Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Nucleic Acids Res ; 44(4): 1732-45, 2016 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-26712563

RESUMO

Multiple DNA double-strand break (DSB) repair pathways are active in S phase of the cell cycle; however, DSBs are primarily repaired by homologous recombination (HR) in this cell cycle phase. As the non-homologous end-joining (NHEJ) factor, Ku70/80 (Ku), is quickly recruited to DSBs in S phase, we hypothesized that an orchestrated mechanism modulates pathway choice between HR and NHEJ via displacement of the Ku heterodimer from DSBs to allow HR. Here, we provide evidence that phosphorylation at a cluster of sites in the junction of the pillar and bridge regions of Ku70 mediates the dissociation of Ku from DSBs. Mimicking phosphorylation at these sites reduces Ku's affinity for DSB ends, suggesting that phosphorylation of Ku70 induces a conformational change responsible for the dissociation of the Ku heterodimer from DNA ends. Ablating phosphorylation of Ku70 leads to the sustained retention of Ku at DSBs, resulting in a significant decrease in DNA end resection and HR, specifically in S phase. This decrease in HR is specific as these phosphorylation sites are not required for NHEJ. Our results demonstrate that the phosphorylation-mediated dissociation of Ku70/80 from DSBs frees DNA ends, allowing the initiation of HR in S phase and providing a mechanism of DSB repair pathway choice in mammalian cells.


Assuntos
Antígenos Nucleares/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/genética , Proteínas de Ligação a DNA/genética , Fase S/genética , Animais , Dano ao DNA/genética , Reparo do DNA/genética , Fibroblastos/metabolismo , Células HCT116 , Recombinação Homóloga , Humanos , Autoantígeno Ku , Camundongos , Transdução de Sinais
2.
Int J Mol Sci ; 19(8)2018 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-30110966

RESUMO

In recent years several approaches have been developed to address the chromatin status and its changes in eukaryotic cells under different conditions-but only few are applicable in living cells. Fluorescence lifetime imaging microscopy (FLIM) is a functional tool that can be used for the inspection of the molecular environment of fluorophores in living cells. Here, we present the use of single organic minor groove DNA binder dyes in FLIM for measuring chromatin changes following modulation of chromatin structure in living cells. Treatment with histone deacetylase inhibitors led to an increased fluorescence lifetime indicating global chromatin decompaction, whereas hyperosmolarity decreased the lifetime of the used dyes, thus reflecting the expected compaction. In addition, we demonstrate that time domain FLIM data based on single photon counting should be optimized using pile-up and counting loss correction, which affect the readout even at moderate average detector count rates in inhomogeneous samples. Using these corrections and utilizing Hoechst 34580 as chromatin compaction probe, we measured a pan nuclear increase in the lifetime following irradiation with X-rays in living NIH/3T3 cells thus providing a method to measure radiation-induced chromatin decompaction.


Assuntos
Benzimidazóis/farmacologia , Montagem e Desmontagem da Cromatina/efeitos da radiação , DNA/metabolismo , Corantes Fluorescentes/farmacologia , Raios X , Animais , Camundongos , Microscopia de Fluorescência , Células NIH 3T3
3.
Nucleic Acids Res ; 43(17): 8352-67, 2015 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-26240375

RESUMO

The MRE11/RAD50/NBS1 (MRN) complex plays a central role as a sensor of DNA double strand breaks (DSB) and is responsible for the efficient activation of ataxia-telangiectasia mutated (ATM) kinase. Once activated ATM in turn phosphorylates RAD50 and NBS1, important for cell cycle control, DNA repair and cell survival. We report here that MRE11 is also phosphorylated by ATM at S676 and S678 in response to agents that induce DNA DSB, is dependent on the presence of NBS1, and does not affect the association of members of the complex or ATM activation. A phosphosite mutant (MRE11S676AS678A) cell line showed decreased cell survival and increased chromosomal aberrations after radiation exposure indicating a defect in DNA repair. Use of GFP-based DNA repair reporter substrates in MRE11S676AS678A cells revealed a defect in homology directed repair (HDR) but single strand annealing was not affected. More detailed investigation revealed that MRE11S676AS678A cells resected DNA ends to a greater extent at sites undergoing HDR. Furthermore, while ATM-dependent phosphorylation of Kap1 and SMC1 was normal in MRE11S676AS678A cells, there was no phosphorylation of Exonuclease 1 consistent with the defect in HDR. These results describe a novel role for ATM-dependent phosphorylation of MRE11 in limiting the extent of resection mediated through Exonuclease 1.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas de Ligação a DNA/metabolismo , Exodesoxirribonucleases/metabolismo , Reparo de DNA por Recombinação , Transdução de Sinais , Linhagem Celular , Linhagem Celular Tumoral , Dano ao DNA , Proteínas de Ligação a DNA/química , Humanos , Fosforilação , Radiação Ionizante
4.
EMBO J ; 30(6): 1079-92, 2011 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-21317870

RESUMO

DNA non-homologous end joining (NHEJ) and homologous recombination (HR) function to repair DNA double-strand breaks (DSBs) in G2 phase with HR preferentially repairing heterochromatin-associated DSBs (HC-DSBs). Here, we examine the regulation of repair pathway usage at two-ended DSBs in G2. We identify the speed of DSB repair as a major component influencing repair pathway usage showing that DNA damage and chromatin complexity are factors influencing DSB repair rate and pathway choice. Loss of NHEJ proteins also slows DSB repair allowing increased resection. However, expression of an autophosphorylation-defective DNA-PKcs mutant, which binds DSBs but precludes the completion of NHEJ, dramatically reduces DSB end resection at all DSBs. In contrast, loss of HR does not impair repair by NHEJ although CtIP-dependent end resection precludes NHEJ usage. We propose that NHEJ initially attempts to repair DSBs and, if rapid rejoining does not ensue, then resection occurs promoting repair by HR. Finally, we identify novel roles for ATM in regulating DSB end resection; an indirect role in promoting KAP-1-dependent chromatin relaxation and a direct role in phosphorylating and activating CtIP.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Fase G2 , Linhagem Celular , Heterocromatina/metabolismo , Humanos , Cinética , Redes e Vias Metabólicas , Recombinação Genética
5.
FASEB J ; 28(3): 1412-21, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24347608

RESUMO

Carbon ion irradiation is an emerging therapeutic option for various tumor entities. Radiation resistance of solid tumors toward photon irradiation is caused by attenuation of DNA damage in less oxygenated tumor areas and by increased hypoxia-inducible factor (HIF)-1 signaling. Carbon ion irradiation acts independently of oxygen; however, the role of HIF-1 is unclear. We analyzed the effect of HIF-1 signaling after carbon ions in comparison to photons by using biological equivalent radiation doses in a human non-small-cell cancer model. The studies were performed in cultured A549 and H1299 cell lines and in A549 xenografts. Knockdown of HIF-1α in vivo combined with photon irradiation delayed tumor growth (23 vs. 13 d; P<0.05). Photon irradiation induced HIF-1α and target genes, predominantly in oxygenated cells (1.6-fold; P<0.05), with subsequent enhanced tumor angiogenesis (1.7-fold; P<0.05). These effects were not observed after carbon ion irradiation. Micro-DNA array analysis indicated that photons, but not carbon ions, significantly induced components of the mTOR (mammalian target of rapamycin) pathway (gene set enrichment analysis; P<0.01) as relevant for HIF-1α induction. After carbon ion irradiation in vivo, we observed substantially decreased HIF-1α levels (8.9-fold; P<0.01) and drastically delayed tumor growth (P<0.01), an important finding that indicates a higher relative biological effectiveness (RBE) than anticipated from the cell survival data. Taken together, the evidence showed that carbon ions mediate an improved therapeutic effectiveness without tumor-promoting HIF-1 signaling.


Assuntos
Radioisótopos de Carbono/uso terapêutico , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Neoplasias Pulmonares/radioterapia , Animais , Sequência de Bases , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Primers do DNA , Regulação para Baixo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase
6.
Nucleic Acids Res ; 41(12): 6109-18, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23620287

RESUMO

DNA double-strand breaks (DSB) are considered as the most deleterious DNA lesions, and their repair is further complicated by increasing damage complexity. However, the molecular effects of clustered lesions are yet not fully understood. As the locally restricted phosphorylation of H2AX to form γH2AX is a key step in facilitating efficient DSB repair, we investigated this process after localized induction of clustered damage by ionizing radiation. We show that in addition to foci at damaged sites, H2AX is also phosphorylated in undamaged chromatin over the whole-cell nucleus in human and rodent cells, but this is not related to apoptosis. This pan-nuclear γH2AX is mediated by the kinases ataxia telangiectasia mutated and DNA-dependent protein kinase (DNA-PK) that also phosphorylate H2AX at DSBs. The pan-nuclear response is dependent on the amount of DNA damage and is transient even under conditions of impaired DSB repair. Using fluorescence recovery after photobleaching (FRAP), we found that MDC1, but not 53BP1, binds to the nuclear-wide γH2AX. Consequently, the accumulation of MDC1 at DSBs is reduced. Altogether, we show that a transient dose-dependent activation of the kinases occurring on complex DNA lesions leads to their nuclear-wide distribution and H2AX phosphorylation, yet without eliciting a full pan-nuclear DNA damage response.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/enzimologia , Quebras de DNA de Cadeia Dupla , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Linhagem Celular , Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Células Cultivadas , Cromatina/química , Cricetinae , Histonas/análise , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Proteínas Nucleares/metabolismo , Fosforilação , Radiação Ionizante , Transativadores/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
7.
BMC Cancer ; 13: 151, 2013 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-23530619

RESUMO

BACKGROUND: To determine whether ceramide is responsible for the induction of p53-independent early or late apoptosis in response to high- and low-Linear-Energy-Transfer (LET) irradiation. METHODS: Four cell lines displaying different radiosensitivities and p53-protein status were irradiated with photons or 33.4 or 184 keV/µm carbon ions. The kinetics of ceramide production was quantified by fluorescent microscopy or High-Performance-Liquid-Chromatogaphy and the sequence of events leading to apoptosis by flow cytometry. RESULTS: Regardless of the p53-status, both low and high-LET irradiation induced an early ceramide production in radiosensitive cells and late in the radioresistant. This production strongly correlated with the level of early apoptosis in radiosensitive cells and delayed apoptosis in the radioresistant ones, regardless of radiation quality, tumor type, radiosensitivity, or p53-status. Inhibition of caspase activity or ceramide production showed that, for both types of radiation, ceramide is essential for the initiation of early apoptosis in radiosensitive cells and late apoptosis following mitotic catastrophe in radioresistant cells. CONCLUSIONS: Ceramide is a determining factor in the onset of early and late apoptosis after low and high-LET irradiation and is the mediator of the p53-independent-apoptotic pathway. We propose that ceramide is the molecular bridge between mitotic catastrophe and the commitment phase of delayed apoptosis in response to irradiation.


Assuntos
Apoptose/genética , Apoptose/efeitos da radiação , Ceramidas/metabolismo , Radiação Ionizante , Proteína Supressora de Tumor p53/genética , Carbono , Caspases/metabolismo , Linhagem Celular Tumoral , Ceramidas/biossíntese , Relação Dose-Resposta à Radiação , Humanos , Cinética , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Fótons
8.
Mutat Res ; 756(1-2): 30-6, 2013 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-23628434

RESUMO

Chromatin modifications are long known as an essential part of the orchestrated response resulting in the repair of radiation-induced DNA double-strand breaks (DSBs). Only recently, however, the influence of the chromatin architecture itself on the DNA damage response has been recognised. Thus for heterochromatic DSBs the sensing and early recruitment of repair factors to the lesion occurs within the heterochromatic compartments, but the damage sites are subsequently relocated from the inside to the outside of the heterochromatin. While previous studies were accomplished at the constitutive heterochromatin of centromeric regions in mouse and flies, here we examine the DSB repair at the facultative heterochromatin of the inactive X chromosome (Xi) in humans. Using heavy ion irradiation we show that at later times after irradiation the DSB damage streaks bend around the Xi verifying that the relocation process is conserved between species and not specialised to repetitive sequences only. In addition, to measure chromatin relaxation at rare positions within the genome, we established live cell microscopy at the GSI microbeam thus allowing the aimed irradiation of small nuclear structures like the Xi. Chromatin decondensation at DSBs within the Xi is clearly visible within minutes as a continuous decrease of the DNA staining over time, comparable to the DNA relaxation revealed at DSBs in mouse chromocenters. Furthermore, despite being conserved between species, slight differences in the underlying regulation of these processes in heterochromatic DSBs are apparent.


Assuntos
Cromatina/genética , Cromossomos Humanos X/genética , Dano ao DNA/genética , Reparo do DNA/genética , Fibroblastos/patologia , Heterocromatina/genética , Animais , Cromossomos Humanos X/efeitos da radiação , Dano ao DNA/efeitos da radiação , Feminino , Fibroblastos/metabolismo , Imunofluorescência , Histonas/genética , Histonas/metabolismo , Humanos , Camundongos , Células NIH 3T3
9.
Nucleic Acids Res ; 39(15): 6489-99, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21511815

RESUMO

DNA double-strand breaks (DSBs) can induce chromosomal aberrations and carcinogenesis and their correct repair is crucial for genetic stability. The cellular response to DSBs depends on damage signaling including the phosphorylation of the histone H2AX (γH2AX). However, a lack of γH2AX formation in heterochromatin (HC) is generally observed after DNA damage induction. Here, we examine γH2AX and repair protein foci along linear ion tracks traversing heterochromatic regions in human or murine cells and find the DSBs and damage signal streaks bending around highly compacted DNA. Given the linear particle path, such bending indicates a relocation of damage from the initial induction site to the periphery of HC. Real-time imaging of the repair protein GFP-XRCC1 confirms fast recruitment to heterochromatic lesions inside murine chromocenters. Using single-ion microirradiation to induce localized DSBs directly within chromocenters, we demonstrate that H2AX is early phosphorylated within HC, but the damage site is subsequently expelled from the center to the periphery of chromocenters within ∼ 20 min. While this process can occur in the absence of ATM kinase, the repair of DSBs bordering HC requires the protein. Finally, we describe a local decondensation of HC at the sites of ion hits, potentially allowing for DSB movement via physical forces.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Eucromatina/metabolismo , Heterocromatina/metabolismo , Histonas/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Humanos , Cinética , Camundongos , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteína 1 Complementadora Cruzada de Reparo de Raio-X
10.
J Biol Chem ; 286(11): 9107-19, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21149446

RESUMO

The recognition and signaling of DNA double strand breaks involves the participation of multiple proteins, including the protein kinase ATM (mutated in ataxia-telangiectasia). ATM kinase is activated in the vicinity of the break and is recruited to the break site by the Mre11-Rad50-Nbs1 complex, where it is fully activated. In human cells, the activation process involves autophosphorylation on three sites (Ser(367), Ser(1893), and Ser(1981)) and acetylation on Lys(3016). We now describe the identification of a new ATM phosphorylation site, Thr(P)(1885) and an additional autophosphorylation site, Ser(P)(2996), that is highly DNA damage-inducible. We also confirm that human and murine ATM share five identical phosphorylation sites. We targeted the ATM phosphorylation sites, Ser(367) and Ser(2996), for further study by generating phosphospecific antibodies against these sites and demonstrated that phosphorylation of both was rapidly induced by radiation. These phosphorylations were abolished by a specific inhibitor of ATM and were dependent on ATM and the Mre11-Rad50-Nbs1 complex. As found for Ser(P)(1981), ATM phosphorylated at Ser(367) and Ser(2996) localized to sites of DNA damage induced by radiation, but ATM recruitment was not dependent on phosphorylation at these sites. Phosphorylation at Ser(367) and Ser(2996) was functionally important because mutant forms of ATM were defective in correcting the S phase checkpoint defect and restoring radioresistance in ataxia-telangiectasia cells. These data provide further support for the importance of autophosphorylation in the activation and function of ATM in vivo.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Quebras de DNA de Cadeia Dupla , Proteínas de Ligação a DNA/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Hidrolases Anidrido Ácido , Animais , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Linhagem Celular Transformada , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Ativação Enzimática/efeitos da radiação , Humanos , Proteína Homóloga a MRE11 , Camundongos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Fosforilação/efeitos da radiação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/genética , Radiação Ionizante , Fase S/efeitos dos fármacos , Fase S/genética , Fase S/efeitos da radiação , Proteínas Supressoras de Tumor/genética
11.
J Biol Chem ; 286(36): 31542-56, 2011 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-21757780

RESUMO

The Mre11/Rad50/NBN complex plays a central role in coordinating the cellular response to DNA double-strand breaks. The importance of Rad50 in that response is evident from the recent description of a patient with Rad50 deficiency characterized by chromosomal instability and defective ATM-dependent signaling. We report here that ATM (defective in ataxia-telangiectasia) phosphorylates Rad50 at a single site (Ser-635) that plays an important adaptor role in signaling for cell cycle control and DNA repair. Although a Rad50 phosphosite-specific mutant (S635G) supported normal activation of ATM in Rad50-deficient cells, it was defective in correcting DNA damage-induced signaling through the ATM-dependent substrate SMC1. This mutant also failed to correct radiosensitivity, DNA double-strand break repair, and an S-phase checkpoint defect in Rad50-deficient cells. This was not due to disruption of the Mre11/Rad50/NBN complex revealing for the first time that phosphorylation of Rad50 plays a key regulatory role as an adaptor for specific ATM-dependent downstream signaling through SMC1 for DNA repair and cell cycle checkpoint control in the maintenance of genome integrity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiologia , Ciclo Celular , Proteínas Cromossômicas não Histona/fisiologia , Enzimas Reparadoras do DNA/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Hidrolases Anidrido Ácido , Ataxia Telangiectasia/patologia , Proteínas Mutadas de Ataxia Telangiectasia , Enzimas Reparadoras do DNA/deficiência , Proteínas de Ligação a DNA/deficiência , Instabilidade Genômica , Humanos , Proteínas Mutantes , Fosforilação/fisiologia , Tolerância a Radiação , Fase S , Transdução de Sinais
12.
J Cell Biol ; 177(2): 219-29, 2007 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-17438073

RESUMO

The DNA-dependent protein kinase catalytic subunit (DNA-PK(CS)) plays an important role during the repair of DNA double-strand breaks (DSBs). It is recruited to DNA ends in the early stages of the nonhomologous end-joining (NHEJ) process, which mediates DSB repair. To study DNA-PK(CS) recruitment in vivo, we used a laser system to introduce DSBs in a specified region of the cell nucleus. We show that DNA-PK(CS) accumulates at DSB sites in a Ku80-dependent manner, and that neither the kinase activity nor the phosphorylation status of DNA-PK(CS) influences its initial accumulation. However, impairment of both of these functions results in deficient DSB repair and the maintained presence of DNA-PK(CS) at unrepaired DSBs. The use of photobleaching techniques allowed us to determine that the kinase activity and phosphorylation status of DNA-PK(CS) influence the stability of its binding to DNA ends. We suggest a model in which DNA-PK(CS) phosphorylation/autophosphorylation facilitates NHEJ by destabilizing the interaction of DNA-PK(CS) with the DNA ends.


Assuntos
Domínio Catalítico , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteína Quinase Ativada por DNA/metabolismo , Animais , Antígenos Nucleares/metabolismo , Células CHO , Cricetinae , Cricetulus , DNA/metabolismo , Proteína Quinase Ativada por DNA/química , Proteínas de Ligação a DNA/metabolismo , Humanos , Autoantígeno Ku , Lasers , Fosforilação , Fotodegradação
13.
Nucleic Acids Res ; 38(5): 1489-503, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20008512

RESUMO

Aprataxin, defective in the neurodegenerative disorder ataxia oculomotor apraxia type 1, resolves abortive DNA ligation intermediates during DNA repair. Here, we demonstrate that aprataxin localizes at sites of DNA damage induced by high LET radiation and binds to mediator of DNA-damage checkpoint protein 1 (MDC1/NFBD1) through a phosphorylation-dependent interaction. This interaction is mediated via the aprataxin FHA domain and multiple casein kinase 2 di-phosphorylated S-D-T-D motifs in MDC1. X-ray structural and mutagenic analysis of aprataxin FHA domain, combined with modelling of the pSDpTD peptide interaction suggest an unusual FHA binding mechanism mediated by a cluster of basic residues at and around the canonical pT-docking site. Mutation of aprataxin FHA Arg29 prevented its interaction with MDC1 and recruitment to sites of DNA damage. These results indicate that aprataxin is involved not only in single strand break repair but also in the processing of a subset of double strand breaks presumably through its interaction with MDC1.


Assuntos
Caseína Quinase II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Ciclo Celular , Linhagem Celular , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Humanos , Transferência Linear de Energia , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosforilação , Domínios e Motivos de Interação entre Proteínas , Transativadores/metabolismo
14.
Cells ; 11(16)2022 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-36010636

RESUMO

DNA double-strand breaks (DSBs) represent the molecular origin of ionizing-radiation inflicted biological effects. An increase in the ionization density causes more complex, clustered DSBs that can be processed by resection also in G1 phase, where repair of resected DSBs is considered erroneous and may contribute to the increased biological effectiveness of heavy ions in radiotherapy. To investigate the resection regulation of complex DSBs, we exposed G1 cells depleted for different candidate factors to heavy ions or α-particle radiation. Immunofluorescence microscopy was used to monitor the resection marker RPA, the DSB marker γH2AX and the cell-cycle markers CENP-F and geminin. The Fucci system allowed to select G1 cells, cell survival was measured by clonogenic assay. We show that in G1 phase the ubiquitin ligase RNF138 functions in resection regulation. RNF138 ubiquitinates the resection factor CtIP in a radiation-dependent manner to allow its DSB recruitment in G1 cells. At complex DSBs, RNF138's participation becomes more relevant, consistent with the observation that also resection is more frequent at these DSBs. Furthermore, deficiency of RNF138 affects both DSB repair and cell survival upon induction of complex DSBs. We conclude that RNF138 is a regulator of resection that is influenced by DSB complexity and can affect the quality of DSB repair in G1 cells.


Assuntos
Quebras de DNA de Cadeia Dupla , Endodesoxirribonucleases/metabolismo , Ubiquitina , Proteínas de Transporte/genética , DNA , Fase G1/genética , Humanos , Ligases , Ubiquitina/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
15.
Hum Mol Genet ; 18(21): 4102-17, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19643912

RESUMO

Aprataxin, defective in the neurodegenerative disorder ataxia oculomotor apraxia type 1 (AOA1), is a DNA repair protein that processes the product of abortive ligations, 5' adenylated DNA. In addition to its interaction with the single-strand break repair protein XRCC1, aprataxin also interacts with poly-ADP ribose polymerase 1 (PARP-1), a key player in the detection of DNA single-strand breaks. Here, we reveal reduced expression of PARP-1, apurinic endonuclease 1 (APE1) and OGG1 in AOA1 cells and demonstrate a requirement for PARP-1 in the recruitment of aprataxin to sites of DNA breaks. While inhibition of PARP activity did not affect aprataxin activity in vitro, it retarded its recruitment to sites of DNA damage in vivo. We also demonstrate the presence of elevated levels of oxidative DNA damage in AOA1 cells coupled with reduced base excision and gap filling repair efficiencies indicative of a synergy between aprataxin, PARP-1, APE-1 and OGG1 in the DNA damage response. These data support both direct and indirect modulating functions for aprataxin on base excision repair.


Assuntos
Dano ao DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Células Cultivadas , DNA Glicosilases/genética , DNA Glicosilases/metabolismo , Reparo do DNA , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Proteínas de Ligação a DNA/genética , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Peróxido de Hidrogênio/farmacologia , Immunoblotting , Camundongos , Camundongos Knockout , Microscopia Confocal , Proteínas Nucleares/genética , Estresse Oxidativo , Poli(ADP-Ribose) Polimerases/genética , Ligação Proteica , Mapeamento de Interação de Proteínas , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/metabolismo , Ataxias Espinocerebelares/patologia
16.
Radiat Environ Biophys ; 49(3): 427-35, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20535615

RESUMO

Metastasis and recurrences are major problems regarding an effective treatment of solid malignant tumors in clinical oncology. Since the impact of radiation on cell motility is not yet well understood, intrinsic and radiation-induced changes in cell migration have been discussed as possible mechanisms involved in the limitations of radiotherapy. This holds true for conventional radiation treatment and even more for the cellular and molecular effects of therapeutically relevant (12)C heavy ions. The present study is therefore focused on the investigation of tumor cell migration in vitro after irradiation with X-rays and (12)C heavy ions and on radiation-induced changes in the expression of proteins that are potentially relevant for motility. Two colon carcinoma cell lines, HCT116 and HCT116 p21-/-, were chosen for this study, which should be isogenic except for their p21-status. We can show here that cells lacking p21 react almost alike to radiation as wild type cells regarding survival and tumor cell migration 24 h after irradiation. Interestingly, differences in protein expression 24 h after irradiation of beta(1) integrin and protein kinase B isoforms Akt1 and Akt2 seem to exist. We conclude that tumor cell migration is unaffected by the p21-status in colorectal carcinoma cells and that the expression of the aforementioned proteins alone is not accountable for the differences observed.


Assuntos
Carbono , Movimento Celular/efeitos da radiação , Neoplasias do Colo/patologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Íons Pesados , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Integrina beta1/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Raios X
17.
Radiat Environ Biophys ; 49(3): 457-62, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20490530

RESUMO

The aim of this interdisciplinary project is to establish slice culture preparations from rodents and humans as a new model system for studying effects of X-rays and heavy ions within normal and tumor tissues. The advantage of such slice cultures relies on the conservation of an organotypic environment, the easy treatment and observation by live-imaging microscopy, and the independence from genetic immortalization strategies used to generate cell lines. Rat brains as well as human tumors were cut into 300-mum-thick sections and cultivated in an incubator in a humidified atmosphere at 37 degrees C. This is realized by a membrane-based culture system with a liquid-air interface. With this system, it is possible to keep rodent slices viable for several months. Human brain tumor slices remained vital for at least 21 days. Slices were irradiated with X-rays at the radiation facility of the University Hospital in Frankfurt/Main at doses up to 40 Gy. Heavy ion irradiations were performed at GSI (Darmstadt) with different ions, energies, and doses. The irradiated slices were analyzed by 3D-confocal microscopy following immunostaining for DNA damage, microglia, and proliferation markers. The phosphorylated histone gammaH2AX proved to be suitable for the detection of ion traversals in this system.


Assuntos
Íons Pesados , Técnicas de Cultura de Tecidos/métodos , Animais , Carbono , Proliferação de Células/efeitos da radiação , Dano ao DNA , Humanos , Microglia/citologia , Microglia/metabolismo , Microglia/efeitos da radiação , Microscopia Confocal , Ratos , Raios X , Xenônio
18.
Sci Rep ; 10(1): 1443, 2020 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-31996740

RESUMO

DNA double-strand break (DSB) repair is crucial to maintain genomic stability. The fidelity of the repair depends on the complexity of the lesion, with clustered DSBs being more difficult to repair than isolated breaks. Using live cell imaging of heavy ion tracks produced at a high-energy particle accelerator we visualised simultaneously the recruitment of different proteins at individual sites of complex and simple DSBs in human cells. NBS1 and 53BP1 were recruited in a few seconds to complex DSBs, but in 40% of the isolated DSBs the recruitment was delayed approximately 5 min. Using base excision repair (BER) inhibitors we demonstrate that some simple DSBs are generated by enzymatic processing of base damage, while BER did not affect the complex DSBs. The results show that DSB processing and repair kinetics are dependent on the complexity of the breaks and can be different even for the same clastogenic agent.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA/genética , DNA/genética , Neoplasias/genética , Sítios de Ligação/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Íons Pesados , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ligação Proteica , Síncrotrons , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
19.
Radiat Res ; 170(3): 316-26, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18763861

RESUMO

Intracellular calcium is an important second messenger that regulates many cell functions. Recent studies have shown that calcium ions can also regulate the cellular responses to ionizing radiation. However, previous data are restricted to cells treated with low-LET radiations (X rays, gamma rays and beta particles). In this work, we investigated the calcium levels in cells exposed to heavy-ion radiation of high LET. The experiments were performed at the single ion hit facility of the GSI heavy-ion microprobe. Using a built-in online calcium imaging system, the intracellular calcium concentrations were examined in HeLa cells and human foreskin fibroblast AG1522-D cells before and after irradiation with 4.8 MeV/nucleon carbon or argon ions. Although the experiment was sensitive enough to detect the calcium response to other known stimuli, no response to heavy-ion radiation was found in these two cell types. We also found that heavy-ion radiation has no impact on calcium oscillation induced by hypoxia stress in fibroblast cells.


Assuntos
Cálcio/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/efeitos da radiação , Íons Pesados , Linhagem Celular , Relação Dose-Resposta à Radiação , Células HeLa , Humanos , Doses de Radiação
20.
Radiat Res ; 169(6): 649-59, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18494540

RESUMO

The investigation of fragment length distributions of plasmid DNA gives insight into the influence of localized energy distribution on the induction of DNA damage, particularly the clustering of double-strand breaks. We present an approach that determines the fragment length distributions of plasmid DNA after heavy-ion irradiation by using the Local Effect Model. We find a good agreement of our simulations with experimental fragment distributions derived from atomic force microscopy (AFM) studies by including experimental constraints typical for AFM. Our calculations reveal that by comparing the fragmentation in terms of fluence, we can uniquely distinguish the effect of different radiation qualities. For very high-LET irradiation using nickel or uranium ions, no difference between their fragment distributions can be expected for the same dose level. However, for carbon ions with an intermediate LET, the fragmentation pattern differs from the distribution for very high-LET particles. The results of the model calculations can be used to determine the optimal experimental parameters for a demonstration of the influence of track structure on primary radiation damage. Additionally, we compare the results of our model for two different plasmid geometries.


Assuntos
Biofísica/métodos , DNA/química , Íons Pesados , Microscopia de Força Atômica/métodos , Plasmídeos/análise , Raios X , Carbono/química , DNA Super-Helicoidal , Íons , Modelos Estatísticos , Modelos Teóricos , Níquel/química , Distribuição Normal , Conformação de Ácido Nucleico , Plasmídeos/química , Plasmídeos/metabolismo , Urânio/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA