Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 12(9)2020 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-32825012

RESUMO

For precision cancer radiotherapy, high linear energy transfer (LET) particle irradiation offers a substantial advantage over photon-based irradiation. In contrast to the sparse deposition of low-density energy by χ- or γ-rays, particle irradiation causes focal DNA damage through high-density energy deposition along the particle tracks. This is characterized by the formation of multiple damage sites, comprising localized clustered patterns of DNA single- and double-strand breaks as well as base damage. These clustered DNA lesions are key determinants of the enhanced relative biological effectiveness (RBE) of energetic nuclei. However, the search for a fingerprint of particle exposure remains open, while the mechanisms underlying the induction of chromothripsis-like chromosomal rearrangements by high-LET radiation (resembling chromothripsis in tumors) await to be elucidated. In this work, we investigate the transformation of clustered DNA lesions into chromosome fragmentation, as indicated by the induction and post-irradiation repair of chromosomal damage under the dynamics of premature chromosome condensation in G0 human lymphocytes. Specifically, this study provides, for the first time, experimental evidence that particle irradiation induces localized shattering of targeted chromosome domains. Yields of chromosome fragments and shattered domains are compared with those generated by γ-rays; and the RBE values obtained are up to 28.6 for α-particles (92 keV/µm), 10.5 for C-ions (295 keV/µm), and 4.9 for protons (28.5 keV/µm). Furthermore, we test the hypothesis that particle radiation-induced persistent clustered DNA lesions and chromatin decompaction at damage sites evolve into localized chromosome shattering by subsequent chromatin condensation in a single catastrophic event-posing a critical risk for random rejoining, chromothripsis, and carcinogenesis. Consistent with this hypothesis, our results highlight the potential use of shattered chromosome domains as a fingerprint of high-LET exposure, while conforming to the new model we propose for the mechanistic origin of chromothripsis-like rearrangements.

2.
DNA Repair (Amst) ; 89: 102828, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32143127

RESUMO

Four repair pathways process DNA double-strand breaks (DSBs). Among these pathways the homologous recombination repair (HRR) subpathway of gene conversion (GC) affords error-free processing, but functions only in S- and G2-phases of the cell cycle. Classical non-homologous end-joining (c-NHEJ) operates throughout the cell cycle, but causes small deletions and translocations. Similar deficiencies in exaggerated form, combined with reduced efficiency, are associated with alternative end-joining (alt-EJ). Finally, single-strand annealing (SSA) causes large deletions and possibly translocations. Thus, processing of a DSB by any pathway, except GC, poses significant risks to the genome, making the mechanisms navigating pathway-engagement critical to genome stability. Logically, the cell ought to attempt engagement of the pathway ensuring preservation of the genome, while accommodating necessities generated by the types of DSBs induced. Thereby, inception of DNA end-resection will be key determinant for GC, SSA and alt-EJ engagement. We reported that during G2-phase, where all pathways are active, GC engages in the processing of almost 50 % of DSBs, at low DSB-loads in the genome, and that this contribution rapidly drops to nearly zero with increasing DSB-loads. At the transition between these two extremes, SSA and alt-EJ compensate, but at extremely high DSB-loads resection-dependent pathways are suppressed and c-NHEJ remains mainly active. We inquired whether in this processing framework all DSBs have similar fates. Here, we analyze in G2-phase the processing of a subset of DSBs defined by their ability to break chromosomes. Our results reveal an absolute requirement for GC in the processing of chromatid breaks at doses in the range of 1 Gy. Defects in c-NHEJ delay significantly the inception of processing by GC, but leave processing kinetics unchanged. These results delineate the essential role of GC in chromatid break repair before mitosis and classify DSBs that underpin this breakage as the exclusive substrate of GC.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , DNA/efeitos da radiação , Fase G2 , Conversão Gênica , Radiação Ionizante , Animais , Quebra Cromossômica , Cricetulus/genética , DNA/metabolismo , Células HCT116 , Humanos , Reparo de DNA por Recombinação
3.
PLoS One ; 14(5): e0216081, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31059552

RESUMO

A sensitive biodosimetry tool is required for rapid individualized dose estimation and risk assessment in the case of radiological or nuclear mass casualty scenarios to prioritize exposed humans for immediate medical countermeasures to reduce radiation related injuries or morbidity risks. Unlike the conventional Dicentric Chromosome Assay (DCA), which takes about 3-4 days for radiation dose estimation, cell fusion mediated Premature Chromosome Condensation (PCC) technique in G0 lymphocytes can be rapidly performed for radiation dose assessment within 6-8 hrs of sample receipt by alleviating the need for ex vivo lymphocyte proliferation for 48 hrs. Despite this advantage, the PCC technique has not yet been fully exploited for radiation biodosimetry. Realizing the advantage of G0 PCC technique that can be instantaneously applied to unstimulated lymphocytes, we evaluated the utility of G0 PCC technique in detecting ionizing radiation (IR) induced stable and unstable chromosomal aberrations for biodosimetry purposes. Our study demonstrates that PCC coupled with mFISH and mBAND techniques can efficiently detect both numerical and structural chromosome aberrations at the intra- and inter-chromosomal levels in unstimulated T- and B-lymphocytes. Collectively, we demonstrate that the G0 PCC technique has the potential for development as a biodosimetry tool for detecting unstable chromosome aberrations (chromosome fragments and dicentric chromosomes) for early radiation dose estimation and stable chromosome exchange events (translocations) for retrospective monitoring of individualized health risks in unstimulated lymphocytes.


Assuntos
Aberrações Cromossômicas/efeitos da radiação , Linfócitos/efeitos da radiação , Radiometria/métodos , Animais , Células CHO/efeitos da radiação , Fusão Celular , Centrômero/efeitos da radiação , Cricetulus , Feminino , Raios gama/efeitos adversos , Humanos , Hibridização in Situ Fluorescente , Masculino , Lesões por Radiação/diagnóstico , Lesões por Radiação/genética , Radiação Ionizante , Estudos Retrospectivos , Cariotipagem Espectral/métodos , Telômero/efeitos da radiação , Raios X/efeitos adversos
4.
EMBO Rep ; 20(3)2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30665945

RESUMO

Genome-wide studies in tumor cells have indicated that chromatin-modifying proteins are commonly mutated in human cancers. The lysine-specific methyltransferase 2C (KMT2C/MLL3) is a putative tumor suppressor in several epithelia and in myeloid cells. Here, we show that downregulation of KMT2C in bladder cancer cells leads to extensive changes in the epigenetic status and the expression of DNA damage response and DNA repair genes. More specifically, cells with low KMT2C activity are deficient in homologous recombination-mediated double-strand break DNA repair. Consequently, these cells suffer from substantially higher endogenous DNA damage and genomic instability. Finally, these cells seem to rely heavily on PARP1/2 for DNA repair, and treatment with the PARP1/2 inhibitor olaparib leads to synthetic lethality, suggesting that cancer cells with low KMT2C expression are attractive targets for therapies with PARP1/2 inhibitors.


Assuntos
Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Neoplasias/enzimologia , Neoplasias/patologia , Animais , Sequência de Bases , Linhagem Celular Tumoral , Dano ao DNA/genética , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Reparo do DNA/genética , Proteínas de Ligação a DNA/genética , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Elementos Facilitadores Genéticos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Instabilidade Genômica/efeitos dos fármacos , Instabilidade Genômica/genética , Recombinação Homóloga/genética , Humanos , Masculino , Camundongos SCID , Neoplasias/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Regiões Promotoras Genéticas/genética
5.
Artigo em Inglês | MEDLINE | ID: mdl-26520366

RESUMO

Ionizing radiation (IR) induces double strand breaks (DSBs) in cellular DNA, which if not repaired correctly can cause chromosome translocations leading to cell death or cancer. Incorrect joining of DNA ends generating chromosome translocations can be catalyzed either by the dominant DNA-PKcs-dependent, classical non-homologous end-joining (c-NHEJ), or by an alternative end-joining (alt-EJ) process, functioning as backup to abrogated c-NHEJ, or homologous recombination repair. Alt-EJ operates with slower kinetics as compared to c-NHEJ and generates larger alterations at the junctions; it is also considered crucial to chromosome translocation-formation. A recent report posits that this view only holds for rodent cells and that in human cells c-NHEJ is the main mechanism of chromosome translocation formation. Since this report uses designer nucleases that induce DSBs with unique characteristics in specific genomic locations and PCR to detect translocations, we revisit the issue using stochastically distributed DSBs induced in the human genome by IR during the G2-phase of the cell cycle. For visualization and analysis of chromosome translocations, which manifest as chromatid translocations in cells irradiated in G2, we employ classical cytogenetics. In wild-type cells, we observe a significant contribution of alt-EJ to translocation formation, as demonstrated by a yield-reduction after treatment with inhibitors of Parp, or of DNA ligases 1 and 3 (Lig1, Lig3). Notably, a nearly fourfold increase in translocation formation is seen in c-NHEJ mutants with defects in DNA ligase 4 (Lig4) that remain largely sensitive to inhibitors of Parp, and of Lig1/Lig3. We conclude that similar to rodent cells, chromosome translocation formation from randomly induced DSBs in human cells largely relies on alt-EJ. We discuss DSB localization in the genome, characteristics of the DSB and the cell cycle as potential causes of the divergent results generated with IR and designer nucleases.


Assuntos
Cromossomos Humanos/efeitos da radiação , Reparo do DNA por Junção de Extremidades , Fase G2/efeitos da radiação , Translocação Genética , Linhagem Celular , Análise Citogenética , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , DNA Ligase Dependente de ATP , DNA Ligases/antagonistas & inibidores , DNA Ligases/genética , DNA Ligases/metabolismo , Fase G2/efeitos dos fármacos , Células HCT116 , Humanos , Poli(ADP-Ribose) Polimerase-1 , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Pirimidinas/farmacologia , Bases de Schiff/farmacologia , Translocação Genética/efeitos dos fármacos
6.
Cancer Lett ; 356(1): 34-42, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24333869

RESUMO

Radiation-induced bystander effects (RIBE), demonstrate the induction of biological non-targeted effects in cells which have not directly hit by radiation or by free radicals produced by ionization events. Although RIBE have been demonstrated using a variety of biological endpoints the mechanism(s) of this phenomenon still remain unclear. The controversial results of the in vitro RIBE and the evidence of non-targeted effects in various in vivo systems are discussed. The experimental evidence on RIBE, indicate that a more analytical and mechanistic in depth approach is needed to secure an answer to one of the most intriguing questions in radiobiology.


Assuntos
Efeito Espectador/efeitos da radiação , Citocinas/metabolismo , Dano ao DNA/efeitos da radiação , DNA/efeitos da radiação , Lesões por Radiação , Animais , Apoptose/efeitos da radiação , Arabidopsis/efeitos da radiação , Cricetinae , Citocinas/biossíntese , Instabilidade Genômica/efeitos da radiação , Humanos , Camundongos , Oncorhynchus mykiss , Lesões por Radiação/genética , Lesões por Radiação/metabolismo , Lesões por Radiação/patologia , Radiobiologia/tendências , Ratos , Transdução de Sinais , Peixe-Zebra
7.
Nucleic Acids Res ; 42(10): 6380-92, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24748665

RESUMO

In mammalian cells, ionizing radiation (IR)-induced DNA double-strand breaks (DSBs) are repaired in all phases of the cell cycle predominantly by classical, DNA-PK-dependent nonhomologous end joining (D-NHEJ). Homologous recombination repair (HRR) is functional during the S- and G2-phases, when a sister chromatid becomes available. An error-prone, alternative form of end joining, operating as backup (B-NHEJ) functions robustly throughout the cell cycle and particularly in the G2-phase and is thought to backup predominantly D-NHEJ. Parp-1, DNA-ligases 1 (Lig1) and 3 (Lig3), and Xrcc1 are implicated in B-NHEJ. Chromosome and chromatid translocations are manifestations of erroneous DSB repair and are crucial culprits in malignant transformation and IR-induced cell lethality. We analyzed shifts in translocation formation deriving from defects in D-NHEJ or HRR in cells irradiated in the G2-phase and identify B-NHEJ as the main DSB repair pathway backing up both of these defects at the cost of a large increase in translocation formation. Our results identify Parp-1 and Lig1 and 3 as factors involved in translocation formation and show that Xrcc1 reinforces the function of Lig3 in the process without being required for it. Finally, we demonstrate intriguing connections between B-NHEJ and DNA end resection in translocation formation and show that, as for D-NHEJ and HRR, the function of B-NHEJ facilitates the recovery from the G2-checkpoint. These observations advance our understanding of chromosome aberration formation and have implications for the mechanism of action of Parp inhibitors.


Assuntos
Reparo do DNA por Junção de Extremidades , DNA Ligases/fisiologia , Proteínas de Ligação a DNA/fisiologia , Poli(ADP-Ribose) Polimerases/fisiologia , Translocação Genética , Animais , Células Cultivadas , Cricetinae , Quebras de DNA de Cadeia Dupla , DNA Ligase Dependente de ATP , Fase G2/genética , Fase G2/efeitos da radiação , Pontos de Checagem da Fase G2 do Ciclo Celular , Humanos , Camundongos , Poli(ADP-Ribose) Polimerase-1 , Proteínas de Ligação a Poli-ADP-Ribose , Radiação Ionizante , Reparo de DNA por Recombinação , Proteína 1 Complementadora Cruzada de Reparo de Raio-X , Proteínas de Xenopus
8.
PLoS One ; 9(2): e88879, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586425

RESUMO

The etiology of acute myeloid leukemia (AML) underlies the influence of genetic variants in candidate genes. The CYP2B6 enzyme detoxifies many genotoxic xenobiotics, protecting cells from oxidative damage. The CYP2B6 gene is subjected to a single-nucleotide polymorphism (G5¹6T) with heterozygotes (GT) and homozygotes (TT) presenting decreased enzymatic activity. This case-control study aimed to investigate the association of CYP2B6 G5¹6T polymorphism with the susceptibility of AML and its cytogenetic and clinical characteristics. Genotyping was performed on 619 AML patients and 430 healthy individuals using RCR-RFLP and a novel LightSNip assay. The major finding was a statistically higher frequency of the variant genotypes (GT and TT) in patients compared to the controls (GT:38.8% vs 29.8% and TT:9.3% vs 5.3% respectively) (p<0.001). More specifically, a significantly higher frequency of GT+TT genotypes in de novo AML patients (46.6%) and an immensely high frequency of TT in secondary AML (s-AML) (20.5%) were observed. The statistical analysis showed that the variant T allele was approximately 1.5-fold and 2.4-fold higher in de novo and s-AML respectively than controls. Concerning FAB subtypes, the T allele presented an almost 2-fold increased in AML-M2. Interestingly, a higher incidence of the TT genotype was observed in patients with abnormal karyotypes. In particular, positive correlations of the mutant allele were found in patients carrying specific chromosomal aberrations [-7/del(7q), -5/del(5q), +8, +21 or t(8;21)], complex or monosomal karyotypes. Finally, a strikingly higher frequency of TT genotype was also observed in patients stratified to the poor risk group. In conclusion, our results provide evidence for the involvement of the CYP2B6 polymorphism in AML susceptibility and suggest a possible role of the CYP2B6 genetic background on the development of specific chromosomal aberrations.


Assuntos
Transtornos Cromossômicos/genética , Citocromo P-450 CYP2B6/genética , Predisposição Genética para Doença/genética , Mutação em Linhagem Germinativa/genética , Leucemia Mieloide Aguda/genética , Polimorfismo Genético/genética , Alelos , Estudos de Casos e Controles , Aberrações Cromossômicas , Genótipo , Humanos , Cariótipo , Cariotipagem/métodos , Risco
9.
Leuk Lymphoma ; 54(8): 1756-61, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23278642

RESUMO

Models for the pathogenesis of myelodysplastic syndrome (MDS) imply the role of individual genetic variations in genes involved in detoxification mechanisms. GSTP1 enzyme plays a key role in the biotransformation of a variety of carcinogens. The corresponding gene is subject to a single nucleotide polymorphism (A(313)G) leading to abolished enzyme activity. In order to evaluate whether the GSTP1 polymorphism influences MDS susceptibility, we conducted a case-control study comprising 310 de novo patients and 370 healthy controls using a real-time polymerase chain reaction (PCR) genotyping method. The GSTP1 gene status was also evaluated in relation to patients' characteristics and chromosomal abnormalities. A significantly higher incidence of the GSTP1 variant genotypes was observed in patients with MDS compared to controls (p < 0.0001). The results revealed increased frequencies of heterozygotes in patients younger than 60 years old and of homozygotes G/G in older patients (p = 0.007). Our results provide evidence for a pathogenetic role of the GSTP1 polymorphism in MDS risk, probably in an age-dependent manner.


Assuntos
Predisposição Genética para Doença , Glutationa S-Transferase pi/genética , Síndromes Mielodisplásicas/genética , Polimorfismo de Nucleotídeo Único , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Substituição de Aminoácidos , Estudos de Casos e Controles , Aberrações Cromossômicas , Feminino , Frequência do Gene , Genótipo , Mutação em Linhagem Germinativa , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
10.
Med Hypotheses ; 80(1): 70-4, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23111201

RESUMO

Cancer development is an evolutionary process that has been highly conserved among centuries within organisms. Based on this, the interest in cancer research focuses on cells, organelles and genes that possess a genetic conservatism from yeasts to human. Towards this thought, mitochondria, the highly conserved and responsible for the cellular bioenergetic activity organelles, might play crucial role in carcinogenesis. Interestingly, tumors with low bioenergetic signature have worse prognosis and show a decreased expression of ATPase protein. Furthermore, according to the stem-cell theory of carcinogenesis, aggressive tumors are characterized by an increase number of malignant stem-like cell population and their resistance to chemotherapy has been found to be mitochondrially driven. The above considerations triggered us to hypothesize that mitochondrial bioenergetic processes in stem-like cancer cells plays a crucial role in the highly conserved process of carcinogenesis. Specifically, we support that mitochondrial and/or nuclear DNA alterations that control stem cells' ATP production drive stem cells to "immortalization" (Otto Warburg theory) that mediates cancer initiation and progression. Substantiation of our hypothesis requires evidence that: (1) alterations in mitochondria bioenergetic metabolites and enzymes encoded either from the mtDNA or the nuclear DNA are linked to human cancer and (2) mitochondrial functions are regulated by highly conserved genes involved in cancer-related cellular processes such as apoptosis, aging and autophagy. Experimental approach on how this hypothesis might be tested and promising strategies in cancer therapeutics are also discussed. In case the hypothesis of stem-cell bioenergetic malformations' related carcinogenesis proves to be correct, it would contribute to the development of new prognostic, diagnostic and even more effective therapeutic interventions against various types of cancer.


Assuntos
Trifosfato de Adenosina/metabolismo , Transformação Celular Neoplásica/metabolismo , Metabolismo Energético/fisiologia , Mitocôndrias/fisiologia , Neoplasias/fisiopatologia , Células-Tronco Neoplásicas/fisiologia , Apoptose/fisiologia , Autofagia/fisiologia , Humanos , Modelos Biológicos
11.
Mol Biol Rep ; 39(1): 251-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21556769

RESUMO

Glutaraldehyde (GA) is a high production volume chemical that is very reactive with a wide spectrum of medical, scientific and industrial applications. Concerning the genotoxic and carcinogenic effect of GA, controversial results have been reported, while in humans no studies with positive carcinogenic results for GA have been published. However, our previous study concerning the combined effects of exposure to both GA and ionising radiation (IR) in peripheral blood lymphocytes of healthy donors has shown that non-genotoxic doses of the chemical induces a statistically significant increase in chromosomal radiosensitivity. The lack of information concerning the radiosensitizing potential of GA on cancerous cells triggered us to test the radiosensitizing effect of GA on breast cancer cells (MCF7). For this purpose the G2-chromosomal radiosensitivity assay (G2-assay) was used. The assay involves G2-phase irradiation and quantitation of the chromosomal fragility in the subsequent metaphase. The experimental data show that 48 h exposure to GA, at doses that are not clastogenic to MCF7 breast cancer cells enhances G2-chromosomal radiosensitivity of this cell line. In an effort to evaluate whether the observed increase in GAs-induced G2-chromosomal radiosensitization is linked to GA-induced alterations in the cell cycle and feedback control mechanism, Mitotic Index analysis was performed. The results have shown that such a mechanism cannot be directly related to the observed GA-induced increase in G2-chromosomal radiosensitivity. Since increased G2-chromosomal radiosensitivity has been linked with cancer proneness, the radiosensitizing effect of GA at non-clastogenic doses highlights its potential carcinogenic profile.


Assuntos
Cromossomos Humanos/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Glutaral/farmacologia , Radiossensibilizantes/farmacologia , Bromodesoxiuridina , Linhagem Celular Tumoral , Cromossomos Humanos/efeitos da radiação , Feminino , Fase G2/efeitos da radiação , Humanos , Cariotipagem , Índice Mitótico , Troca de Cromátide Irmã , Estatísticas não Paramétricas
12.
Radiother Oncol ; 101(1): 28-34, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22014898

RESUMO

BACKGROUND AND PURPOSE: An increased yield of chromatid breaks following G2-phase irradiation could be a marker of radiosensitivity-predisposing genes that respond to DNA damage. We have shown that the dynamic nature of chromatin-nucleoprotein complex, which is capable of rapid unfolding, disassembling, assembling and refolding, affects repair of radiation-induced DNA-lesions and causes chromatid breaks during G2-M transition in damaged DNA sites. Here, we investigate induction and repair kinetics of chromatid breaks, their potential role in radiosensitivity predisposition and a standardized G2-assay is proposed to assess individual radiosensitivity. MATERIALS AND METHODS: Lymphocytes from 125 blood donors with significant inter-individual radiosensitivity variation (healthy, cancer, AT-patients) are used to correlate G2-checkpoint efficiency with chromatid breakage and individual radiosensitivity. Experiments involve repair kinetics of chromatid breaks using colcemid-block and treatment with caffeine to abrogate G2-checkpoint, generate internal controls and standardize the G2-assay. RESULTS: Radiation-induced chromatid breaks during G2-M transition, following 4h repair, remained unchanged and a significant correlation between G2-chromosomal radiosensitivity and G2-checkpoint efficiency to prevent chromatid breakage was found. A standardized G2-assay is developed by introducing normalization to conditions reflecting lack of checkpoint and repair similar to those of AT-patients, generating a unique standard for individual radiosensitivity testing. CONCLUSIONS: The standardized G2-assay can minimize inter-laboratory and intra-experimental variations and may have straightforward application in clinical practice for individualization of radiotherapy protocols.


Assuntos
Pontos de Checagem da Fase G2 do Ciclo Celular , Linfócitos/efeitos da radiação , Neoplasias/genética , Neoplasias/radioterapia , Tolerância a Radiação/genética , Estudos de Casos e Controles , Aberrações Cromossômicas , Dano ao DNA/efeitos da radiação , Reparo do DNA/genética , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Humanos , Masculino , Neoplasias/patologia , Valor Preditivo dos Testes , Doses de Radiação , Tolerância a Radiação/efeitos da radiação , Valores de Referência , Estudos de Amostragem
13.
Free Radic Biol Med ; 51(3): 713-8, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21605663

RESUMO

Oxidative stress plays a pivotal role in the pathogenesis of multiple sclerosis (MS). Inactivating polymorphisms of genes encoding detoxification enzymes, such as NAD(P)H:quinone oxidoreductase 1 (NQO1), could influence susceptibility to MS. To test this hypothesis we performed a case-control study in which we compared the distribution of NQO1 genotypes between 231 MS patients and 380 controls, using both PCR-RFLP and real-time PCR assays. Correlations with MS clinical subtype classification and gender were also evaluated. A significantly higher frequency of the homozygous (T/T) and heterozygous (C/T) NQO1 C(609)T variant genotypes was observed among MS patients compared to controls (P=0.01), with MS patients showing a 1.5-fold increased risk of carrying at least one variant T allele (P=0.009). Interestingly, patients belonging to the primary progressive subgroup exhibited a significantly higher incidence of the heterozygous C/T variant genotype, compared to the other forms of MS (P=0.019). There was no correlation of the NQO1 polymorphism with gender. These results provide the first evidence for a pathogenetic role for the NQO1 C(609)T polymorphism in MS susceptibility and suggest a possible role for the NQO1 genetic background in the development of primary progressive MS.


Assuntos
Esclerose Múltipla Crônica Progressiva/genética , Esclerose Múltipla Crônica Progressiva/fisiopatologia , NAD(P)H Desidrogenase (Quinona)/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Análise Mutacional de DNA , Progressão da Doença , Feminino , Frequência do Gene , Estudos de Associação Genética , Predisposição Genética para Doença , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla Crônica Progressiva/enzimologia , Mutação/genética , NAD(P)H Desidrogenase (Quinona)/genética , Estresse Oxidativo , Polimorfismo Genético , Risco , Fatores Sexuais
14.
Mutat Res ; 711(1-2): 174-86, 2011 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-21185845

RESUMO

The formation of diverse chromosomal aberrations following irradiation and the variability in radiosensitivity at different cell-cycle stages remain a long standing controversy, probably because most of the studies have focused on elucidating the enzymatic mechanisms involved using simple DNA substrates. Yet, recognition, processing and repair of DNA damage occur within the nucleoprotein complex of chromatin which is dynamic in nature, capable of rapid unfolding, disassembling, assembling and refolding. The present work reviews experimental work designed to investigate the impact of chromatin dynamics and chromosome conformation changes during cell-cycle in the formation of chromosomal aberrations. Using conventional cytogenetics and premature chromosome condensation to visualize interphase chromatin, the data presented support the hypothesis that chromatin dynamic changes during cell-cycle are important determinants in the conversion of sub-microscopic DNA lesions into chromatid breaks. Consequently, the type and yield of radiation-induced chromosomal aberrations at a given cell-cycle-stage depends on the combined effect of DNA repair processes and chromatin dynamics, which is cell-cycle-regulated and subject to up- or down-regulation following radiation exposure or genetic alterations. This new hypothesis is used to explain the variability in radiosensitivity observed at various cell-cycle-stages, among mutant cells and cells of different origin, or among different individuals, and to revisit unresolved issues and unanswered questions. In addition, it is used to better understand hypersensitivity of AT cells and to provide an improved predictive G2-assay for evaluating radiosensitivity at individual level. Finally, experimental data at single cell level obtained using hybrid cells suggest that the proposed hypothesis applies only to the irradiated component of the hybrid.


Assuntos
Ciclo Celular/genética , Cromátides/genética , Cromatina/fisiologia , Aberrações Cromossômicas , Dano ao DNA , Animais , Ciclo Celular/efeitos da radiação , Cromátides/efeitos da radiação , Cromatina/efeitos da radiação , Dano ao DNA/efeitos da radiação , Fase G2 , Humanos , Modelos Genéticos
15.
Mutat Res ; 701(1): 27-37, 2010 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-20398788

RESUMO

Experiments were carried out to explore the correlation between chromatin conformation changes in the presence of DNA lesions and the formation of radiation-induced chromosomal aberrations. To modulate the onset and dynamics of chromatin conformation changes following irradiation, premature chromosome condensation (PCC) was induced by means of cell fusion. G2-check point abrogation by caffeine or elevated heat treatment was also applied. In addition, transfer of irradiated mitotic cells was employed either into depleted media to restrain them from proceeding through G1/S, or holding them further in colcemid to avoid M/G1 transition. To investigate the correlation between efficiency of chromosomal conformation changes and chromosomal breakage in irradiated G0 peripheral blood lymphocytes, cell fusion with different mitotic PCC-inducer cells was used. The experimental evidence supports the hypothesis that functional cell-cycle chromatin conformation changes in the presence of DNA damage are important determinants in the formation of radiation-induced chromosomal aberrations. Specifically, it is proposed here that following irradiation, chromatin structure may not be broken but instead it unfolds to a conformation that is more accessible to repair enzymes at the sites of DNA lesions. If subsequent chromosomal conformation changes occur while DNA is still being repaired, such changes will lead into an energetically unfavorable state, thus exerting mechanical stress on the unfolded chromatin at the damaged sites, which will in turn result into chromatid breaks that may not be able to restitute or mis-rejoin. Therefore, this biophysical conversion process of DNA damage into chromatid breaks as such is antagonistic to the DNA repair process. Alternatively, in the absence of chromosomal conformation changes, either DNA repair will take place efficiently or DNA misrepair will cause the formation of exchanges and chromosomal rearrangements. Consequently, the type and yield of radiation-induced chromosomal aberrations at a given cell cycle stage will be the combined effect of the interaction, at that particular stage, of the DNA repair process and the proposed conversion process of DNA lesions into chromatid breaks.


Assuntos
Cromatina/química , Aberrações Cromossômicas , Dano ao DNA , Radiação Ionizante , Animais , Células CHO , Cricetinae , Cricetulus , Quebras de DNA de Cadeia Dupla , Humanos , Interfase , Modelos Genéticos , Conformação Molecular , Fosforilação , Radiogenética
16.
Chem Biol Interact ; 188(2): 350-8, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20371364

RESUMO

Oxidatively-induced clustered DNA lesions are considered the signature of any ionizing radiation like the ones human beings are exposed daily from various environmental sources (medical X-rays, radon, etc.). To evaluate the role of BRCA1 deficiencies in the mitigation of radiation-induced toxicity and chromosomal instability we have used two human breast cancer cell lines, the BRCA1 deficient HCC1937 cells and as a control the BRCA1 wild-type MCF-7 cells. As an additional control for the DNA damage repair measurements, the HCC1937 cells with partially reconstituted BRCA1 expression were used. Since clustered DNA damage is considered the signature of ionizing radiation, we have measured the repair of double strand breaks (DSBs), non-DSB bistranded oxidative clustered DNA lesions (OCDLs) as well as single strand breaks (SSBs) in cells exposed to radiotherapy-relevant γ-ray doses. Parallel measurements were performed in the accumulation of chromatid and isochromatid breaks. For the measurement of OCDL repair, we have used a novel adaptation of the denaturing single cell gel electrophoresis (Comet assay) and pulsed field gel electrophoresis with Escherichia coli repair enzymes as DNA damage probes. Independent monitoring of the γ-H2AX foci was also performed while metaphase chromatid lesions were measured as an indicator of chromosomal instability. HCC1937 cells showed a significant accumulation of all types of DNA damage and chromatid breaks compared to MCF-7 while BRCA1 partial expression contributed significantly in the overall repair of OCDLs. These results further support the biological significance of repair resistant clustered DNA damage leading to chromosomal instability. The current results combined with previous findings on the minimized ability of base clusters to induce cell death (mainly induced by DSBs), enhance the potential association of OCDLs with breast cancer development especially in the case of a BRCA1 deficiency leading to the survival of breast cells carrying a high load of unrepaired DNA damage clusters.


Assuntos
Proteína BRCA1/metabolismo , Neoplasias da Mama/metabolismo , Instabilidade Cromossômica/efeitos da radiação , Dano ao DNA/efeitos da radiação , Reparo do DNA , Proteína BRCA1/genética , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Histonas/metabolismo , Humanos
17.
Int J Oncol ; 35(5): 1223-30, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19787278

RESUMO

Increased yield of chromatid breaks, following in vitro G2-phase lymphocyte irradiation, can be a marker of individual radiosensitivity and cancer predisposing genes whose role is to respond to DNA damage. Mutations or polymorphisms of genes encoding DNA repair pathways may underlie the increased chromosomal radiosensitivity. However, genes that facilitate DNA damage recognition, using signal transduction pathways to activate cell cycle arrest and preserve genomic integrity, are perhaps the most important determinant. Based on the latter hypothesis, an individual radiosensitivity parameter (IRP) is introduced, which expresses, at individual level, the G2-checkpoint potential to facilitate DNA damage recognition and repair of radiation-induced chromosomal damage during G2 to M-phase transition. Based on this parameter a new methodology for assessment of individual radiosensitivity is proposed, which involves G2-checkpoint abrogation by caffeine to obtain the IRP values. To evaluate the proposed methodology, blood samples from 52 healthy donors were taken for inter-individual radiosensitivity analysis using both the conventional G2 chromosomal radiosensitivity assay as well as the new approach using caffeine-induced G2-checkpoint abrogation. The two assays were compared in experiments using samples from 5 hypersensitive patients, 3 AT-homozygotes, 3 AT-heterozygotes, and the GM15786, GM03188A, GM09899, HCC1937 and MCF-7 cell lines. Using the G2 chromosomal radiosensitivity assay, donors are predicted as G2 radiosensitive or normal, while according to the new approach, individuals can be classified as highly radiosensitive, radiosensitive, normal, radioresistant and highly radioresistant. Overall, the new approach provides better individual radiosensitivity discrimination and intra-experimental reproducibility. Therefore, the proposed methodology using IRP values may provide a clinically applicable predictive assay for individual radiosensitivity and predisposition to cancer.


Assuntos
Fase G2/efeitos da radiação , Predisposição Genética para Doença , Técnicas Genéticas , Linfócitos/efeitos da radiação , Neoplasias/genética , Tolerância a Radiação/genética , Dano ao DNA/efeitos da radiação , Fase G2/genética , Testes Hematológicos , Humanos , Reprodutibilidade dos Testes
19.
Cancer Genet Cytogenet ; 187(2): 85-94, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19027489

RESUMO

Jumping translocations (JT) are rare cytogenetic aberrations in hematological malignancies that include unbalanced translocations involving a donor chromosome arm or chromosome segment that has fused to two or more different recipient chromosomes in different cell lines. We report five cases associated with different hematologic disorders and JT to contribute to the investigation of the origin, pathogenesis, and clinical significance of JT. These cases involve JT of 1q in a case of acute myeloblastic leukemia (AML)-M1, a case of Burkitt lymphoma, and a case of BCR/ABL-positive acute lymphoblastic leukemia, as well as a JT of 13q in a case of AML-M5, and a JT of 11q segment in a case of undifferentiated leukemia. To our knowledge, with regard to hematologic malignancies, this study presents the first case of JT associated with AML-M1, the first case of JT involving 13q as a donor chromosome, and the first report of JT involving a segment of 11q containing two copies of the MLL gene, jumping on to two recipient chromosomes in each cell line and resulting in six copies of the MLL gene. Our investigation suggests that JT may not contribute to the pathogenesis but rather to the progression of the disease, and it demonstrates that chromosome band 1q10 as a breakpoint of the donor chromosome 1q is also implicated in AML, not only in multiple myeloma as it has been known until now.


Assuntos
Neoplasias Hematológicas/genética , Translocação Genética , Adulto , Idoso , Linfoma de Burkitt/diagnóstico , Linfoma de Burkitt/genética , Análise Citogenética , Feminino , Humanos , Cariotipagem , Leucemia/diagnóstico , Leucemia/genética , Leucemia Monocítica Aguda/diagnóstico , Leucemia Monocítica Aguda/genética , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Masculino , Pessoa de Meia-Idade , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Adulto Jovem
20.
Int J Oncol ; 33(4): 871-9, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18813802

RESUMO

Cells of higher eukaryotes process double strand breaks (DSBs) in their genome using a non-homologous end joining apparatus that utilizes DNA-PK and other well characterized factors (D-NHEJ). Cells with defects in D-NHEJ, repair the majority of DSBs using a slow-repair pathway which is independent of genes of the RAD52 epistasis group and functions as a backup (B-NHEJ). Recent studies implicate DNA ligase III, PARP-1 and histone H1 in this pathway of NHEJ. The present study investigates the operation of B-NHEJ in the repair of interphase chromosome breaks visualized in irradiated G0 human lymphocytes by premature chromosome condensation (PCC). Chromosome breaks are effectively repaired in human lymphocytes, but repair is significantly compromised after treatment with wortmannin, a DNA-PK inhibitor. Despite slower kinetics, cells exposed to wortmannin rejoin the majority of IR induced chromosome breaks suggesting that B-NHEJ is also functional at the chromosome level. Complementation of D-NHEJ defect in wortmannin-treated lymphocytes by newly made DNA-PK is only possible under conditions of nuclear envelope break down and premature chromosome condensation, suggesting that in interphase cells the shunting of chromosome breaks from D-NHEJ to B-NHEJ is irreversible. The understanding of chromosomal aberration formation allows mechanistic explanations for the carcinogenic potential of D-NHEJ defects.


Assuntos
Cromossomos/ultraestrutura , Dano ao DNA , Proteína Quinase Ativada por DNA/metabolismo , Epistasia Genética , Androstadienos/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Reparo do DNA , Histonas/metabolismo , Humanos , Cinética , Linfócitos/metabolismo , Modelos Biológicos , Wortmanina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...