Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Chromosome Res ; 30(4): 459-476, 2022 12.
Article in English | MEDLINE | ID: mdl-35604590

ABSTRACT

ATM and DNA-PKcs coordinate the DNA damage response at multiple levels following the exposure to chemotherapy. The Topoisomerase II poison etoposide (ETO) is an effective chemotherapeutic agent that induces DNA double-strand breaks (DSB), but it is responsible from the chromosomal rearrangements frequently found in therapy-related secondary tumors. Targeted inhibition of DNA-PKcs in ATM-defective tumors combined with radio- or chemotherapy has been proposed as relevant therapies. Here, we explored the DNA repair mechanisms and the genetic consequences of targeting the non-oncogenic addiction to DNA-PKcs of ATM-defective tumor cells after exposure to ETO. We demonstrated that chemical inhibition of DNA-PKcs followed by treatment with ETO resulted in the accumulation of chromatid breaks and decreased mitotic index in both A-T cells and ATM-knocked-down (ATMkd) tumor cells. The HR repair process in DNA-PKcs-inhibited ATMkd cells amplified the RAD51 foci number, with no correlated increase in sister chromatid exchanges. The analysis of post-mitotic DNA lesions presented an augmented number of persistent unresolved DSB, without alterations in the cell cycle progression. Long-term examination of chromosome aberrations revealed a strikingly high number of chromatid and chromosome exchanges. By using genetic and pharmacological abrogation of PARP-1, we demonstrated that alternative end-joining (alt-EJ) repair pathway is responsible for those chromosome abnormalities generated by limiting c-NHEJ activities during directed inhibition of DNA-PKcs in ATM-deficient cells. Targeting the non-oncogenic addiction to DNA-PKcs of ATM-defective tumors stimulates the DSB repair by alt-EJ, which is liable for the origin of cells carrying stable chromosome aberrations that may eventually restrict the therapeutic strategy.


Subject(s)
Chromosome Aberrations , DNA Breaks, Double-Stranded , Humans , Etoposide/pharmacology , Cell Line , DNA Repair , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , DNA/genetics , DNA End-Joining Repair , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism
2.
Mutagenesis ; 34(3): 245-252, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31037299

ABSTRACT

Genomic instability is a hallmark of cancer, contributing to tumour development and transformation, being chromosome instability (CIN) the most common form in human cancer. Chronic lymphocytic leukaemia (CLL) is the most frequent adult leukaemia in the Western world. In this study, we have evaluated basal CIN in untreated patients with CLL by measuring chromosome aberrations (CAs) and micronucleus (MN) frequency and their association with different prognostic factors. Seventy-two patients and 21 normal controls were analysed. Cytogenetic and fluorescence in situ hybridisation (FISH) studies were performed. IGHV (immunoglobulin heavy chain variable region) mutational status was evaluated by reverse transcription polymerase chain reaction and sequencing. An increased number of CA in patients compared with controls (P = 0.0001) was observed. Cases with abnormal karyotypes showed increased CA rate than those with normal karyotypes (P = 0.0026), with a particularly highest frequency in cases with complex karyotypes. Among FISH risk groups, a significant low frequency of CA was found in patients with no FISH alterations compared to those with del13q14 and ≥2 FISH alterations (P = 0.0074). When mean CA value (6.7%) was considered, significant differences in the distribution of low and high CA frequency between cases with normal and abnormal karyotypes (P = 0.002) were observed. By MN analysis, higher frequency in patients compared to controls (P = 0.0001) was also found, as well as between cases with ≥2 FISH abnormalities and those with no FISH alterations (P = 0.026). Similarly, significant differences were observed when patients were divided according to mean MN frequency (2.2%; P ≤ 0.04). Interestingly, patients with high MN frequency had shorter time to first treatment than those with low frequency (P = 0.024). Cases with mutated and unmutated IGHV status showed increased CA and MN frequencies compared to controls (P ≤ 0.0007), but no differences between both groups were found. Our results support the strong interaction between CIN and genomic complexity as well as their influence on poor outcome in this pathology.


Subject(s)
Chromosomal Instability , Chromosome Aberrations , Genetic Predisposition to Disease , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Carcinoma in Situ , Female , Genetic Association Studies , Genomic Instability , Humans , Immunoglobulin Variable Region , In Situ Hybridization, Fluorescence , Karyotyping , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Male , Middle Aged , Mutation
3.
Cytometry A ; 89(9): 852-60, 2016 09.
Article in English | MEDLINE | ID: mdl-27517472

ABSTRACT

Topoisomerase II (Top2) is an important target for anticancer therapy. A variety of drugs that poison Top2, including several epipodophyllotoxins, anthracyclines, and anthracenediones, are widely used in the clinic for both hematologic and solid tumors. The poisoning of Top2 involves the formation of a reaction intermediate Top2-DNA, termed Top2 cleavage complex (Top2cc), which is persistent in the presence of the drug and involves a 5' end of DNA covalently bound to a tyrosine from the enzyme through a phosphodiester group. Drug-induced Top2cc leads to Top2 linked-DNA breaks which are the major responsible for their cytotoxicity. While biochemical detection is very laborious, quantification of drug-induced Top2cc by immunofluorescence-based microscopy techniques is time consuming and requires extensive image segmentation for the analysis of a small population of cells. Here, we developed a flow cytometry-based method for the analysis of drug-induced Top2cc. This method allows a rapid analysis of a high number of cells in their cell cycle phase context. Moreover, it can be applied to almost any human cell type, including clinical samples. The methodology is useful for a high-throughput analysis of drugs that poison Top2, allowing not just the discrimination of the Top2 isoform that is targeted but also to track its removal. © 2016 International Society for Advancement of Cytometry.


Subject(s)
DNA Topoisomerases, Type II/isolation & purification , DNA-Binding Proteins/isolation & purification , Flow Cytometry/methods , Neoplasms/drug therapy , Topoisomerase II Inhibitors/chemistry , Anthraquinones/chemistry , Anthraquinones/isolation & purification , Anthraquinones/therapeutic use , DNA Damage/drug effects , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/drug effects , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/drug effects , Drug Resistance, Neoplasm/genetics , High-Throughput Screening Assays , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/isolation & purification , Topoisomerase II Inhibitors/isolation & purification , Topoisomerase II Inhibitors/therapeutic use
4.
Chromosome Res ; 23(4): 719-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26152239

ABSTRACT

Etoposide (ETO), a drug used for the treatment of human tumors, is associated with the development of secondary malignancies. Recently, therapeutic strategies have incorporated chemosensitizing agents to improve the tumoral response to this drug. ETO creates DNA double-strand breaks (DSB) via inhibition of DNA topoisomerase II (Top2). To repair DSB, homologous recombination (HR) and non-homologous end-joining (NHEJ), involving D-NHEJ (dependent of the catalytic subunit of DNA-dependent protein kinase, DNA-PKcs) and B-NHEJ (backup repair pathway) are activated. We evaluated the progression of the DNA damage induced by the Top2 poison ETO in G2 phase of human HeLa cells after chemical inhibition of DNA-PKcs with NU7026. Compared to ETO treatment alone, this combined treatment resulted in a twofold higher rate of chromatid breaks and exchanges when analysis was performed in the following metaphase. Moreover, when analysis was performed in the second metaphase following treatment, increases in the percentage of micronuclei with H2AX (biomarker for DSB) foci in binucleated cells and dicentric chromosomes were seen. In post-mitotic G1 phase, a close association between unresolved DSB and meiotic recombination 11 homolog A (MRE11) signals was observed, demonstrating the contribution of MRE11 in the DSB repair by B-NHEJ. Hence, chemical inhibition of DNA-PKcs impaired both D-NHEJ and HR repair pathways, altering the maintenance of chromosomal integrity and cell proliferation. Our results suggest that the chemosensitizing effectiveness of the DNA-PKcs inhibitor and the survival rate of aberrant cells may contribute to the development of therapy-related tumors.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Chromosome Aberrations/chemically induced , DNA-Activated Protein Kinase/deficiency , Etoposide/toxicity , G2 Phase/drug effects , G2 Phase/genetics , DNA Breaks, Double-Stranded/drug effects , DNA End-Joining Repair/drug effects , DNA-Binding Proteins/metabolism , Gene Rearrangement , HeLa Cells , Histones/metabolism , Homologous Recombination , Humans , MRE11 Homologue Protein , Mitotic Index , Protein Binding , Rad51 Recombinase/metabolism , Sister Chromatid Exchange/genetics
5.
DNA Repair (Amst) ; 6(5): 626-38, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17218167

ABSTRACT

Genome integrity and cell proliferation and survival are regulated by an intricate network of pathways that includes cell cycle checkpoints, DNA repair and recombination, and programmed cell death. It makes sense that there should be a coordinated regulation of these different processes, but the components of such mechanisms remain unknown. In this report, we demonstrate that p19INK4d expression enhances cell survival under genotoxic conditions. By using p19INK4d-overexpressing clones, we demonstrated that p19INK4d expression correlates with the cellular resistance to UV treatment with increased DNA repair activity against UV-induced lesions. On the contrary, cells transfected with p19INK4d antisense cDNA show reduced ability to repair DNA damage and increased sensitivity to genotoxic insult when compared with their p19INK4d-overexpressing counterparts. Consistent with these findings, our studies also show that p19INK4d-overexpressing cells present not only a minor accumulation of UV-induced chromosomal aberrations but a lower frequency of spontaneous chromosome abnormalities than p19INK4d-antisense cells. Lastly, we suggest that p19INK4d effects are dissociated from its role as CDK4/6 inhibitor. The results presented herein support a crucial role for p19INK4d in regulating genomic stability and overall cell viability under conditions of genotoxic stress. We propose that p19INK4d would belong to a protein network that would integrate DNA repair, apoptotic and checkpoint mechanisms in order to maintain the genomic integrity.


Subject(s)
Cell Survival/physiology , Chromosome Aberrations , Cyclin-Dependent Kinase Inhibitor p19/metabolism , Cyclin-Dependent Kinase Inhibitor p19/pharmacology , DNA Damage/radiation effects , DNA Repair/physiology , Animals , Blotting, Northern , Blotting, Western , Cell Survival/radiation effects , Colony-Forming Units Assay , DNA Repair/radiation effects , Genomic Instability , Humans , Immunoprecipitation , Mice , Pyrimidine Dimers , RNA, Messenger/genetics , Radiation Tolerance , Thymidine/metabolism , Ultraviolet Rays
6.
Mutat Res ; 646(1-2): 8-16, 2008 Nov 10.
Article in English | MEDLINE | ID: mdl-18812179

ABSTRACT

Fludarabine (FLU), an analogue of adenosine, interferes with DNA synthesis and inhibits the chain elongation leading to replication arrest and DNA double strand break (DSB) formation. Mammalian cells use two main pathways of DSB repair to maintain genomic stability: homologous recombination (HR) and non-homologous end joining (NHEJ). The aim of the present work was to evaluate the repair pathways employed in the restoration of DSB formed following replication arrest induced by FLU in mammalian cells. Replication inhibition was induced in human lymphocytes and fibroblasts by FLU. DSB occurred in a dose-dependent manner on early/middle S-phase cells, as detected by gammaH2AX foci formation. To test whether conservative HR participates in FLU-induced DSB repair, we measured the kinetics of Rad51 nuclear foci formation in human fibroblasts. There was no significant induction of Rad51 foci after FLU treatment. To further confirm these results, we analyzed the frequency of sister chromatid exchanges (SCE) in both human cells. We did not find increased frequencies of SCE after FLU treatment. To assess the participation of NHEJ pathway in the repair of FLU-induced damage, we used two chemical inhibitors of the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs), vanillin and wortmannin. Human fibroblasts pretreated with DNA-PKcs inhibitors showed increased levels of chromosome breakages and became more sensitive to cell death. An active role of NHEJ pathway was also suggested from the analysis of Chinese hamster cell lines. XR-C1 (DNA-PKcs-deficient) and XR-V15B (Ku80-deficient) cells showed hypersensitivity to FLU as evidenced by the increased frequency of chromosome aberrations, decreased mitotic index and impaired survival rates. In contrast, CL-V4B (Rad51C-deficient) and V-C8 (Brca2-deficient) cell lines displayed a FLU-resistant phenotype. Together, our results suggest a major role for NHEJ repair in the preservation of genome integrity against FLU-induced DSB in mammalian cells.


Subject(s)
DNA Ligases/metabolism , DNA Repair , DNA-Activated Protein Kinase/metabolism , DNA/metabolism , Adult , Androstadienes/pharmacology , Animals , Benzaldehydes/pharmacology , CHO Cells , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromosome Aberrations/drug effects , Cricetinae , Cricetulus , DNA/genetics , DNA Breaks, Double-Stranded , DNA Ligases/genetics , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/genetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/toxicity , Female , Fibroblasts/metabolism , Foreskin/cytology , Humans , Lymphocytes/metabolism , Male , Mitotic Index , S Phase/drug effects , Vidarabine/analogs & derivatives , Vidarabine/toxicity , Wortmannin
7.
Cancer Chemother Pharmacol ; 59(3): 349-60, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17009037

ABSTRACT

PURPOSE: Chronic myeloid leukemia is a clonal myeloproliferative disorder characterized by the presence of the fusion gene BCR/ABL. We had previously demonstrated an increased proapoptotic effect of imatinib (STI571) in combination with amifostine (AMI) in K562 cell line. In this study, we used genomic scale gene expression profiling to monitor changes at transcriptional level in K562 cells during the treatment with AMI + STI571. MATERIALS AND METHODS: cRNA from Control and treated K562 cells were mixed in equal amounts and incubated with a microarray slide for hybridization. RNA from six independent paired experiments was subjected to transcriptional profiling. With the aim to automate the process of biological theme determination, selected genes were further analyzed by EASE. Validation of the expression was carried out by quantitative real-time PCR and western blotting. RESULTS: As expected, a small percentage of genes accounts for the effects of the combined drug treatment. We identified 61 sequences corresponding to known genes; 17 of the 61 genes were up regulated, such as RHO6, PPP2R5E, PPM1E and BTF that appear to reflect favorable events for apoptosis induction. Between down regulated genes, API5, TUBB2 and TLK1 are also of considerable interest. CONCLUSION: We identified a transcriptional repressor of survival genes, known as BTF, which triggers a proapoptotic signal, potentially helpful to overcome the resistance to STI571. This finding could be particularly useful to design novel therapeutic strategies for leukemia patients. This study demonstrates the importance of in vitro testing of a novel drug combination most likely to predict its potential usefulness for in vivo application.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Gene Expression Regulation, Neoplastic/physiology , Transcription, Genetic/drug effects , Amifostine/administration & dosage , Benzamides , Cell Survival/drug effects , DNA-Binding Proteins/genetics , Drug Combinations , Drug Screening Assays, Antitumor , Gene Expression Profiling , Humans , Imatinib Mesylate , K562 Cells/drug effects , K562 Cells/pathology , Oligonucleotide Array Sequence Analysis/methods , Piperazines/administration & dosage , Pyrimidines/administration & dosage , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
8.
Curr Protoc Cytom ; 81: 7.48.1-7.48.8, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28678420

ABSTRACT

The poisoning of Topoisomerase II (Top2) has been found to be useful as a therapeutic strategy for the treatment of several tumors. The mechanism of Top2 poisons involves a drug-mediated stabilization of a Top2-DNA complex, termed Top2 cleavage complex (Top2cc), which maintains a 5' end of DNA covalently bound to a tyrosine from Top2 through a phosphodiester group. Drug-stabilized Top2cc leads to Top2-linked-DNA breaks, which are believed to mediate their cytotoxicity. Several time-consuming or cell type-limiting assays have been used in the past to study drug-stabilized Top2cc. Here, we describe a flow cytometry-based method that allows a rapid assessment of drug-induced Top2cc, which is suitable for high throughput analysis in almost any kind of human cell. The analyses of the drug-induced Top2cc in the cell cycle context and the possibility to track its removal are additional benefits from this methodology. © 2017 by John Wiley & Sons, Inc.


Subject(s)
DNA Topoisomerases, Type II/analysis , DNA/analysis , Etoposide/chemistry , Flow Cytometry/methods , Animals , DNA/chemistry , DNA/metabolism , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/metabolism , Etoposide/pharmacology , HL-60 Cells , Humans
9.
Cancer Chemother Pharmacol ; 55(6): 602-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15700189

ABSTRACT

PURPOSE: To study the apoptotic effect of the 2-phenylaminopyrimidine derivative STI571 in combination with antioxidant agents on K-562 cell line derived from a Philadelphia chromosome-positive chronic myeloid leukemia patient. MATERIALS AND METHODS: K-562 (BCR/ABL+), U-937, and HL60 (BCR/ABL-) leukemic cell lines were incubated with STI571 and the antioxidant agents catalase, glutathione, superoxide dismutase, and amifostine (AMI). Apoptotic effect was analyzed by morphological and flow cytometric criteria. RESULTS: STI571 at concentrations higher than 0.25 mumol L(-1) produced apoptosis (P<0.05) in K-562 cells only after treatment for 72 h. At the mentioned concentrations, STI571 also induced an increase in the loss of mitochondrial transmembrane potential from 24.6 to 40%. Combination of STI571 (0.5 micromol L(-1)) with antioxidant agents showed that the cytoprotective agent AMI (0.75 mg mL(-1)) produced an additive effect in the proapoptotic activity of STI571 in K-562 cells at nuclear (58.8%+/-2.0 vs. 28.9%+/-3.3) and mitochondrial (53.3%+/-3.6 vs. 29.5%+/-1.2) levels. CONCLUSIONS: Our results show that only AMI in combination with STI571, at submicromolar concentration, has an additive effect in K-562 cell line, and it does not have severe toxic effects on Philadelphia chromosome negative cells.


Subject(s)
Amifostine/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Pyrimidines/pharmacology , Antioxidants/pharmacology , Benzamides , Cell Cycle/drug effects , Cell Proliferation/drug effects , Comet Assay , Cytoprotection , Drug Screening Assays, Antitumor , Drug Synergism , HL-60 Cells , Humans , Imatinib Mesylate , K562 Cells , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Piperazines
10.
Mutat Res ; 781: 37-48, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26421495

ABSTRACT

Tyrosyl-DNA-phosphodiesterase 1 (TDP1) is a DNA repair enzyme that removes irreversible protein-linked 3' DNA complexes, 3' phosphoglycolates, alkylation damage-induced DNA breaks, and 3' deoxyribose nucleosides. In addition to its extended spectrum of substrates, TDP1 interacts with several DNA damage response factors. To determine whether TDP1 participates in the repair of topoisomerase II (Top2) induced DNA lesions, we generated TDP1 depleted (TDP1kd) human tumoral cells. We found that TDP1kd cells are hypersensitive to etoposide (ETO). Moreover, we established in a chromatin context that following treatment with ETO, TDP1kd cells accumulate increased amounts of Top2α cleavage complexes, removing them with an altered kinetics. We also showed that TDP1 depleted cells accumulate increased γH2AX and pS296Chk1 signals following treatment with ETO. Similarly, cytogenetics analyses following Top2 poisoning revealed increased amounts of chromatid and chromosome breaks and exchanges on TDP1kd cells in the presence or not of the DNA-PKcs inhibitor NU7026. However, the levels of sister chromatid exchanges were similar in both TDP1kd and control non-silenced cell lines. This suggests a role of TDP1 in both canonical non-homologous end joining and alternative end joining, but not in the homologous recombination repair pathway. Finally, micronucleus analyses following ETO treatment revealed a higher frequency of micronucleus containing γH2AX signals on TDP1kd cells. Together, our results highlight an active role of TDP1 in the repair of Top2-induced DNA damage and its relevance on the genome stability maintenance in human cells.


Subject(s)
Antigens, Neoplasm/toxicity , DNA Damage/genetics , DNA End-Joining Repair/genetics , DNA Topoisomerases, Type II/toxicity , DNA-Binding Proteins/toxicity , Phosphoric Diester Hydrolases/metabolism , Chromones , Colony-Forming Units Assay , DNA End-Joining Repair/physiology , DNA Primers/genetics , Etoposide/pharmacology , Flow Cytometry , Fluorescent Antibody Technique , Gentian Violet , HeLa Cells , Histones/metabolism , Humans , Immunoblotting , Micronucleus Tests , Morpholines , Phosphoric Diester Hydrolases/deficiency , Phosphoric Diester Hydrolases/genetics , Poly-ADP-Ribose Binding Proteins , Real-Time Polymerase Chain Reaction
12.
Leuk Res ; 26(7): 669-76, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12008085

ABSTRACT

We have evaluated the apoptotic and DNA damaging activity of Idarubicin (IDA) on K-562 cells alone and following the uptake of modified antisense-oligodeoxynucleotides (AS-ODNs) targeting b3a2 mRNA of bcr/abl hybrid gene, after treatment with AS-ODNs/DCChol-DOPE (liposomes) complexes. The uptake of FITC-labeled oligonucleotide-liposomes complexes (FITC-ODNs/DCChol-DOPE) was analyzed by flow cytometry and fluorescence microscopy. Both techniques indicated cytoplasmic accumulation of labeled liposome complexes following 24h of exposure. In absence of liposomes, AS-ODNs uptake was minimal. Pre-treatment of cells with AS-ODNs/DCChol-DOPE increased the capability of IDA to induce apoptosis as determined by morphology and the comet assay. In contrast, the use of a non-sense oligodeoxynucleotide conjugated with liposomes, in the presence of IDA, did not increase K-562 cell apoptosis. Nevertheless, DNA damage in IDA treated cells was not related to ODNs/liposomes pre-treatment, as determined by the comet assay. Our data suggests that DCChol-DOPE increases the uptake of ODNs in K-562 cells, and these modified AS-ODNs increase IDA induced apoptosis by decreasing p210(bcr/abl) levels in K-562 cells.


Subject(s)
Apoptosis/drug effects , Cholesterol/analogs & derivatives , Idarubicin/pharmacology , K562 Cells/drug effects , Oligodeoxyribonucleotides, Antisense/metabolism , Biological Transport , Cations/chemistry , Cholesterol/chemistry , Comet Assay , DNA Damage , DNA, Neoplasm/analysis , Drug Synergism , Flow Cytometry , Fusion Proteins, bcr-abl/biosynthesis , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic/drug effects , Humans , K562 Cells/metabolism , Lipids/chemistry , Liposomes/administration & dosage , Liposomes/chemistry , Microscopy, Fluorescence , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/pharmacology , Phosphatidylethanolamines/chemistry , Quaternary Ammonium Compounds/chemistry , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Neoplasm/antagonists & inhibitors , RNA, Neoplasm/genetics
13.
Environ Mol Mutagen ; 39(1): 3-9, 2002.
Article in English | MEDLINE | ID: mdl-11813290

ABSTRACT

In this study we evaluated the antigenotoxic and cytoprotective capabilities of WR-2721 [S-2-(3-aminopropylamino)-ethylphosphorothioic acid (amifostine)] in different normal tissues of BALB/c mice treated with idarubicin [4-demethoxydaunorubicin (IDA)]. The aminothiol WR-2721 is a pro-drug that requires dephosphorylation to its active metabolite WR-1065, to produce selectively cytoprotective activity in normal tissues exposed to radio- and chemotherapeutic agents, without protecting malignant tissues. IDA is an effective chemotherapeutic agent against hematological diseases, but produces a broad spectrum of toxicity in nontumoral cells. Animals were injected intravenously with WR-2721 (250 mg/kg) or IDA (6 mg/kg) and WR-2721/IDA. Micronuclei frequency in bone marrow was measured 24 and 48 hr after initiation of the treatments. The IDA-treated group showed increased levels of micronuclei. However, the WR-2721- and WR-2721/IDA-treated groups did not show differences from the controls. Genetic damage was evaluated by alkaline single-cell gel electrophoresis at 24-hr posttreatments. Important DNA damage was observed in liver, spleen, and peripheral blood cells of mice treated with IDA. The presence of WR-2721 diminished that damaging effect only in liver cells. The apoptotic index was measured in liver and spleen tissues by the TUNEL assay 14 and 24 hr after treatment. In liver we observed an increased percentage of apoptotic cells at 24 hr for the IDA-treated group, whereas the WR-2721 and WR-2721/IDA groups remained at low levels. Splenic cells treated with IDA and WR-2721/IDA showed increased DNA fragmentation levels at any time. In conclusion, WR-2721 has a tissue-specific antigenotoxic and cytoprotective effect in IDA-treated mice using these experimental conditions.


Subject(s)
Amifostine/pharmacology , Antibiotics, Antineoplastic/adverse effects , Antimutagenic Agents/pharmacology , Idarubicin/adverse effects , Protective Agents/pharmacology , Animals , Apoptosis/drug effects , Blood/drug effects , Bone Marrow/drug effects , Comet Assay/methods , DNA Damage/drug effects , Female , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred BALB C , Micronucleus Tests , Spleen/drug effects , Spleen/pathology , Toxicity Tests
14.
Toxicology ; 171(2-3): 215-22, 2002 Feb 28.
Article in English | MEDLINE | ID: mdl-11836027

ABSTRACT

Fludarabine (FLU, a fluorinated purine analog) and idarubicin (IDA, a DNA-topoisomerase II poison) are frequently used in cancer chemotherapy. The effects of these drugs on cultured normal human lymphocytes were studied to establish the possible involvement of chromosome damage in the apoptotic program. Chromosome aberrations (CA) were evaluated in first division metaphases and the apoptotic process was measured by morphological and electrophoretical techniques. The percentage of abnormal cells was increased from the doses of FLU 1.0 microg/ml and IDA 0.005 microg/ml (P<0.0001) with an important decrease in the mitotic index (MI) for the highest doses assayed. A significant dose-dependent induction of abnormal cells was observed for both drugs. An increase of apoptotic cells was found at 5.0 and 10.0 microg/ml of FLU (P<0.001) while IDA activated apoptosis at 0.05 microg/ml (P<0.01) and markedly from 0.1 microg/ml (P<0.001). These increments were dose dependent. Apoptotic cell morphology was associated with DNA fragmentation at the highest doses. The increased induction of abnormal cells and the decreased MI were in correlation with the apoptotic index for FLU and IDA, suggesting the role of CA in drug-induced cell death.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Antineoplastic Agents/toxicity , Idarubicin/toxicity , Lymphocytes/drug effects , Mutagens/toxicity , Vidarabine/toxicity , Apoptosis , Cells, Cultured , Chromosome Aberrations , DNA Fragmentation , Dose-Response Relationship, Drug , Fluorescent Dyes , Humans , Lymphocytes/pathology , Lymphocytes/ultrastructure , Vidarabine/analogs & derivatives
15.
Environ Mol Mutagen ; 53(8): 608-18, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22987276

ABSTRACT

The role of DNA double strand break (DSB) repair pathways, non-homologous end joining (NHEJ), and homologous recombination (HR) was evaluated to prevent the chromosome instability induced by the topoisomerase II (Top2) poisons, idarubicin, and etoposide in Chinese hamster cell lines. XR-C1 (DNA-PKcs deficient) and V-C8 (BRCA2 deficient) showed higher sensitivity to increased concentrations of Top2 poisons compared with their normal counterparts, CHO9 and V79. Both proficient and deficient cells exhibited a marked DSB induction in all phases of the cell cycle. Additionally, deficient cells showed persistent DNA damage 24 hr post-treatment. Chromosomal aberrations increased in the first mitosis following Top2 poison-treatments in G1 or G2 in proficient and deficient cells. CHO9 and V79 demonstrated chromosome and chromatid exchanges following treatments in G1 and G2 phases, respectively. Deficient cells showed high frequencies of chromatid exchanges following treatments in G1 and G2. Simultaneously, we analyzed the micronuclei (MN) induction in interphase cells after treatments in G1, S, or G2 of the previous cell cycle. Both Top2 poisons induced an important increase in MN in CHO9, V79, and V-C8 cells. XR-C1 exhibited an increased MN frequency when cells were treated in G1 phase but not in S or G2. This MN reduction was due to a cell accumulation at G2/M and death in G2-treated cells. Our data suggest that NHEJ and HR operate differentially throughout the cell cycle to protect from Top2 poison-induced chromosome instability, and that DNA-PKcs-dependent NHEJ pathway allows the survival of chromosome damaged cells during S/G2 to the next interphase.


Subject(s)
Chromosome Aberrations/drug effects , DNA Breaks, Double-Stranded/drug effects , DNA Topoisomerases, Type II/metabolism , Animals , CHO Cells , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line , Cricetinae , Cricetulus , Etoposide/toxicity , G1 Phase/drug effects , G1 Phase/genetics , G2 Phase/drug effects , G2 Phase/genetics , Idarubicin/toxicity , Micronuclei, Chromosome-Defective/chemically induced
16.
PLoS One ; 5(9)2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20824055

ABSTRACT

Topoisomerase II (Top2) is a nuclear enzyme involved in several metabolic processes of DNA. Chemotherapy agents that poison Top2 are known to induce persistent protein-mediated DNA double strand breaks (DSB). In this report, by using knock down experiments, we demonstrated that Top2alpha was largely responsible for the induction of gammaH2AX and cytotoxicity by the Top2 poisons idarubicin and etoposide in normal human cells. As DSB resulting from Top2 poisons-mediated damage may be repaired by non-homologous end joining (NHEJ) or homologous recombination (HR), we aimed to analyze both DNA repair pathways. We found that DNA-PKcs was rapidly activated in human cells, as evidenced by autophosphorylation at serine 2056, following Top2-mediated DNA damage. The chemical inhibition of DNA-PKcs by wortmannin and vanillin resulted in an increased accumulation of DNA DSB, as evaluated by the comet assay. This was supported by a hypersensitive phenotype to Top2 poisons of Ku80- and DNA-PKcs- defective Chinese hamster cell lines. We also showed that Rad51 protein levels, Rad51 foci formation and sister chromatid exchanges were increased in human cells following Top2-mediated DNA damage. In support, BRCA2- and Rad51C- defective Chinese hamster cells displayed hypersensitivity to Top2 poisons. The analysis by immunofluorescence of the DNA DSB repair response in synchronized human cell cultures revealed activation of DNA-PKcs throughout the cell cycle and Rad51 foci formation in S and late S/G2 cells. Additionally, we found an increase of DNA-PKcs-mediated residual repair events, but not Rad51 residual foci, into micronucleated and apoptotic cells. Therefore, we conclude that in human cells both NHEJ and HR are required, with cell cycle stage specificity, for the repair of Top2-mediated reversible DNA damage. Moreover, NHEJ-mediated residual repair events are more frequently associated to irreversibly damaged cells.


Subject(s)
Cell Cycle , DNA Damage , DNA Repair , DNA Topoisomerases, Type II/metabolism , Recombination, Genetic , Animals , Cell Line , Cricetinae , Cricetulus , DNA Breaks, Double-Stranded , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans
17.
Acta toxicol. argent ; 14(supl): 48-51, ago. 2006. tab, graf
Article in Spanish | LILACS | ID: lil-576396

ABSTRACT

El arsénico es un contaminante ampliamente distribuido en el mundo. En Argentina se estima que cerca de un millón y medio de personas están expuestas a sus efectos deletéreos por ingesta de agua con altos tenores de este metaloide. El presente trabajotuvo como objetivo establecer si existe correlación entre la exposición a arsénico inorgánico (AsI) a través del consumo de aguas y el nivel de daño al ADN. Muestras de orina y sangre fueron recolectadas de una población de la provincia de Santiago del Estero. Sobre éstas se evaluó el contenido de arsénico urinario (Asu) como marcador de exposición reciente y el daño producidoal ADN mediante el ensayo de electroforesis de células únicas (test del cometa) en sangre como índice de genotoxicidad. De la población total estudiada (n=65), el 41,54% fueron niños. El 57% de la población infantil presentó valores de Asu superiores al valor máximo referencial (hasta 40μgAs/g creatinina) en un rango comprendido entre 41,78 y 3918,10 μgAs/g creatinina. La evaluación genotóxica en sangre reveló que los niños expuestos a elevados niveles de arsénico presentaron un porcentajede células con alto daño al ADN significativamente mayor al compararlo con aquellos con Asu menor al valor de referencia (32,79% ± 3,32 vs 9,77% ± 6,59; p<0,001). A pesar del bajo número de muestras analizadas en este estudio preliminar, existe una buena correlación entre el contenido de arsénico urinario en la población general y el daño al ADN (r=0,6509; p=0,0117). Los resultados obtenidos muestran que la población estudiada expuesta a altos niveles de arsénico, representa un grupo de riesgo para el desarrollo de patologías relacionadas con este tóxico.


Arsenic is an environmental pollutantwidely distributed in the world. In Argentine about a million and a half of inhabitants are exposed to its deleterious effect by drinking water with high levels of this metalloid. In this work the correlation between the exposure to inorganic arsenic and thelevel of DNA damage in blood cells was assessed. Urine and blood samples were taken from a population of Santiago del Estero province. Urinary total arsenic (Asu) content (a recent exposure biomarker) and single cell gel electrophoresis (Comet assay) wereperformed. Children represented the 41.54% of the sampled population (n=65) and 57% of them had Asu levels between 41.78 and 3918.10 μgAs/g creatinine (reference value: less than 40μgAs/g creatinine). Comet assay showed that children with high levelof Asu presented a percentage of cells with high DNA damage increased respect to those with Asu below the reference value (32.79% ± 3.32 vs 9.77% ± 6.59, p< 0.001). Despite the small number of samples analyzed in this preliminary study, there is a clear tendency of correlation between total arsenic content in urine and DNA damage (r=0.6509, p=0.0117). The data obtained showed that the children population exposed to high levels of arsenic represent a group of risk for developing pathologies related with this toxic.


Subject(s)
Humans , Adolescent , Child , Arsenic Poisoning , Arsenic/urine , Arsenic/blood , Water Pollution/analysis , Argentina/epidemiology , DNA Damage , Genotoxicity/adverse effects
18.
Bol. Acad. Nac. Med. B.Aires ; 72(2): 557-64, jul.-dic. 1994. tab
Article in Spanish | LILACS | ID: lil-151798

ABSTRACT

La azidotimidina (AZT) es la primera droga antirretrovira permitida para el tratamiento de la infección con el virus de la inmunodeficiencia humana. En esta investigación se evaluó la actividad genotóxica de la AZT en dos sistemas celulares en cultivo. En linfocitos humanos, la AZT produjo un aumento estadísticamente significativo en las roturas cromosómicas a la dosis de 100 µg/ml y en las células mixonucleadas a la dosis más alta utilizada (500 µg/ml). El intercambio de cromátidas hermanas (ICH) experimentó un significativo incremento a partir de 50 µg/ml. La proliferación celular de los linfocitos mostró un importante enlentecimiento a la concentración de 500 µg/ml. En la Línea de ovario de hamster chino (CHO), AZT produjo un significativo aumento en las aberraciones cromosómicas y en el ICH a las dosis de 1000 y 500 µg/ml, respectivamente. A la concentración de 2500 µg/ml se observó una demora en la progresión celular. Estos resultados sugieren que la droga AZT daña al ADN en los dos sistemas celulares estudiados, siendo los linfocitos humanos más sensibles que la línea CHO. Teniendo en cuenta este efecto genotóxico la droga AZT debe ser administrada con precaución a las poblaciones en riesgo


Subject(s)
Chromosome Aberrations , Culture Techniques , Zidovudine
SELECTION OF CITATIONS
SEARCH DETAIL