Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
1.
J Biol Chem ; 299(6): 104800, 2023 06.
Article in English | MEDLINE | ID: mdl-37164156

ABSTRACT

For cells, it is important to repair DNA damage, such as double-strand and single-strand DNA breaks, because unrepaired DNA can compromise genetic integrity, potentially leading to cell death or cancer. Cells have multiple DNA damage repair pathways that have been the subject of detailed genetic, biochemical, and structural studies. Recently, the scientific community has started to gain evidence that the repair of DNA double-strand breaks may occur within biomolecular condensates and that condensates may also contribute to DNA damage through concentrating genotoxic agents used to treat various cancers. Here, we summarize key features of biomolecular condensates and note where they have been implicated in the repair of DNA double-strand breaks. We also describe evidence suggesting that condensates may be involved in the repair of other types of DNA damage, including single-strand DNA breaks, nucleotide modifications (e.g., mismatch and oxidized bases), and bulky lesions, among others. Finally, we discuss old and new mysteries that could now be addressed considering the properties of condensates, including chemoresistance mechanisms.


Subject(s)
DNA Repair , DNA , Drug Resistance, Neoplasm , DNA/chemistry , DNA/drug effects , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , Drug Resistance, Neoplasm/drug effects , DNA Breaks, Single-Stranded/drug effects , Base Pair Mismatch/drug effects
2.
ACS Appl Mater Interfaces ; 13(39): 46303-46316, 2021 Oct 06.
Article in English | MEDLINE | ID: mdl-34569240

ABSTRACT

The antibacterial and cell-proliferative character of atmospheric pressure plasma jets (APPJs) helps in the healing process of chronic wounds. However, control of the plasma-biological target interface remains an open issue. High vacuum ultraviolet/ultraviolet (VUV/UV) radiation and RONS flux from plasma may cause damage of a treated tissue; therefore, controlled interaction is essential. VUV/UV emission from argon APPJs and radiation control with aerosol injection in plasma effluent is the focus of this research. The aerosol effect on radiation is studied by a fluorescent target capable of resolving the plasma oxidation footprint. In addition, DNA damage is evaluated by plasmid DNA radiation assay and cell proliferation assay to assess safety aspects of the plasma jet, the effect of VUV/UV radiation, and its control with aerosol injection. Inevitable emission of VUV/UV radiation from plasmas during treatment is demonstrated in this work. Plasma has no antiproliferative effect on fibroblasts in short treatments (t < 60 s), while long exposure has a cytotoxic effect, resulting in decreased cell survival. Radiation has no effect on cell survival in the medium due to absorption. However, a strong cytotoxic effect on the attached fibroblasts without the medium is apparent. VUV/UV radiation contributes 70% of the integral plasma effect in induction of single- and double-strand DNA breaks and cytotoxicity of the attached cells without the medium. Survival of the attached cells increases by 10% when aerosol is introduced between plasma and the cells. Injection of aerosol in the plasma effluent can help to control the plasma-cell/tissue interaction. Aerosol droplets in the effluent partially absorb UV emission from the plasma, limiting photon flux in the direction of the biological target. Herein, cold and safe plasma-aerosol treatment and a safe operational mode of treatment are demonstrated in a murine model.


Subject(s)
Aerosols/toxicity , Argon/toxicity , Plasma Gases/toxicity , Aerosols/radiation effects , Animals , Argon/radiation effects , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , DNA/drug effects , DNA/radiation effects , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Breaks, Single-Stranded/drug effects , DNA Breaks, Single-Stranded/radiation effects , Female , Mice, Inbred BALB C , Plasma Gases/radiation effects , Plasmids/drug effects , Plasmids/radiation effects , Skin/drug effects , Skin/radiation effects , Ultraviolet Rays
3.
Mutagenesis ; 35(5): 391-404, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32914844

ABSTRACT

DNA ligase I (LIG1) joins DNA strand breaks during DNA replication and repair transactions and contributes to genome integrity. The mutations (P529L, E566K, R641L and R771W) in LIG1 gene are described in patients with LIG1-deficiency syndrome that exhibit immunodeficiency. LIG1 senses 3'-DNA ends with a mismatch or oxidative DNA base inserted by a repair DNA polymerase. However, the ligation efficiency of the LIG1 variants for DNA polymerase-promoted mutagenesis products with 3'-DNA mismatches or 8-oxo-2'-deoxyguanosine (8-oxodG) remains undefined. Here, we report that R641L and R771W fail in the ligation of nicked DNA with 3'-8-oxodG, leading to an accumulation of 5'-AMP-DNA intermediates in vitro. Moreover, we found that the presence of all possible 12 non-canonical base pairs variously impacts the ligation efficiency by P529L and R771W depending on the architecture at the DNA end, whereas E566K exhibits no activity against all substrates tested. Our results contribute to the understanding of the substrate specificity and mismatch discrimination of LIG1 for mutagenic repair intermediates and the effect of non-synonymous mutations on ligase fidelity.


Subject(s)
DNA Ligase ATP/genetics , DNA Mismatch Repair/genetics , Mutagenesis/genetics , 8-Hydroxy-2'-Deoxyguanosine/genetics , Adenosine Monophosphate/genetics , DNA Breaks, Single-Stranded/drug effects , DNA Damage/genetics , DNA Replication/genetics , Genome/drug effects , Humans , Mutation/genetics , Oxidative Stress/drug effects
4.
Int J Mol Sci ; 21(14)2020 Jul 11.
Article in English | MEDLINE | ID: mdl-32664581

ABSTRACT

Glioblastoma multiforme (GBM) is a severe brain tumor whose ability to mutate and adapt to therapies is at the base for the extremely poor survival rate of patients. Despite multiple efforts to develop alternative forms of treatment, advances have been disappointing and GBM remains an arduous tumor to treat. One of the leading causes for its strong resistance is the innate upregulation of DNA repair mechanisms. Since standard therapy consists of a combinatory use of ionizing radiation and alkylating drugs, which both damage DNA, targeting the DNA damage response (DDR) is proving to be a beneficial strategy to sensitize tumor cells to treatment. In this review, we will discuss how recent progress in the availability of the DDR kinase inhibitors will be key for future therapy development. Further, we will examine the principal existing DDR inhibitors, with special focus on those currently in use for GBM clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , DNA Repair , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Molecular Targeted Therapy , Antineoplastic Agents/therapeutic use , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , Brain Neoplasms/genetics , Cell Line, Tumor , Clinical Trials, Phase I as Topic , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Single-Stranded/drug effects , DNA Damage , DNA, Neoplasm/drug effects , DNA, Neoplasm/genetics , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , Drug Screening Assays, Antitumor , Enzyme Inhibitors/therapeutic use , Glioblastoma/genetics , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Nitrosourea Compounds/pharmacology , Nitrosourea Compounds/therapeutic use , Signal Transduction/drug effects , Temozolomide/pharmacology , Temozolomide/therapeutic use
6.
PLoS One ; 15(5): e0232724, 2020.
Article in English | MEDLINE | ID: mdl-32374749

ABSTRACT

DNA damage in the A549 human lung cancer cell line treated with cold plasma irradiation was investigated. We confirmed that cold atmospheric plasma generated reactive oxygen and nitrogen species (RONS) in a liquid, and the intracellular RONS level was increased in plasma-irradiated cells. However, a notable decrease in cell viability was not observed 24 hours after plasma irradiation. Because RONS induce oxidative damage in cells, strand breaks and chemical modification of DNA in the cancer cells were investigated. We found that 8-oxoguanine (8-oxoG) formation as well as DNA strand breaks, which have been thoroughly investigated, were induced by plasma irradiation. In addition, up-regulation of 8-oxoG repair enzyme was observed after plasma irradiation.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Single-Stranded/drug effects , DNA Glycosylases/metabolism , DNA Repair , Guanine/analogs & derivatives , Plasma Gases/pharmacology , A549 Cells , Cell Survival/drug effects , DNA/metabolism , Guanine/biosynthesis , Humans , Oxidative Stress/drug effects , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Up-Regulation
7.
J Pharm Biomed Anal ; 187: 113326, 2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32413832

ABSTRACT

Anti-inflammatory drugs are reported to induce changes in nucleic-acids upon UV-irradiation. Such changes have the potential to cause apoptosis, carcinogenesis, and mutagenesis. In this work, the kinetics of the damage induced in DNA by some anti-inflammatory drugs were compared after UV-irradiation. Five commonly used anti-inflammatory drugs; diclofenac, ketoprofen, leflunomide, piroxicam and tolmetin, were studied. Simple, sensitive and eco-friendly methods for the analysis of DNA-damage were proposed including absorption spectroscopy, MALDI-TOF mass spectrometry and fluorescence using TbCl3. Results show that all drugs induced DNA-damage after UV-irradiation. Absorption spectroscopy results demonstrated hyperchromic shift in the absorption band characteristic to DNA, indicating distortion of the double-strand. Mass spectra showed a significant decrease of the molecular-ion-peak of DNA, together with peaks of smaller m/z that indicated the formation of DNA strand-breaks. TbCl3 fluorescence was observed to increase with incubation time of each drug with DNA, indicating the presence of more single-stranded regions in DNA due to damage. TbCl3 fluorescence was used to obtain the kinetics of the induced damage. Results show that DNA-damage occurred via photoinduced oxidative mechanism. Also, the potency of the studied drugs was examined on calf-thymus real DNA samples using TbCl3 fluorescence with ketoprofen and leflunomide being the most photogenotoxic anti-inflammatory drugs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , DNA Damage/drug effects , DNA/drug effects , Animals , Cattle , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Single-Stranded/drug effects , Spectrometry, Fluorescence/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Ultraviolet Rays/adverse effects
8.
ACS Nano ; 14(3): 2827-2846, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32049495

ABSTRACT

Therapy for intracerebral hemorrhage (ICH) remains elusive, in part dependent on the severity of the hemorrhage itself as well as multiple deleterious effects of blood and its breakdown products such as hemin and free iron. While oxidative injury and genomic damage have been seen following ICH, the details of this injury and implications remain unclear. Here, we discovered that, while free iron produced mostly reactive oxygen species (ROS)-related single-strand DNA breaks, hemin unexpectedly induced rapid and persistent nuclear and mitochondrial double-strand breaks (DSBs) in neuronal and endothelial cell genomes and in mouse brains following experimental ICH comparable to that seen with γ radiation and DNA-complexing chemotherapies. Potentially as a result of persistent DSBs and the DNA damage response, hemin also resulted in senescence phenotype in cultured neurons and endothelial cells. Subsequent resistance to ferroptosis reported in other senescent cell types was also observed here in neurons. While antioxidant therapy prevented senescence, cells became sensitized to ferroptosis. To address both senescence and resistance to ferroptosis, we synthesized a modified, catalytic, and rapidly internalized carbon nanomaterial, poly(ethylene glycol)-conjugated hydrophilic carbon clusters (PEG-HCC) by covalently bonding the iron chelator, deferoxamine (DEF). This multifunctional nanoparticle, DEF-HCC-PEG, protected cells from both senescence and ferroptosis and restored nuclear and mitochondrial genome integrity in vitro and in vivo. We thus describe a potential molecular mechanism of hemin/iron-induced toxicity in ICH that involves a rapid induction of DSBs, senescence, and the consequent resistance to ferroptosis and provide a mechanistic-based combinatorial therapeutic strategy.


Subject(s)
Carbon/pharmacology , Cerebral Hemorrhage/drug therapy , Nanoparticles/chemistry , Animals , Cell Differentiation/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/metabolism , DNA Breaks, Single-Stranded/drug effects , DNA Damage , Deferoxamine/pharmacology , Hemin/antagonists & inhibitors , Hemin/pharmacology , Humans , Iron/pharmacology , Mice , Mitochondria/drug effects , Polyethylene Glycols/pharmacology , Reactive Oxygen Species/metabolism
9.
Biomed Pharmacother ; 125: 109875, 2020 May.
Article in English | MEDLINE | ID: mdl-32036211

ABSTRACT

Efficient DNA repair is critical for cell survival following exposure to DNA topoisomerase I (Top1) inhibitors camptothecin, a nature product from which the common chemotherapeutic drugs irinotecan and topotecan are derived. The camptothecin-derived agents exert their antitumor activities by specifically stabilizing the Top1-DNA covalent complexes (Top1cc) and blocking the DNA religation step. When exposed to these DNA damage agents, tumor cells quickly activate DNA damage response. This allows sufficient time to remove the Top1ccs and prevent tumor cells from apoptosis. Several repair pathways have been implicated in this process. One of the most relevant repair modes is DNA single strand break repair (SSBR) pathway. The expression level or mutagenesis of specific repair factors involved in SSBR pathway may play an indispensable role in individual's capacity of repairing camptothecin induced DNA damage. Therefore, understanding of the tolerance pathways counteracted to camptothecin cytotoxicity is crucial in alleviating chemotherapy resistance. This review focus on the SSBR pathway in repair camptothecin induced DNA damage, aiming to provide insights into the potential molecular determinants of camptothecin chemosensitivity.


Subject(s)
Antineoplastic Agents, Phytogenic/adverse effects , Camptothecin/adverse effects , DNA Breaks, Single-Stranded/drug effects , DNA Damage/drug effects , DNA Repair , Signal Transduction , Topoisomerase I Inhibitors/adverse effects , Animals , Camptothecin/pharmacology , Cell Survival/drug effects , DNA Topoisomerases, Type I/metabolism , Drug Resistance, Neoplasm , Genomic Instability , Humans , Protein Binding
10.
Biochemistry ; 59(7): 892-900, 2020 02 25.
Article in English | MEDLINE | ID: mdl-31977191

ABSTRACT

Colibactin is a genotoxic gut microbiome metabolite long suspected of playing an etiological role in colorectal cancer. Evidence suggests that colibactin forms DNA interstrand cross-links (ICLs) in eukaryotic cells and activates ICL repair pathways, leading to the production of ICL-dependent DNA double-strand breaks (DSBs). Here we show that colibactin ICLs can evolve directly to DNA DSBs. Using the topology of supercoiled plasmid DNA as a proxy for alkylation adduct stability, we find that colibactin-derived ICLs are unstable toward depurination and elimination of the 3' phosphate. This ICL degradation pathway leads progressively to single strand breaks (SSBs) and subsequently DSBs. The spontaneous conversion of ICLs to DSBs is consistent with the finding that nonhomologous end joining repair-deficient cells are sensitized to colibactin-producing bacteria. The results herein refine our understanding of colibactin-derived DNA damage and underscore the complexities underlying the DSB phenotype.


Subject(s)
Cross-Linking Reagents/pharmacology , DNA/metabolism , Peptides/pharmacology , Polyketides/pharmacology , Cross-Linking Reagents/chemistry , DNA/chemistry , DNA/genetics , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Single-Stranded/drug effects , DNA Repair , Deoxyribonuclease IV (Phage T4-Induced)/chemistry , Escherichia coli/chemistry , Peptides/chemistry , Plasmids/chemistry , Polyketides/chemistry
11.
Nucleic Acids Res ; 48(4): 2013-2025, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31930301

ABSTRACT

The rolling-circle replication is the most common mechanism for the replication of small plasmids carrying antibiotic resistance genes in Gram-positive bacteria. It is initiated by the binding and nicking of double-stranded origin of replication by a replication initiator protein (Rep). Duplex unwinding is then performed by the PcrA helicase, whose processivity is critically promoted by its interaction with Rep. How Rep and PcrA proteins interact to nick and unwind the duplex is not fully understood. Here, we have used magnetic tweezers to monitor PcrA helicase unwinding and its relationship with the nicking activity of Staphylococcus aureus plasmid pT181 initiator RepC. Our results indicate that PcrA is a highly processive helicase prone to stochastic pausing, resulting in average translocation rates of 30 bp s-1, while a typical velocity of 50 bp s-1 is found in the absence of pausing. Single-strand DNA binding protein did not affect PcrA translocation velocity but slightly increased its processivity. Analysis of the degree of DNA supercoiling required for RepC nicking, and the time between RepC nicking and DNA unwinding, suggests that RepC and PcrA form a protein complex on the DNA binding site before nicking. A comprehensive model that rationalizes these findings is presented.


Subject(s)
Bacterial Proteins/genetics , DNA Helicases/genetics , DNA Replication/genetics , Drug Resistance, Bacterial/genetics , DNA Breaks, Single-Stranded/drug effects , DNA-Binding Proteins/genetics , Geobacillus stearothermophilus/drug effects , Geobacillus stearothermophilus/genetics , Geobacillus stearothermophilus/pathogenicity , Plasmids/drug effects , Plasmids/genetics , Protein Binding/genetics , Protein Interaction Domains and Motifs/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Tetracycline/pharmacology , Trans-Activators/genetics
12.
Drug Chem Toxicol ; 43(2): 174-181, 2020 Mar.
Article in English | MEDLINE | ID: mdl-30052082

ABSTRACT

Non-steroidal anti-inflammatory drugs are drugs with analgesic, antipyretic, and anti-inflammatory effects. This study uses in vitro methods to investigate the potential and unknown genotoxic effects of dexketoprofen trometamol, an active substance in painkillers, on healthy human lymphocytes. In this study, a cytokinesis-block micronucleus cytome assay is used to investigate potential clastogenic, aneugenic activity and to identify chromosome breakages caused by the active drug substance dexketoprofen trometamol; a comet assay is performed to identify the genotoxic damage resulting from DNA single-strand breaks; a real-time reverse transcription polymerase chain reaction panel system is used to evaluate the potential negative effects on the expression of the genes responsible for DNA damage assessment. Dexketoprofen trometamol induces toxic effects in healthy human lymphocytes at concentrations of 750-1000 µg/mL and above, and shows clastogenic, aneugenic activity by inducing micronucleus formations at exposures of 750-500 µg/mL. At concentration intervals of 1000, 500, 250, 100 µg/mL, dexketoprofen trometamol also resulted in DNA damage in the form of strand breaks, as demonstrated by highly significant increases in DNA tail length and density comet parameters when compared to spontaneous values. Human lymphocytes exposed to 750-100 µg/mL dexketoprofen trometamol were found to have significantly increased levels of expression of the XPC, XRCC6, PNKP genes in the DNA damage signaling pathway. It can be concluded that dexketoprofen trometamol may have cytotoxic, cytostatic, genotoxic effects on healthy human lymphocytes in vitro, depending on the concentration and duration of exposure. It is anticipated that this outcome will be supported by advanced studies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , DNA Damage/drug effects , Ketoprofen/analogs & derivatives , Lymphocytes/drug effects , Tromethamine/toxicity , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Comet Assay , DNA Breaks, Single-Stranded/drug effects , Dose-Response Relationship, Drug , Humans , Ketoprofen/administration & dosage , Ketoprofen/toxicity , Lymphocytes/pathology , Micronucleus Tests , Mutagens/administration & dosage , Mutagens/toxicity , Time Factors , Tromethamine/administration & dosage
13.
Nat Commun ; 10(1): 4846, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31649282

ABSTRACT

DNA topoisomerases are required to resolve DNA topological stress. Despite this essential role, abortive topoisomerase activity generates aberrant protein-linked DNA breaks, jeopardising genome stability. Here, to understand the genomic distribution and mechanisms underpinning topoisomerase-induced DNA breaks, we map Top2 DNA cleavage with strand-specific nucleotide resolution across the S. cerevisiae and human genomes-and use the meiotic Spo11 protein to validate the broad applicability of this method to explore the role of diverse topoisomerase family members. Our data characterises Mre11-dependent repair in yeast and defines two strikingly different fractions of Top2 activity in humans: tightly localised CTCF-proximal, and broadly distributed transcription-proximal, the latter correlated with gene length and expression. Moreover, single nucleotide accuracy reveals the influence primary DNA sequence has upon Top2 cleavage-distinguishing sites likely to form canonical DNA double-strand breaks (DSBs) from those predisposed to form strand-biased DNA single-strand breaks (SSBs) induced by etoposide (VP16) in vivo.


Subject(s)
DNA Repair , DNA Topoisomerases, Type II/metabolism , DNA/metabolism , Endodeoxyribonucleases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Antineoplastic Agents, Phytogenic/pharmacology , Base Sequence , CCCTC-Binding Factor/genetics , DNA/drug effects , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Single-Stranded/drug effects , Etoposide/pharmacology , Humans , Nucleotide Mapping
14.
Mol Carcinog ; 58(12): 2327-2339, 2019 12.
Article in English | MEDLINE | ID: mdl-31544312

ABSTRACT

Autologous stem cell transplant (ASCT) with high-dose melphalan (HDM) is the standard treatment for fit multiple myeloma (MM) patients. It is generally believed that some DNA repair proteins impact the activity to repair melphalan-induced DNA damage, thus potentially contributing to the patient's clinical response. However, knowledge of these proteins is limited. In the current study, we investigated the roles of XRCC1, a protein involved in base excision repair and single-strand break repair, in melphalan response in MM cells. Small interfering RNA knockdown of XRCC1 significantly increased the accumulation of melphalan-induced DNA damage in MM cells and sensitized them to melphalan treatment, indicating that genetic variation in XRCC1 may impact response to melphalan treatment. We then evaluated the association between an XRCC1 variant with reduced activity, rs25487 (R399Q), and clinical outcomes of 108 MM patients with melphalan therapy. Our results showed that XRCC1 rs25487 was associated with prolonged progression-free survival (PFS) in MM patients. The adjusted hazard ratio for PFS between patients carrying rs25487 AA/AG and GG was 0.42 (95% confidence interval: 0.25, 0.84, P = .014). Taken together, these results indicate that XRCC1 is involved in the repair of melphalan-induced DNA damage and XRCC1 rs25487 variant with impaired DNA repair function influences the clinical responses of HDM in MM patients.


Subject(s)
DNA Repair , Hematopoietic Stem Cell Transplantation/methods , Melphalan/therapeutic use , Multiple Myeloma/therapy , X-ray Repair Cross Complementing Protein 1/metabolism , Aged , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/therapeutic use , DNA Breaks, Single-Stranded/drug effects , DNA Damage , Dose-Response Relationship, Drug , Female , Humans , Kaplan-Meier Estimate , Male , Melphalan/adverse effects , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Polymorphism, Single Nucleotide , Progression-Free Survival , RNA Interference , Transplantation, Autologous , X-ray Repair Cross Complementing Protein 1/genetics
15.
J Biochem Mol Toxicol ; 33(11): e22396, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31557364

ABSTRACT

The furocoumarin backbone is a promising platform for chemical modifications aimed at creating new pharmaceutical agents. However, the high level of biological activity of furocoumarins is associated with a number of negative effects. For example, some of the naturally occurring ones and their derivatives can show genotoxic and mutagenic properties as a result of their forming crosslinks with DNA molecules. Therefore, a particularly important area for the chemical modification of natural furocoumarins is to reduce the negative aspects of their bioactivity. By studying a group of 21 compounds-1,2,3-triazolyl modified derivatives of furocoumarin and peucedanin-using the SOS chromotest, the Ames test, and DNA-comet assays, we revealed modifications that can neutralize the structure's genotoxic properties. Theoretical aspects of the interaction of the compound library were studied using molecular modeling and this identified the leading role of the polyaromatic molecular core that takes part in stacking-interactions with the pi-systems of the nitrogenous bases of DNA.


Subject(s)
Coumarins/chemistry , Furocoumarins/chemistry , Intercalating Agents/chemistry , Mutagens/chemistry , Plant Extracts/chemistry , Allium/cytology , Apiaceae/chemistry , Comet Assay , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Single-Stranded/drug effects , Escherichia coli/drug effects , Guanine/chemistry , Hydrogen Bonding , Meristem/drug effects , Molecular Docking Simulation , Salmonella typhimurium/drug effects
16.
J Pharm Biomed Anal ; 176: 112786, 2019 Nov 30.
Article in English | MEDLINE | ID: mdl-31398506

ABSTRACT

Electrochemical techniques were used to investigate the behavior of lomustine (CCNU) and its degradation in aqueous solution at a glassy carbon electrode (GCE). The in situ interaction of CCNU and chemically degraded CCNU (cdCCNU) with dsDNA was then investigated in dsDNA incubated solutions, using dsDNA electrochemical biosensors and comet assays. CCNU undergoes electrochemical reduction in two irreversible, diffusion-controlled, and pH-dependent redox processes, each with transfer of two electrons and one proton. At pH ≥ 10.1, the peak potential for the two processes was essentially pH-independent and involved only one electron. A mechanism was proposed for the reduction of CCNU in a neutral medium. In addition, it was found that CCNU underwent spontaneous degradation during incubation in aqueous solution, without the formation of electroactive degradation products. The degradation process was faster in basic media. Moreover, this pro-drug interacted with the DNA. Its metabolite(s) initially caused condensation of the double helix chains, followed by the unwinding of these chains. In addition, free guanine (Gua) was released from the dsDNA and oxidative damage to the DNA by the CCNU metabolite(s) was evidenced from the detection of 8-oxoGua and 2,8-oxoAde. These results were confirmed by the poly(dA)- and poly(dG)-polyhomonucleotide biosensors, which revealed the oxidative damage caused to both bases (guanine and adenine) of the dsDNA by the CCNU metabolite(s). The comet assay indicated breaks in the single strand DNA, complementing the results of the studies using differential pulse voltammetry. Conformational changes of dsDNA caused by CCNU and cdCCNU were confirmed using comet assays.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Breaks, Single-Stranded/drug effects , DNA/drug effects , Lomustine/pharmacology , Antineoplastic Agents, Alkylating/chemistry , Biosensing Techniques , DNA/chemistry , Diffusion , Drug Stability , Electrochemical Techniques , Electrodes , Lomustine/chemistry , Nucleic Acid Conformation/drug effects , Water
17.
Nucleic Acids Res ; 47(17): 9132-9143, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31329989

ABSTRACT

Poly(ADP-ribose) polymerases (PARPs) facilitate the repair of DNA single-strand breaks (SSBs). When PARPs are inhibited, unrepaired SSBs colliding with replication forks give rise to cytotoxic double-strand breaks. These are normally rescued by homologous recombination (HR), but, in cells with suboptimal HR, PARP inhibition leads to genomic instability and cell death, a phenomenon currently exploited in the therapy of ovarian cancers in BRCA1/2 mutation carriers. In spite of their promise, resistance to PARP inhibitors (PARPis) has already emerged. In order to identify the possible underlying causes of the resistance, we set out to identify the endogenous source of DNA damage that activates PARPs. We argued that if the toxicity of PARPis is indeed caused by unrepaired SSBs, these breaks must arise spontaneously, because PARPis are used as single agents. We now show that a significant contributor to PARPi toxicity is oxygen metabolism. While BRCA1-depleted or -mutated cells were hypersensitive to the clinically approved PARPi olaparib, its toxicity was significantly attenuated by depletion of OGG1 or MYH DNA glycosylases, as well as by treatment with reactive oxygen species scavengers, growth under hypoxic conditions or chemical OGG1 inhibition. Thus, clinical resistance to PARPi therapy may emerge simply through reduced efficiency of oxidative damage repair.


Subject(s)
BRCA1 Protein/genetics , DNA Glycosylases/genetics , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerases/genetics , Cell Line, Tumor , DNA Breaks, Single-Stranded/drug effects , DNA Damage/drug effects , DNA Glycosylases/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Female , Homologous Recombination/drug effects , Homologous Recombination/genetics , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Oxidation-Reduction/drug effects , Phthalazines/adverse effects , Phthalazines/pharmacology , Piperazines/adverse effects , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Synthetic Lethal Mutations/genetics
18.
Environ Pollut ; 252(Pt A): 917-923, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31226516

ABSTRACT

Glyphosate-based herbicides (GBH) are the most widely used pesticides in the world. The extensive use of them increases the potential human health risk, including the human inhalation toxicity risk. We studied the effect of the most famous GBH Roundup® (RDP) in the concentration range from 50 to 125 µg/mL on Mitochondria-Associated apoptosis and DNA damage in Human alveolar carcinoma cells (A549 cells). Alkaline comet assay, immunofluorescence assay and Flow Cytometric Analysis assay were employed to detect DNA damages and apoptosis of A549 cells. We found RDP caused concentration-dependent increases in DNA damages and proportion of apoptotic cells in A549 cells. RDP induced the DNA single-strand breaks and double-strand breaks; the collapse of mitochondrial membrane by increasing Bax/Bcl-2, resulting in the release of cytochrome c into cytosol and then activated caspase-9/-3, cleaved poly (ADP-ribose) polymerase (PARP) in human lung tissue cells. The results demonstrate that RDP can induce A549 cells cytotoxic effects in vitro at the concentration lower than the occupational exposures level of workers, which means RDP has a potential threat to human health.


Subject(s)
Apoptosis/drug effects , Glycine/analogs & derivatives , Herbicides/toxicity , Lung/pathology , Mitochondria/drug effects , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/genetics , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Comet Assay , Cytochromes c/metabolism , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Single-Stranded/drug effects , Glycine/toxicity , Humans , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism , Glyphosate
19.
Future Oncol ; 15(17): 2053-2068, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30942614

ABSTRACT

DC Bead is designed for the embolization of liver malignancies combined with local sustained chemotherapy delivery. It was first demonstrated around a decade ago that irinotecan could be loaded into DC Bead and used in a transarterially directed procedure to treat colorectal liver metastases, commonly referred to as drug-eluting bead with irinotecan (DEBIRI). Despite numerous reports of its safe and effective use in treating colorectal liver metastases patients, there remains a perceived fundamental paradox as to how this treatment works. This review of the mechanism of action of DEBIRI provides a rationale for why intra-arterial delivery of this prodrug from an embolic bead provides for enhanced tumor selectivity, sparing the normal liver while reducing adverse side effects associated with the irinotecan therapy.


Subject(s)
Chemoembolization, Therapeutic/methods , Drug Delivery Systems/methods , Irinotecan/pharmacology , Liver Neoplasms/drug therapy , Topoisomerase I Inhibitors/pharmacology , Clinical Trials as Topic , Colorectal Neoplasms/pathology , DNA Breaks, Single-Stranded/drug effects , DNA Repair/drug effects , DNA Replication/drug effects , Drug Compounding/methods , Humans , Injections, Intra-Arterial , Irinotecan/therapeutic use , Liver Neoplasms/secondary , Particle Size , Topoisomerase I Inhibitors/therapeutic use , Treatment Outcome
20.
Food Chem Toxicol ; 124: 349-358, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30562542

ABSTRACT

The cyanobacterial pentapeptide nodularin (NOD), mainly produced by genus Nodularia, is a potent inhibitor of protein phosphatases PP1 and PP2A, and causes animal mortality. The few studies available indicate that NOD is a potential non-genotoxic carcinogen. In the present study we evaluated NOD (0.01, 0.1 and 1 µg/ml) genotoxic activity in human hepatoma (HepG2) cells with the comet, γH2AX and cytokinesis block micronucleus cytome assays. In addition, induction of oxidative stress was studied. Moreover changes in the expression of selected genes from the P53 pathway, involved in the response to DNA damage (P53, GADD45α, CDKN1A, MDM2), apoptosis (BAX, BCL2) and oxidative stress (GPX1, GSR, GCLC, CAT, SOD1) were determined using qPCR. Non-cytotoxic concentrations induced time and dose dependant increase in reactive oxygen species (ROS) production and substantially increased the formation of oxidative DNA damage. In addition, elevated formation of micronuclei was detected. For the first time it has been shown that NOD deregulated the mRNA level of DNA damage (CDKN1A, GADD45α) and oxidative stress (GPX1, GSR, GCLC, CAT and SOD1) responsive genes and anti-apoptotic gene BCL2. Our results provide new evidence that NOD genotoxic effects are mediated through ROS production, already at low environmentally relevant concentrations.


Subject(s)
Mutagens/toxicity , Peptides, Cyclic/toxicity , Apoptosis/drug effects , DNA/metabolism , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Single-Stranded/drug effects , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...