Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
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.
Eur J Hum Genet ; 27(4): 603-611, 2019 04.
Article in English | MEDLINE | ID: mdl-30626931

ABSTRACT

Among other applications of long-distance haplotype phasing in clinical genetics, determination of linked DNA markers as surrogate for problematic structural variants (e.g., repeat-mediated rearrangements) is essential to perform diagnosis from low-quality DNA samples. We describe a next-of-kin-independent (physical) phasing approach based on inverse-PCR (iPCR) paired-end amplification (PI). This method enables typing the multialleles of the short tandem repeat (STR) F8Int21[CA]n at the F8-intron 21, as a surrogate DNA marker for the F8-intron 22 inversion (Inv22), the hemophilia A-causative hotspot, within the transmitted haplotype in informative carriers. We provide proof-of-concept by blindly validating the PI approach in 15 carrier mother/affected-son duos. Every F8Int21[CA]n STR allele determined in phase with the Inv22 allele in the female carriers from the informative duos was confirmed in the hemizygous proband (P = 0.00003). A second surrogate STR locus at the F8-IVS22 was obtained by the PI approach improving severe-HA preimplantation genetic diagnosis by augmenting heterozygosity in Inv22 carriers bypassing the requirement for family linkage analysis. The ability of the PI-assay to combine other marker pairs was demonstrated by haplotyping a SNV (F8:c.6118T > C) with a >28kb-distant F8-IVS22 STR. The PI approach has proven flexibility to target different marker pairs and has potential for multiplex characterization of iPCR products by massively parallel sequencing.


Subject(s)
Hemophilia A/genetics , Microsatellite Repeats/genetics , Polymerase Chain Reaction/methods , Preimplantation Diagnosis , Alleles , Female , Genetic Markers/genetics , Genotype , Haplotypes/genetics , Hemophilia A/diagnosis , Hemophilia A/physiopathology , Humans , Introns , Male , Pregnancy
4.
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
5.
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
6.
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
7.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...