation Channel , Animals , Rats , Apoptosis , Cardiotoxicity/metabolism , Cell Death , Daunorubicin/toxicity , Dynamins/metabolism , MAP Kinase Signaling System , Mitochondrial Dynamics , Myocytes, Cardiac/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , TRPC6 Cation Channel/metabolism
4.
Cardiovasc Toxicol ; 22(8): 701-712, 2022 08.
Article in English | MEDLINE | ID: mdl-35596909

ABSTRACT

Cardiac troponin T (encoded by TNNT2) is involved in the contraction of cardiomyocytes during beating. The alternative splicing of TNNT2 results in four transcript variants with differential Ca2+ sensitivity. The splicing of TNNT2 involves phosphorylation of the splicing factor SRSF6 by DYRK1A. Altered TNNT2 splicing patterns have been identified in failing human hearts. There is a paucity of studies describing DYRK1A-SRSF6-TNNT2 interplays in human cardiomyocytes. Also, it is not known whether the sensitivity of cardiomyocytes to cardiotoxic anthracyclines is modified in the context of variable DYRK1A-TNNT2 expression. In this study, we investigated the impact of DYRK1A on the endogenous expression of TNNT2 splicing variants in iPSC-derived cardiomyocytes. We also examined whether DYRK1A expression modifies the sensitivity of cardiomyocytes to the cardiotoxic drug daunorubicin (DAU). DYRK1A over-expression increased the abundance of TNNT2 fetal variants by ~ 58% whereas the abundance of the adult cTnT3 variant decreased by ~ 27%. High DYRK1A expression increased the phosphorylation of SRSF6 by ~ 25-65%. DAU cytotoxicity was similar between cardiomyocytes with variable levels of DYRK1A expression. DYRK1A over-expression ameliorated the impact of DAU on beating frequency. This study lays the foundation to further investigate the contribution of variable DYRK1A-TNNT2 expression to Ca2+ handling and beating in human cardiomyocytes.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Adult , Cardiotoxicity/metabolism , Daunorubicin/metabolism , Daunorubicin/toxicity , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Serine-Arginine Splicing Factors/metabolism , Troponin T/genetics , Dyrk Kinases
5.
ACS Appl Mater Interfaces ; 13(36): 42382-42395, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34473471

ABSTRACT

Anthracycline-induced liver injury (AILI) is becoming an increasingly serious and potential clinical complication and is linked to reactive oxygen species (ROS) production and subsequent inflammatory response. Herein, we demonstrated that artificial Prussian blue nanozymes (PBZs) prevented daunorubicin-induced liver injury, a prototype of AILI, by attenuating ROS production and regulating inflammation. PBZs exhibited multienzyme activity and could scavenge ROS and free radicals. At the cellular level, PBZs could effectively eliminate ROS, suppress hepatocyte apoptosis, reduce deoxyribonucleic acid damage, and decrease the levels of inflammatory cytokines and chemokines. According to the results of the in vivo study, pretreatment with PBZs also resulted in a desirable protective effect against AILI, as indicated by both a decrease in biochemical indicator levels and hepatocyte necrosis. PBZs upregulated antioxidative genes by activating the Nrf2 pathway to reduce oxidative stress. Meanwhile, PBZs counteracted the inflammatory response based on the decreased expression levels of myeloperoxidase and F4/80 in the liver. Collectively, our findings indicate that PBZ-based nanotherapy is a novel strategy for protecting against AILI.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Chemical and Drug Induced Liver Injury/prevention & control , Daunorubicin/toxicity , Free Radical Scavengers/therapeutic use , Inflammation/drug therapy , Nanoparticles/therapeutic use , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/toxicity , Apoptosis/drug effects , Catalysis , Chemical and Drug Induced Liver Injury/metabolism , Chemokines/metabolism , DNA/drug effects , DNA Damage/drug effects , Ferrocyanides/chemistry , Ferrocyanides/therapeutic use , Ferrocyanides/toxicity , Free Radical Scavengers/chemistry , Free Radical Scavengers/toxicity , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/toxicity , Oxidative Stress/drug effects , Povidone/chemistry , Povidone/toxicity , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
6.
J Med Chem ; 64(7): 3997-4019, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33750129

ABSTRACT

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


Subject(s)
Cardiotonic Agents/therapeutic use , Cardiotoxicity/drug therapy , Piperazines/therapeutic use , Topoisomerase II Inhibitors/therapeutic use , Animals , Animals, Newborn , Cardiotonic Agents/chemical synthesis , Cardiotonic Agents/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Topoisomerases, Type II/metabolism , Daunorubicin/toxicity , Diketopiperazines , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Myocytes, Cardiac/drug effects , Piperazines/chemical synthesis , Piperazines/metabolism , Protein Binding , Rats, Wistar , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/metabolism
7.
Cardiovasc Toxicol ; 21(2): 142-151, 2021 02.
Article in English | MEDLINE | ID: mdl-32880787

ABSTRACT

Cardiotoxicity is a major limitation for anthracycline chemotherapy although anthracyclines are potent antitumor agents. The precise mechanism underlying clinical heart failure due to anthracycline treatment is not fully understood, but is believed to be due, in part, to lipid peroxidation and the generation of free radicals by anthracycline-iron complexes. Thioredoxin (Trx) is a small redox-active antioxidant protein with potent disulfide reductase properties. Here, we present evidence that cancer cells overexpressing Trx undergo enhanced apoptosis in response to daunomycin. In contrast, cells overexpressing redox-inactive mutant Trx were not effectively killed. However, rat embryonic cardiomyocytes (H9c2 cells) overexpressing Trx were protected against daunomycin-mediated apoptosis, but H9c2 cells with decreased levels of active Trx showed enhanced apoptosis in response to daunomycin. We further demonstrate that increased level of Trx is specifically effective in anthracycline toxicity, but not with other topoisomerase II inhibitors such as etoposide. Collectively these data demonstrate that whereas high levels of Trx protect cardiomyocytes against anthracycline toxicity, it potentiates toxicity of anthracyclines in cancer cells.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Apoptosis/drug effects , Daunorubicin/toxicity , Myocytes, Cardiac/drug effects , Neoplasms/drug therapy , Thioredoxins/metabolism , Animals , Cardiotoxicity , HCT116 Cells , Humans , MCF-7 Cells , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Poly(ADP-ribose) Polymerases/metabolism , Rats , Thioredoxins/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , U937 Cells
8.
J Ethnopharmacol ; 261: 113118, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-32621953

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Green tea is the most ancient and popular beverage worldwide and its main constituent epigallocatechin-3-gallate (EGCG) has a potential role in the management of cancer through the modulation of cell signaling pathways. However, EGCG is frangible to oxidation and exhibits low lipid solubility and bioavailability, and we synthesized a derivative of EGCG in an attempt to overcome these limitations. AIM OF THE STUDY: The anthracycline antibiotic daunorubicin (DNR) is a potent anticancer agent. However, its severe cardiotoxic limits its clinical efficacy. Human carbonyl reductase 1 (CBR1) is one of the most effective human reductases for producing hydroxyl metabolites and thus may be involved in increasing the cardiotoxicity and decreasing the antineoplastic effect of anthracycline antibiotics. Accordingly, in this study, we investigated the co-therapeutic effect of Y6, a novel and potent adjuvant obtained by optimization of the structure of EGCG. MATERIAL AND METHODS: The cellular concentrations of DNR and its metabolite DNRol were measured by HPLC to determine the effects of EGCG and Y6 on the inhibition of DNRol formation. The cytotoxic effects of EGCG and Y6 were tested by MTT assay in order to identify non-toxic concentrations of them. To understand their antitumor and cardioprotective mechanisms, hypoxia-inducible factor-1α (HIF-1α) and CBR1 protein expression was measured via Western blotting and immunohistochemical staining while gene expression was analyzed using RT-PCR. Moreover, PI3K/AKT and MEK/ERK signaling pathways were analyzed via Western blotting. HepG2 xenograft model was used to detect the effects of EGCG and Y6 on the antitumor activity and cardiotoxicity of DNR in vivo. Finally, to obtain further insight into the interactions of Y6 and EGCG with HIF-1α and CBR1, we performed a molecular modeling. RESULTS: Y6(10 µg/ml or 55 mg/kg) decreased the expression of HIF-1α and CBR1 at both the mRNA and protein levels during combined drug therapy in vitro as well as in vivo, thereby inhibiting formation of the metabolite DNRol from DNR, with the mechanisms being related to PI3K/AKT and MEK/ERK signaling inhibition. In a human carcinoma xenograft model established with subcutaneous HepG2 cells, Y6(55 mg/kg) enhanced the antitumor effect and reduced the cardiotoxicity of DNR more effectively than EGCG(40 mg/kg). CONCLUSIONS: Y6 has the ability to inhibit CBR1 expression through the coordinate inhibition of PI3K/AKT and MEK/ERK signaling, then synergistically enhances the antitumor effect and reduces the cardiotoxicity of DNR.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Arrhythmias, Cardiac/prevention & control , Carcinoma, Hepatocellular/drug therapy , Catechin/analogs & derivatives , Daunorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Liver Neoplasms/drug therapy , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Antineoplastic Combined Chemotherapy Protocols/toxicity , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cardiotoxicity , Catechin/pharmacology , Cell Proliferation/drug effects , Daunorubicin/toxicity , Drug Synergism , Female , Gene Expression Regulation, Neoplastic , Heart Rate/drug effects , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Clin Sci (Lond) ; 133(16): 1827-1844, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31409729

ABSTRACT

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


Subject(s)
Anthracyclines/toxicity , Antineoplastic Agents/toxicity , Cardiotoxicity/etiology , Proteasome Inhibitors/toxicity , Animals , Anthracyclines/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/toxicity , Bortezomib/administration & dosage , Bortezomib/toxicity , Daunorubicin/administration & dosage , Daunorubicin/toxicity , Dose-Response Relationship, Drug , Male , Myocytes, Cardiac/drug effects , Oligopeptides/administration & dosage , Oligopeptides/toxicity , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/administration & dosage , Rabbits , Rats , Rats, Wistar
10.
Nanomedicine ; 20: 102004, 2019 08.
Article in English | MEDLINE | ID: mdl-31055076

ABSTRACT

Patients with acute myeloid leukemia have a very poor prognosis related to a high rate of relapse and drug-related toxicity. The ability of leukemia stem cells (LSCs) to survive chemotherapy is primarily responsible for relapse, and eliminating LSCs is ultimately essential for cure. We developed novel disulfide-crosslinked CLL1-targeting micelles (DC-CTM), which can deliver high concentrations of daunorubicin (DNR) into both bulk leukemia cells and LSCs. Compared to free DNR, DC-CTM-DNR had a longer half-life, increased DNR area under the curve concentration by 11-fold, and exhibited a superior toxicity profile. In patient-derived AML xenograft models, DC-CTM-DNR treatment led to significant decreases in AML engraftment and impairment of secondary transplantation compared to control groups. Collectively, we demonstrate superior anti-LSC/AML efficacy, and preferable pharmacokinetic and toxicity profiles of DC-CTM-DNR compared to free DNR. DC-CTM-DNR has the potential to significantly improve treatment outcomes and reduce therapy-related morbidity and mortality for patients with AML.


Subject(s)
Daunorubicin/therapeutic use , Lectins, C-Type/chemistry , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Micelles , Nanoparticles/chemistry , Neoplastic Stem Cells/pathology , Animals , Cross-Linking Reagents/chemistry , Daunorubicin/pharmacokinetics , Daunorubicin/toxicity , Disulfides/chemistry , Humans , Mice, Inbred BALB C , Nanoparticles/ultrastructure , Neoplastic Stem Cells/drug effects , Rats, Sprague-Dawley
11.
Toxicology ; 411: 81-92, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30339824

ABSTRACT

In the present study, the molecular docking mechanism based on pharmacodynamic interactions between the ligands AZD1208 and recognized chemotherapy agents (Vincristine and Daunorubicin) with human ATP-binding cassette (ABC) transporters (ABCB1) was investigated. For the first time, were combined an in silico approaches like molecular docking and ab initio computational simulation based on Density Functional Theory (DFT) to explain the drug-drug interaction mechanism of aforementioned chemotherapy ligands with the transmembrane ligand extrusion binding domains (TMDs) of ABCB1. In this regard, the theoretical pharmacodynamic interactions were characterized by using the Gibbs free energy (FEB, kcal/mol) from the best ABCB1-ligand docking complexes. The molecular docking results pointing that for the three chemotherapy ABCB1-ligand complexes are mainly based in non-covalent hydrophobic and hydrogen-bond interactions showing a similar toxicodynamic behavior in terms of strength of interaction (FEB, kcal/mol) and very close free binding energies when compared with the FEB-values of the ABCB1 specific-inhibitor (Rhodamine B) = -6.0 kcal/mol used as theoretical docking control to compare with FEB (AZD1208-ABCB1) ∼ FEB (Vincristine-ABCB1) ∼ FEB (Daunorubicin-ABCB1) -6.2 kcal/mol as average. Ramachandran plot suggests that the 3D-crystallographic structure from ABCB1 transporter can be efficiently-modeled with conformationally-favored Psi versus Phi dihedral angles for all key TMDs-residues. Though, the results of DFT-simulation corroborate the existence of drug-drug interaction between (AZD1208/Vincristine) > (AZD1208/Daunorubicin). These theoretical pieces of evidence have preclinical relevance potential in the design of the new drugs to understand the polypharmacology influence in the molecular mechanism of multiple-drugs resistance, contributing with a higher success in chemotherapy and prognosis of cancer patients.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Antineoplastic Agents, Phytogenic/toxicity , Biphenyl Compounds/toxicity , Daunorubicin/toxicity , Protein Kinase Inhibitors/toxicity , Protein Transport/drug effects , Thiazolidines/toxicity , Vincristine/toxicity , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/drug effects , Binding Sites/drug effects , Cell Line, Tumor , Density Functional Theory , Drug Interactions , Humans , Ligands , Molecular Conformation , Molecular Docking Simulation
12.
Toxicology ; 392: 1-10, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28941780

ABSTRACT

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


Subject(s)
Cardiotonic Agents/pharmacology , DNA Topoisomerases, Type II/metabolism , Dexrazoxane/pharmacology , Iron Chelating Agents/pharmacology , Animals , Animals, Newborn , Anthracyclines/toxicity , Cardiotoxicity/drug therapy , Cardiotoxicity/etiology , Cell Proliferation/drug effects , Cells, Cultured , Daunorubicin/toxicity , Dexrazoxane/analogs & derivatives , Diketopiperazines/pharmacology , Iron/metabolism , Myocytes, Cardiac/drug effects , Rats , Rats, Wistar , Structure-Activity Relationship
13.
PLoS One ; 12(5): e0177660, 2017.
Article in English | MEDLINE | ID: mdl-28498861

ABSTRACT

Anthracyclines are chemotherapeutic drugs known to induce heart failure in a dose-dependent manner. Mechanisms involved in anthracycline cardiotoxicity are an area of relevant investigation. Caveolins bind, organize and regulate receptors and signaling molecules within cell membranes. Caveolin-3 (Cav-3), integrins and related membrane repair proteins can function as cardioprotective proteins. Expression of these proteins in anthracycline-induced heart failure has not been evaluated. We tested the hypothesis that daunorubicin alters cardioprotective protein expression in the heart. Rabbits were administered daunorubicin (3 mg/kg, IV) weekly, for three weeks or nine weeks. Nine weeks but not three weeks of daunorubicin resulted in progressive reduced left ventricular function. Cav-3 expression in the heart was unchanged at three weeks of daunorubicin and increased in nine week treated rabbits when compared to control hearts. Electron microscopy showed caveolae in the heart were increased and mitochondrial number and size were decreased after nine weeks of daunorubicin. Activated beta-1 (ß1) integrin and the membrane repair protein MG53 were increased after nine weeks of daunorubicin vs. controls with no change at the three week time point. The results suggest a potential pathophysiological role for Cav3, integrins and membrane repair in daunorubicin-induced heart failure.


Subject(s)
Anthracyclines/toxicity , Caveolins/metabolism , Daunorubicin/toxicity , Heart Failure/metabolism , Integrins/metabolism , Animals , Blotting, Western , Cardiotoxicity/blood , Cardiotoxicity/metabolism , Cholesterol/blood , Cholesterol/metabolism , Echocardiography , Heart Failure/chemically induced , Immunohistochemistry , Microscopy, Electron , Myocardium/metabolism , Myocardium/pathology , Rabbits
14.
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
15.
PLoS One ; 11(10): e0163885, 2016.
Article in English | MEDLINE | ID: mdl-27741251

ABSTRACT

A regulatory program involving hundreds of genes is coordinated by p53 to prevent carcinogenesis in response to stress. Given the importance of chromatin loops in gene regulation, we investigated whether DNA interactions participate in the p53 stress response. To shed light on this issue, we measured the binding dynamics of cohesin in response to stress. We reveal that cohesin is remodeled at specific loci during the stress response and that its binding within genes negatively correlates with transcription. At p53 target genes, stress-induced eviction of cohesin from gene bodies is concomitant to spatial reorganization of loci through the disruption of functional chromatin loops. These findings demonstrate that chromatin loops can be remodeled upon stress and contribute to the p53-driven stress response. Additionally, we also propose a mechanism whereby transcription-coupled eviction of cohesin from CDKN1A might act as a molecular switch to control spatial interactions between regulatory elements.


Subject(s)
Chromatin Assembly and Disassembly , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly/drug effects , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Binding Proteins , Daunorubicin/toxicity , Genes, Reporter , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , HCT116 Cells , Humans , Nuclear Proteins/metabolism , Oxidative Stress/drug effects , Phosphoproteins/metabolism , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Sequence Analysis, RNA , Transcriptional Activation/drug effects , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Cohesins
16.
Invest New Drugs ; 34(4): 416-23, 2016 08.
Article in English | MEDLINE | ID: mdl-27146514

ABSTRACT

Compared to classical chemotherapy, peptide-based drug targeting is a promising therapeutic approach for cancer, which can provide increased selectivity and decreased side effects to anticancer drugs. Among various homing devices, gonadotropin-releasing hormone-III (GnRH-III) peptide represents a suitable targeting moiety, in particular in the treatment of hormone independent tumors that highly express GnRH receptors (e.g. colon carcinoma). We have previously shown that GnRH-III[(4)Lys(Ac),(8)Lys(Dau = Aoa)] bioconjugate, in which daunorubicin was attached via oxime linkage to the (8)Lys of a GnRH-III derivative, exerted significant in vivo antitumor effect on subcutaneously developed HT-29 colon tumor. In contrast, results of the study reported here indicated that this compound was not active on an orthotopically developed tumor. However, if Lys in position 4 was acylated with butyric acid instead of acetic acid, the resulting bioconjugate GnRH-III[(4)Lys(Bu),(8)Lys(Dau = Aoa)] had significant tumor growth inhibitory effect. Furthermore, it prevented tumor neovascularization, without detectable side effects. Nevertheless, the development of metastases could not be inhibited by the bioconjugate; therefore, its application in combination with a metastasis preventive agent might be necessary in order to achieve complete tumor remission. In spite of this result, the treatment with GnRH-III[(4)Lys(Bu),(8)Lys(Dau = Aoa)] bioconjugate proved to have significant benefits over the administration of free daunorubicin, which was used at the maximum tolerated dose.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Butyric Acid/therapeutic use , Colorectal Neoplasms/drug therapy , Daunorubicin/therapeutic use , Gonadotropin-Releasing Hormone/therapeutic use , Pyrrolidonecarboxylic Acid/analogs & derivatives , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/toxicity , Butyric Acid/chemistry , Butyric Acid/toxicity , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Daunorubicin/chemistry , Daunorubicin/toxicity , Female , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/toxicity , HT29 Cells , Heart/drug effects , Humans , Liver/drug effects , Male , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Pyrrolidonecarboxylic Acid/chemistry , Pyrrolidonecarboxylic Acid/therapeutic use , Pyrrolidonecarboxylic Acid/toxicity , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
17.
Mater Sci Eng C Mater Biol Appl ; 61: 753-61, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26838906

ABSTRACT

Clinical use of daunorubicin (Dau) in treatment of leukemia has been restricted because of its cardiotoxicity. Targeted delivery of anticancer drugs could decrease their off-target effects and enhance their efficacy. In this study a modified polyvalent aptamers (PA)-Daunorubicin (Dau)-Gold nanoparticles (AuNPs) complex was designed and its efficacy was assessed in Molt-4 cells (human acute lymphoblastic leukemia T-cell, target). Dau was efficiently loaded (10.5 µM) onto 1mL of PA-modified AuNPs. Dau was released from the PA-Dau-AuNPs complex in a pH-sensitive manner (faster release at pH5.5). The results of flow cytometry analysis indicated that the PA-Dau-AuNPs complex was efficiently internalized into target cells, but not into nontarget cells. The results of MTT assay were consistent with the internalization data. PA-Dau-AuNPs complex had less cytotoxicity in U266 cells compared to Dau alone and even Apt-Dau-AuNPs complex. The PA-Dau-AuNPs complex had more cytotoxicity in Molt-4 cells compared to Dau alone and even Apt-Dau-AuNPs complex. Cytotoxicity of PA-Dau-AuNPs complex was effectively antagonized using antisense of polyvalent aptamers. In conclusion, the designed drug delivery system inherited the properties of efficient drug loading, tumor targeting, pH-dependent drug release and controllable delivery of Dau to tumor cells.


Subject(s)
Antineoplastic Agents/chemistry , Aptamers, Nucleotide/chemistry , Daunorubicin/chemistry , Drug Carriers/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Antineoplastic Agents/toxicity , Cell Line, Tumor , Cell Survival/drug effects , Daunorubicin/toxicity , Humans , Hydrogen-Ion Concentration , Leukemia/metabolism , Leukemia/pathology , Oligonucleotides, Antisense/metabolism
18.
Invest New Drugs ; 33(5): 1032-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26268925

ABSTRACT

In this study, we compared the cellular uptake, intracellular localization and cytotoxicity of two groups of anthracycline derivatives in cultured H9c2(2-1) rat cardiomyoblasts. The first group consisted of doxorubicin (DOX) and two of its derivatives containing a formamidino group (-N = CH-N<) at the C-3' position with a morpholine (DOXM) or a hexamethyleneimine (DOXH) ring. The second group consisted of daunorubicin (DRB) and its derivatives containing a morpholine (DRBM) or a hexamethyleneimine (DRBH) ring. DOXH and DRBH were taken up by cardiomyoblasts more efficiently than estimated for other tested anthracyclines. The cellular uptakes of DOXM and DRBM were reduced compared to those of the parent compounds. Applied structural modifications of DOX and DRB influenced the subcellular localization of the tested derivatives. DOX and DOXH were localized primarily in nuclei, whereas the other anthracyclines were found in the nuclei and cytoplasm. The percentages of the compounds that accumulated in the nuclei were 80.2 and 54.2 % for DOX and DOXH, respectively. The lowest nuclear accumulation values were observed for DRBM (19.9 %), DRBH (21.9 %) and DOXM (23.7 %). The ability of anthracyclines to accumulate in the nuclei correlated with their DNA binding constants (r = 0.858, P = 0.029). A correlation was found between the accumulation of the tested anthracyclines in the nuclei of cardiomyoblasts and their cardiotoxicity in vivo, which was observed in our previous study. We suggest that cytotoxicity and the anthracycline accumulation level in the nuclei of cultured cardiomyoblasts could be used for early prediction of their cardiotoxicity.


Subject(s)
Anthracyclines/chemistry , Anthracyclines/toxicity , Cardiotoxicity/prevention & control , Animals , Anthracyclines/metabolism , Cell Cycle , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Daunorubicin/chemistry , Daunorubicin/toxicity , Doxorubicin/chemistry , Doxorubicin/toxicity , Myoblasts, Cardiac , Rats , Structure-Activity Relationship
19.
Int J Cardiol ; 201: 358-67, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26310978

ABSTRACT

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


Subject(s)
Anthracyclines/toxicity , Cardiotoxicity/blood , Troponin I/blood , Troponin T/blood , Animals , Antibiotics, Antineoplastic/toxicity , Biomarkers/blood , Biomarkers/metabolism , Cardiomyopathies/blood , Cardiomyopathies/chemically induced , Cardiotoxicity/diagnostic imaging , Daunorubicin/toxicity , Disease Models, Animal , Echocardiography , Heart/drug effects , Heart/physiology , Male , Predictive Value of Tests , Rabbits , Regression Analysis , Systole/drug effects , Systole/physiology
20.
Int Immunopharmacol ; 28(1): 154-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26072060

ABSTRACT

Inflammation and oxidative stress play important roles in the progression of renal damage. The natural polyphenol naringenin is known to exert potent antioxidant and anti-inflammatory effects. In this study, we have investigated the effect of naringenin on kidney dysfunction, fibrosis, endoplasmic reticulum (ER) stress, angiotensin II type I receptor (AT1R) expression and inflammation in daunorubicin (DNR) induced nephrotoxicity model. Nephrotoxicity was induced in rats by intravenous injection of DNR at a cumulative dose of 9 mg/kg. After 1 week, naringenin (20mg/kg/day. p.o) was administered daily for 6 weeks. Biochemical studies were performed to evaluate renal function. Western blotting was performed to measure the protein levels of AT1R, endothelin (ET)1, ET receptor type A (ETAR), extracellular signal-regulated kinase (ERK)1/2, nuclear factor (NF)κB p65, peroxisome proliferator activated receptor (PPAR)γ, oxidative/ER stress, apoptosis, and inflammatory markers in the kidney of DNR treated rats. Histopathological analysis was done using hemotoxylin eosin and Masson trichrome stained renal sections to investigate the structural abnormalities and fibrosis. DNR treated rats suffered from nephrotoxicity as evidenced by worsened renal function, increased blood urea nitrogen, serum creatinine levels in renal tissues and histopathogical abnormalities. Treatment with naringenin mitigated these changes. Furthermore, naringenin up regulated PPARγ and down regulated AT1R, ET1, ETAR, p-ERK1/2, p-NFκB p65, ER stress, apoptosis, and inflammatory markers. Our results suggest that naringenin has an ability to improve renal function and attenuates AT1R, ERK1/2-NFκB p65 signaling pathway in DNR induced nephrotoxicity in rats.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibiotics, Antineoplastic/toxicity , Daunorubicin/antagonists & inhibitors , Daunorubicin/toxicity , Flavanones/pharmacology , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , MAP Kinase Signaling System/drug effects , Receptor, Angiotensin, Type 1/drug effects , Renal Agents/pharmacology , Transcription Factor RelA/drug effects , Animals , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Fibrosis , Inflammation/drug therapy , Inflammation/physiopathology , Injections, Intravenous , Kidney Diseases/pathology , Male , PPAR gamma/biosynthesis , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL