Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Radiat Res ; 65(3): 263-271, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38461549

RESUMEN

Ionizing radiation (IR)-induced double-strand breaks (DSBs) are primarily repaired by non-homologous end joining or homologous recombination (HR) in human cells. DSB repair requires adenosine-5'-triphosphate (ATP) for protein kinase activities in the multiple steps of DSB repair, such as DNA ligation, chromatin remodeling, and DNA damage signaling via protein kinase and ATPase activities. To investigate whether low ATP culture conditions affect the recruitment of repair proteins at DSB sites, IR-induced foci were examined in the presence of ATP synthesis inhibitors. We found that p53 binding protein 1 foci formation was modestly reduced under low ATP conditions after IR, although phosphorylated histone H2AX and mediator of DNA damage checkpoint 1 foci formation were not impaired. Next, we examined the foci formation of breast cancer susceptibility gene I (BRCA1), replication protein A (RPA) and radiation 51 (RAD51), which are HR factors, in G2 phase cells following IR. Interestingly, BRCA1 and RPA foci in the G2 phase were significantly reduced under low ATP conditions compared to that under normal culture conditions. Notably, RAD51 foci were drastically impaired under low ATP conditions. These results suggest that HR does not effectively progress under low ATP conditions; in particular, ATP shortages impair downstream steps in HR, such as RAD51 loading. Taken together, these results suggest that the maintenance of cellular ATP levels is critical for DNA damage response and HR progression after IR.


Asunto(s)
Adenosina Trifosfato , Proteína BRCA1 , Recombinación Homóloga , Recombinasa Rad51 , Radiación Ionizante , Humanos , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/biosíntesis , Recombinación Homóloga/efectos de la radiación , Recombinasa Rad51/metabolismo , Proteína BRCA1/metabolismo , Roturas del ADN de Doble Cadena/efectos de la radiación , Proteína de Replicación A/metabolismo , Línea Celular Tumoral , Espacio Intracelular/metabolismo , Espacio Intracelular/efectos de la radiación , Reparación del ADN , Histonas/metabolismo
2.
Mol Cell ; 82(14): 2557-2570.e7, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35594857

RESUMEN

Antigen presentation by the human leukocyte antigen (HLA) on the cell surface is critical for the transduction of the immune signal toward cytotoxic T lymphocytes. DNA damage upregulates HLA class I presentation; however, the mechanism is unclear. Here, we show that DNA-damage-induced HLA (di-HLA) presentation requires an immunoproteasome, PSMB8/9/10, and antigen-transporter, TAP1/2, demonstrating that antigen production is essential. Furthermore, we show that di-HLA presentation requires ATR, AKT, mTORC1, and p70-S6K signaling. Notably, the depletion of CBP20, a factor initiating the pioneer round of translation (PRT) that precedes nonsense-mediated mRNA decay (NMD), abolishes di-HLA presentation, suggesting that di-antigen production requires PRT. RNA-seq analysis demonstrates that DNA damage reduces NMD transcripts in an ATR-dependent manner, consistent with the requirement for ATR in the initiation of PRT/NMD. Finally, bioinformatics analysis identifies that PRT-derived 9-mer peptides bind to HLA and are potentially immunogenic. Therefore, DNA damage signaling produces immunogenic antigens by utilizing the machinery of PRT/NMD.


Asunto(s)
Degradación de ARNm Mediada por Codón sin Sentido , Biosíntesis de Proteínas , Presentación de Antígeno , Daño del ADN , Antígenos de Histocompatibilidad Clase I/genética , Humanos
3.
Cell Rep ; 38(5): 110335, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108530

RESUMEN

Single-stranded DNA (ssDNA) arising as an intermediate of cellular processes on DNA is a potential vulnerability of the genome unless it is appropriately protected. Recent evidence suggests that R-loops, consisting of ssDNA and DNA-RNA hybrids, can form in the proximity of DNA double-strand breaks (DSBs) within transcriptionally active regions. However, how the vulnerability of ssDNA in R-loops is overcome during DSB repair remains unclear. Here, we identify RAP80 as a factor suppressing the vulnerability of ssDNA in R-loops, chromosome translocations, and deletions during DSB repair. Mechanistically, RAP80 prevents unscheduled nucleolytic processing of ssDNA in R-loops by CtIP. This mechanism promotes efficient DSB repair via transcription-associated end joining dependent on BRCA1, Polθ, and LIG1/3. Thus, RAP80 suppresses the vulnerability of R-loops during DSB repair, thereby precluding genomic abnormalities in a critical component of the genome caused by deleterious R-loop processing.


Asunto(s)
Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Chaperonas de Histonas/metabolismo , Estructuras R-Loop/fisiología , ADN/genética , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN por Unión de Extremidades/fisiología , ADN de Cadena Simple/metabolismo , Humanos , ARN/genética
4.
Biochem Biophys Res Commun ; 591: 95-101, 2022 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-35007836

RESUMEN

Post-translational modification of proteins by small ubiquitin-like modifier (SUMO) is known to be involved in a variety of cellular events. This modification, called SUMOylation, is carried out by the E1 activating enzyme, the E2 conjugating enzyme, and multiple E3 ligases. Previous studies have demonstrated that the SUMO E3 ligases, protein inhibitors of activated STAT 1 (PIAS1) and 4 (PIAS4), and the SUMO-targeted ubiquitin ligase, RING finger protein 4 (RNF4), play important roles in the repair of DNA double-strand breaks (DSBs). However, the mechanism by which these SUMO-related enzymes promote DSB repair is still poorly understood. In the present study, we focused on homologous recombination (HR), the most accurate DSB repair pathway, and aimed to elucidate the mechanism by which PIAS1, PIAS4, and RNF4 promote HR. In γ-ray-irradiated normal human fibroblasts, DSB end resection and RAD51 loading, the two essential steps of HR, were significantly impaired by small interfering RNA (siRNA)-mediated depletion of PIAS1, PIAS4, or RNF4. The recruitment of BRCA1, a major HR factor, to DSB sites was reduced in cells depleted of these SUMO-related enzymes. Consistent with the role of BRCA1 in counteracting the p53-binding protein 1 (53BP1)-mediated resection blockade, 53BP1 depletion rescued the reduced resection and RAD51 loading in the cells depleted of PIAS1, PIAS4, or RNF4. Moreover, Rap1-interacting factor 1 (RIF1), a resection inhibitor downstream of 53BP1, became more abundant at DSBs when PIAS1, PIAS4, RNF4, or BRCA1 was depleted. Importantly, the concomitant depletion of BRCA1 with either one of the SUMO-related enzymes did not further increase RIF1 at DSBs, when compared to single depletion of BRCA1. Collectively, these results suggest that PIAS1, PIAS4, RNF4, and BRCA1 work epistatically to counteract 53BP1/RIF1-mediated resection blockade, thereby promoting resection.


Asunto(s)
Roturas del ADN de Doble Cadena , Recombinación Homóloga , Proteínas Nucleares/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Factores de Transcripción/metabolismo , Línea Celular , Humanos , Masculino , Unión Proteica , Recombinasa Rad51/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
5.
J Radiat Res ; 63(1): 19-29, 2022 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-34738103

RESUMEN

Malignant pleural mesothelioma (MPM) is a highly malignant disease that develops after asbestos exposure. Although the number of MPM cases is predicted to increase, no effective standard therapies have been established. The novel radiosensitizer α-sulfoquinovosyl-acylpropanediol (SQAP) enhances the effects of γ-radiation in human lung and prostate cancer cell lines and in animal models. In this study, we explored the radiosensitizing effect of SQAP and its mechanisms in MPM. The human MPM cell lines MSTO-211H and MESO-4 were implanted subcutaneously into the backs and thoracic cavities of immunodeficient KSN/Slc mice, then 2 mg/kg SQAP was intravenously administered with or without irradiation with a total body dose of 8 Gy. In both the orthotopic and ectopic xenograft murine models, the combination of irradiation plus SQAP delayed the implanted human MSTO-211H tumor growth. The analysis of the changes in the relative tumor volume of the MSTO-211H indicated a statistically significant difference after 8 Gy total body combined with 2 mg/kg SQAP, compared to both the untreated control (P = 0.0127) and the radiation treatment alone (P = 0.0171). After the treatment in each case, immunostaining of the harvested tumors revealed decreased cell proliferation, increased apoptosis and normalization of tumor blood vessels in the SQAP- and irradiation-treated group. Furthermore, hypoxia-inducible factor (HIF) 1 mRNA and protein expression were decreased, indicating reoxygenation in this group. In conclusion, SQAP improved hypoxic conditions in tumor tissue and may elicit a radiosensitizing effect in malignant mesothelioma models.


Asunto(s)
Antineoplásicos , Mesotelioma Maligno , Mesotelioma , Neoplasias Pleurales , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Humanos , Masculino , Mesotelioma/tratamiento farmacológico , Mesotelioma/metabolismo , Mesotelioma/radioterapia , Ratones , Ratones Desnudos , Neoplasias Pleurales/tratamiento farmacológico , Neoplasias Pleurales/metabolismo , Neoplasias Pleurales/radioterapia , Tolerancia a Radiación
6.
Biochem Biophys Res Commun ; 572: 191-196, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34375929

RESUMEN

Chromosome rearrangements, which are structural chromosomal abnormalities commonly found in human cancer, result from the misrejoining between two or more DNA double-strand breaks arising at different genomic regions. Consequently, chromosome rearrangements can generate fusion genes that promote tumorigenesis. The mechanisms of chromosome rearrangement have been studied using exogenous double-strand break inducers, such as radiation and nucleases. However, the mechanism underlying the occurrence of chromosome rearrangements in the absence of exogenous double-strand break-inducing stimuli is unclear. This study aimed to identify the major source of chromosome rearrangements and the DNA repair pathway that suppresses them. DNA repair factors that potentially suppress gene fusion were screened using The Cancer Genome Atlas dataset. In total, 22 repair factors whose expression levels were negatively correlated with the frequency of gene fusion were identified. More than 60% of these repair factors are involved in homologous recombination, a major double-strand break repair pathway. We hypothesized that DNA single-strand breaks are the source of double-strand breaks that lead to chromosome rearrangements. This study demonstrated that hydrogen peroxide (H2O2)-induced single-strand breaks gave rise to double-strand breaks in a replication-dependent manner. Additionally, H2O2 induced the formation of RPA and RAD51 foci, which indicated that double-strand breaks derived from single-strand breaks were repaired through homologous recombination. Moreover, treatment with H2O2 promoted the formation of radial chromosomes, a type of chromosome rearrangements, only upon the downregulation of homologous recombination factors, such as BRCA1 and CtIP. Thus, single-strand breaks are the major source of chromosome rearrangements when the expression of homologous recombination factors is downregulated.


Asunto(s)
Cromosomas/genética , Reordenamiento Génico/genética , Recombinación Homóloga/genética , Células Cultivadas , Cromosomas/efectos de los fármacos , Cromosomas/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN , Reordenamiento Génico/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología
7.
Genes (Basel) ; 12(8)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34440406

RESUMEN

Chromosome rearrangements are structural variations in chromosomes, such as inversions and translocations. Chromosome rearrangements have been implicated in a variety of human diseases. Ataxia-telangiectasia (A-T) is an autosomal recessive disorder characterized by a broad range of clinical and cellular phenotypes. At the cellular level, one of the most prominent features of A-T cells is chromosome rearrangement, especially that in T lymphocytes. The gene that is defective in A-T is ataxia-telangiectasia mutated (ATM). The ATM protein is a serine/threonine kinase and plays a central role in the cellular response to DNA damage, particularly DNA double-strand breaks. In this review, the mechanisms by which ATM suppresses chromosome rearrangements are discussed.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Ataxia Telangiectasia/genética , Cromosomas/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Humanos
8.
J Radiat Res ; 62(5): 773-781, 2021 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-34196706

RESUMEN

Programmed death ligand 1 (PD-L1) expression on the surface of cancer cells affects the efficacy of anti-PD-1/PD-L1 immune checkpoint therapy. However, the mechanism underlying PD-L1 expression in cancer cells is not fully understood, particularly after ionizing radiation (IR). Here, we examined the impact of high linear energy transfer (LET) carbon-ion irradiation on the expression of PD-L1 in human osteosarcoma U2OS cells. We found that the upregulation of PD-L1 expression after high LET carbon-ion irradiation was greater than that induced by X-rays at the same physical and relative biological effectiveness (RBE) dose, and that the upregulation of PD-L1 induced by high LET carbon-ion irradiation was predominantly dependent on ataxia telangiectasia and Rad3-related (ATR) kinase activity. Moreover, we showed that the downstream signaling, e.g. STAT1 phosphorylation and IRF1 expression, was upregulated to a greater extent after high LET carbon-ion irradiation than X-rays, and that IRF1 upregulation was also ATR dependent. Finally, to visualize PD-L1 molecules on the cell surface in 3D, we applied immunofluorescence-based super-resolution imaging. The three-dimensional structured illumination microscopy (3D-SIM) analyses revealed substantial increases in the number of presented PD-L1 molecules on the cell surface after high LET carbon-ion irradiation compared with X-ray irradiation.


Asunto(s)
Antígeno B7-H1/biosíntesis , Neoplasias Óseas/patología , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Radioterapia de Iones Pesados , Proteínas de Neoplasias/biosíntesis , Osteosarcoma/patología , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/fisiología , Antígeno B7-H1/genética , Línea Celular Tumoral , Humanos , Imagenología Tridimensional , Factor 1 Regulador del Interferón/biosíntesis , Factor 1 Regulador del Interferón/genética , Transferencia Lineal de Energía , Morfolinas/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Fosforilación/efectos de la radiación , Procesamiento Proteico-Postraduccional/efectos de la radiación , Pirazinas/farmacología , Pironas/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Factor de Transcripción STAT1/metabolismo , Sulfonas/farmacología , Regulación hacia Arriba/efectos de la radiación , Rayos X
9.
Sci Rep ; 10(1): 21146, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33273685

RESUMEN

Cellular metabolism is directly or indirectly associated with various cellular processes by producing a variety of metabolites. Metabolic alterations may cause adverse effects on cell viability. However, some alterations potentiate the rescue of the malfunction of the cell system. Here, we found that the alteration of glucose metabolism suppressed genome instability caused by the impairment of chromatin structure. Deletion of the TDH2 gene, which encodes glyceraldehyde 3-phospho dehydrogenase and is essential for glycolysis/gluconeogenesis, partially suppressed DNA damage sensitivity due to chromatin structure, which was persistently acetylated histone H3 on lysine 56 in cells with deletions of both HST3 and HST4, encoding NAD+-dependent deacetylases. tdh2 deletion also restored the short replicative lifespan of cells with deletion of sir2, another NAD+-dependent deacetylase, by suppressing intrachromosomal recombination in rDNA repeats increased by the unacetylated histone H4 on lysine 16. tdh2 deletion also suppressed recombination between direct repeats in hst3∆ hst4∆ cells by suppressing the replication fork instability that leads to both DNA deletions among repeats. We focused on quinolinic acid (QUIN), a metabolic intermediate in the de novo nicotinamide adenine dinucleotide (NAD+) synthesis pathway, which accumulated in the tdh2 deletion cells and was a candidate metabolite to suppress DNA replication fork instability. Deletion of QPT1, quinolinate phosphoribosyl transferase, elevated intracellular QUIN levels and partially suppressed the DNA damage sensitivity of hst3∆ hst4∆ cells as well as tdh2∆ cells. qpt1 deletion restored the short replicative lifespan of sir2∆ cells by suppressing intrachromosomal recombination among rDNA repeats. In addition, qpt1 deletion could suppress replication fork slippage between direct repeats. These findings suggest a connection between glucose metabolism and genomic stability.


Asunto(s)
Eliminación de Gen , Inestabilidad Genómica , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Saccharomyces cerevisiae/enzimología , Acetilación , Cromosomas Fúngicos , Daño del ADN , Replicación del ADN , Glucosa/metabolismo , NAD/metabolismo , Ácido Quinolínico/metabolismo , Saccharomyces cerevisiae/metabolismo
10.
DNA Repair (Amst) ; 91-92: 102872, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32502756

RESUMEN

The cell-killing effect of radiotherapy largely depends on unrepaired DNA double-stranded breaks (DSBs) or lethal chromosome aberrations induced by DSBs. Thus, the capability of DSB repair is critically important for the cancer-cell-killing effect of ionizing radiation. Here, we investigated the involvement of the DNA damage signaling factors ataxia telangiectasia mutated (ATM), ring finger protein 8 (RNF8), and RNF168 in quiescent G0/G1 cells, which are expressed in the majority of cell populations in tumors, after high linear energy transfer (LET) carbon-ion irradiation. Interestingly, ATM inhibition caused a substantial DSB repair defect after high-LET carbon-ion irradiation. Similarly, RNF8 or RNF168 depletion caused a substantial DSB repair defect. ATM inhibition did not exert an additive effect in RNF8-depleted cells, suggesting that ATM and RNF8 function in the same pathway. Importantly, we found that the RNF8 RING mutant showed a similar DSB repair defect, suggesting the requirement of ubiquitin ligase activity in this repair pathway. The RNF8 FHA domain was also required for DSB repair in this axis. Furthermore, the p53-binding protein 1 (53BP1), which is an important downstream factor in RNF8-dependent DSB repair, was also required for this repair. Importantly, either ATM inhibition or RNF8 depletion increased the frequency of chromosomal breaks, but reduced dicentric chromosome formation, demonstrating that ATM/RNF8 is required for the rejoining of DSB ends for the formation of dicentric chromosomes. Finally, we showed that RNF8 depletion augmented radiosensitivity after high-LET carbon-ion irradiation. This study suggests that the inhibition of RNF8 activity or its downstream pathway may augment the efficacy of high-LET carbon-ion therapy.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células A549 , Línea Celular , Aberraciones Cromosómicas , ADN/metabolismo , ADN/efectos de la radiación , Humanos , Transferencia Lineal de Energía , Tolerancia a Radiación , Transducción de Señal , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Rayos X
11.
Cancer Sci ; 111(4): 1357-1366, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31991041

RESUMEN

Survivin belongs to the inhibitor of apoptosis protein family, which is consistently overexpressed in most cancer cells but rarely expressed in normal adult tissues. Therefore, the detection and inhibition of survivin are regarded as attractive strategies for cancer-specific treatment. In this study, we designed and synthesized 7-19 residues of inner centromere protein (INCENP)-derived small peptides (INC peptides) as novel survivin-targeting agents. The INC peptides showed binding affinity for the human survivin protein (Kd  = 91.4-255 nmol L-1 ); INC16-22 , which contains residues 16-22 of INCENP, showed the highest affinity (91.4 nmol L-1 ). Confocal fluorescence imaging showed consistent colocalization of FITC-INC16-22 and survivin in cell lines. Nona-arginine-linked INC16-22 (r9-INC16-22 ) rendered INC16-22 cells penetrable and strongly inhibited cell growth of MIA PaCa-2 cells (52% inhibition at 1.0 µmol L-1 ) and MDA-MB-231 cells (60% inhibition at 10 µmol L-1 ) as determined by MTT assays. The exposure of MIA PaCa-2 cells to 40 µmol L-1 r9-INC16-22 apparently reduced the intracellular protein expression levels of survivin. However, cleaved caspase-3 was significantly increased in cells treated with r9-INC16-22 , even at 10 µmol L-1 , compared to untreated cells. Flow cytometry revealed that r9-INC16-22 strongly induced apoptosis in MIA PaCa-2 cells. These results indicate that the cytotoxic effects of r9-INC16-22 could be mediated mainly through the disruption of survivin-dependent antiapoptotic functions and partly because of the direct degradation of the survivin protein. Our findings suggest that INC peptides can act as useful scaffolds for novel cancer imaging and anticancer agents.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Proteínas Cromosómicas no Histona/genética , Péptidos/farmacología , Survivin/aislamiento & purificación , Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Caspasas/química , Caspasas/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Proteínas Cromosómicas no Histona/química , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis/química , Proteínas Inhibidoras de la Apoptosis/aislamiento & purificación , Imagen Molecular/métodos , Péptidos/síntesis química , Péptidos/química , Survivin/química , Survivin/genética
12.
Oncol Rep ; 42(6): 2293-2302, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31578593

RESUMEN

Ribosomes are important cellular components that maintain cellular homeostasis through overall protein synthesis. The nucleolus is a prominent subnuclear structure that contains ribosomal DNA (rDNA) encoding ribosomal RNA (rRNA), an essential component of ribosomes. Despite the significant role of the rDNA­rRNA­ribosome axis in cellular homeostasis, the stability of rDNA in the context of the DNA damage response has not been fully investigated. In the present study, the number and morphological changes of nucleolin, a marker of the nucleolus, were examined following ionizing radiation (IR) in order to investigate the impact of DNA damage on nucleolar stability. An increase in the number of nucleoli per cell was found in HCT116 and U2OS cells following IR. Interestingly, the IR­dependent increase in nucleolar fragmentation was enhanced by p53 deficiency. In addition, the morphological analysis revealed several distinct types of nucleolar fragmentation following IR. The pattern of nucleolar morphology differed between HCT116 and U2OS cells, and the p53 deficiency altered the pattern of nucleolar morphology. Finally, a significant decrease in rRNA synthesis was observed in HCT116 p53­/­ cells following IR, suggesting that severe nucleolar fragmentation downregulates rRNA transcription. The findings of the present study suggest that p53 plays a key role in protecting the transcriptional activity of rDNA in response to DNA damage.


Asunto(s)
Neoplasias Óseas/genética , Nucléolo Celular/metabolismo , Neoplasias Colorrectales/genética , Osteosarcoma/genética , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Apoptosis , Neoplasias Óseas/patología , Nucléolo Celular/genética , Nucléolo Celular/efectos de la radiación , Neoplasias Colorrectales/patología , Daño del ADN , ADN Ribosómico/genética , ADN Ribosómico/metabolismo , Humanos , Osteosarcoma/patología , Fosfoproteínas/genética , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , Proteínas de Unión al ARN/genética , Radiación Ionizante , Transcripción Genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Nucleolina
13.
J Radiat Res ; 60(1): 69-79, 2019 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-30476166

RESUMEN

Photons, such as X- or γ-rays, induce DNA damage (distributed throughout the nucleus) as a result of low-density energy deposition. In contrast, particle irradiation with high linear energy transfer (LET) deposits high-density energy along the particle track. High-LET heavy-ion irradiation generates a greater number and more complex critical chromosomal aberrations, such as dicentrics and translocations, compared with X-ray or γ irradiation. In addition, the formation of >1000 bp deletions, which is rarely observed after X-ray irradiation, has been identified following high-LET heavy-ion irradiation. Previously, these chromosomal aberrations have been thought to be the result of misrepair of complex DNA lesions, defined as DNA damage through DNA double-strand breaks (DSBs) and single-strand breaks as well as base damage within 1-2 helical turns (<3-4 nm). However, because the scale of complex DNA lesions is less than a few nanometers, the large-scale chromosomal aberrations at a micrometer level cannot be simply explained by complex DNA lesions. Recently, we have demonstrated the existence of clustered DSBs along the particle track through the use of super-resolution microscopy. Furthermore, we have visualized high-level and frequent formation of DSBs at the chromosomal boundary following high-LET heavy-ion irradiation. In this review, we summarize the latest findings regarding the hallmarks of DNA damage structure and the repair pathway following heavy-ion irradiation. Furthermore, we discuss the mechanism through which high-LET heavy-ion irradiation may induce dicentrics, translocations and large deletions.


Asunto(s)
Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/efectos de la radiación , Iones Pesados , Cromosomas/efectos de la radiación , Histonas/metabolismo , Humanos , Investigación Biomédica Traslacional
14.
Cell ; 175(2): 558-570.e11, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30245011

RESUMEN

Given that genomic DNA exerts its function by being transcribed, it is critical for the maintenance of homeostasis that DNA damage, such as double-strand breaks (DSBs), within transcriptionally active regions undergoes accurate repair. However, it remains unclear how this is achieved. Here, we describe a mechanism for transcription-associated homologous recombination repair (TA-HRR) in human cells. The process is initiated by R-loops formed upon DSB induction. We identify Rad52, which is recruited to the DSB site in a DNA-RNA-hybrid-dependent manner, as playing pivotal roles in promoting XPG-mediated R-loop processing and initiating subsequent repair by HRR. Importantly, dysfunction of TA-HRR promotes DSB repair via non-homologous end joining, leading to a striking increase in genomic aberrations. Thus, our data suggest that the presence of R-loops around DSBs within transcriptionally active regions promotes accurate repair of DSBs via processing by Rad52 and XPG to protect genomic information in these critical regions from gene alterations.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Proteínas Nucleares/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Reparación del ADN por Recombinación/fisiología , Factores de Transcripción/metabolismo , Línea Celular , ADN/genética , Roturas del ADN de Doble Cadena , Daño del ADN , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Proteínas de Unión al ADN/fisiología , Endonucleasas/fisiología , Recombinación Homóloga , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , ARN/genética , Proteína Recombinante y Reparadora de ADN Rad52/genética , Factores de Transcripción/fisiología
15.
Hum Immunol ; 79(8): 627-631, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29859207

RESUMEN

Programmed cell death-1 (PD-1) and its ligand (programmed death-ligand 1, PD-L1) are key factors that regulate a cytotoxic immune reaction. Anti-PD-1 therapy provides significant clinical benefits for patients with cancer, even those with advanced-stage cancer. We have recently demonstrated that DNA damage signaling from DNA double-strand breaks (DSBs) promotes PD-L1 upregulation in cancer cells. In the present study, we aimed to investigate PD-L1 expression in primary normal human dermal fibroblasts (NHDFs) in response to DSBs. We demonstrated that PD-L1 expression in NHDFs is not upregulated after ionizing radiation (IR). In addition, interferon (IFN) regulatory factor 1 (IRF1) and signal transducer and activator of transcription 1 (STAT1) phosphorylation do not respond in NHDFs after IR. In contrast, IFNγ treatment upregulates PD-L1 and IRF1 expressions and STAT1 phosphorylation. The nonresponsiveness was also observed after treatment with other DNA-damaging agents, such as camptothecin and etoposide. Treatment with a histone deacetylase inhibitor (HDACi), which causes chromatin relaxation and restores gene silencing, upregulates PD-L1 without exogenous DNA damage; however, IR-dependent upregulation is not observed in NHDFs treated with HDACi. Taken together, our data suggest that DNA-damage signaling is insufficient for upregulating PD-L1 in NHDFs.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígeno B7-H1/metabolismo , Daño del ADN/inmunología , Dermis/patología , Fibroblastos/fisiología , Inmunoterapia/métodos , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Células Cultivadas , Etopósido/farmacología , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Humanos , Factor 1 Regulador del Interferón/metabolismo , Cultivo Primario de Células , Radiación Ionizante , Factor de Transcripción STAT1/metabolismo
16.
Sci Rep ; 7: 41812, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-28155885

RESUMEN

Chromosome rearrangement is clinically and physiologically important because it can produce oncogenic fusion genes. Chromosome rearrangement requires DNA double-strand breaks (DSBs) at two genomic locations and misrejoining between the DSBs. Before DSB misrejoining, two DSB-containing chromatin regions move and pair with each other; however, the molecular mechanism underlying this process is largely unknown. We performed a spatiotemporal analysis of ionizing radiation-induced foci of p53-binding protein 1 (53BP1), a marker for DSB-containing chromatin. We found that some 53BP1 foci were paired, indicating that the two damaged chromatin regions neighboured one another. We searched for factors regulating the foci pairing and found that the number of paired foci increased when Ku80, DNA-PKcs, or ATM was absent. In contrast, 53BP1 depletion reduced the number of paired foci and dicentric chromosomes-an interchromosomal rearrangement. Foci were paired more frequently in heterochromatin than in euchromatin in control cells. Additionally, the reduced foci pairing in 53BP1-depleted cells was rescued by concomitant depletion of a heterochromatin building factor such as Krüppel-associated box-associated protein 1 or chromodomain helicase DNA-binding protein 3. These findings indicate that pairing between DSB-containing chromatin regions was suppressed by Ku80, DNA-PKcs, and ATM, and this pairing was promoted by 53BP1 through chromatin relaxation.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Cromatina/genética , Cromatina/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Autoantígeno Ku/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Biomarcadores , Cromatina/efectos de la radiación , Aberraciones Cromosómicas , Roturas del ADN de Doble Cadena/efectos de la radiación , Proteínas de Unión al ADN/metabolismo , Fibroblastos , Técnica del Anticuerpo Fluorescente , Humanos , Unión Proteica , Radiación Ionizante , Transducción de Señal
17.
Oncotarget ; 8(65): 109370-109381, 2017 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-29312614

RESUMEN

DNA double-strand breaks (DSBs) induced by ionising radiation are considered the major cause of genotoxic mutations and cell death. While DSBs are dispersed throughout chromatin after X-rays or γ-irradiation, multiple types of DNA damage including DSBs, single-strand breaks and base damage can be generated within 1-2 helical DNA turns, defined as a complex DNA lesion, after high Linear Energy Transfer (LET) particle irradiation. In addition to the formation of complex DNA lesions, recent evidence suggests that multiple DSBs can be closely generated along the tracks of high LET particle irradiation. Herein, by using three dimensional (3D)-structured illumination microscopy, we identified the formation of 3D widespread γH2AX foci after high LET carbon-ion irradiation. The large γH2AX foci in G2-phase cells encompassed multiple foci of replication protein A (RPA), a marker of DSBs undergoing resection during homologous recombination. Furthermore, we demonstrated by 3D analysis that the distance between two individual RPA foci within γH2AX foci was approximately 700 nm. Together, our findings suggest that high LET heavy-ion particles induce clustered DSB formation on a scale of approximately 1 µm3. These closely localised DSBs are considered to be a risk for the formation of chromosomal rearrangement after heavy-ion irradiation.

18.
Health Phys ; 110(6): 558-62, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27115222

RESUMEN

From October to December 2010, just before the radiological accident at the Fukushima Daiichi nuclear power plant, 71 radiation professionals from radiation facilities in Japan were asked what they considered as a "safe dose" of radiation for themselves, their partners, parents, children, siblings, and friends. Although the 'safe dose' they noted varied widely, from less than 1 mSv y to more than 100 mSv y, the average dose was 35.6 mSv y, which is around the middle point between the legal exposure dose limits for the annual average and for any single year. Similar results were obtained from other surveys of members of the Japan Radioisotope Association (36.9 mSv y) and of the Oita Prefectural Hospital (36.8 mSv y). Among family members and friends, the minimum average "safe" dose was 8.5 mSv y for children, for whom 50% of the responders claimed a "safe dose" of less than 1 mSv. Gender, age and specialty of the radiation professional also affected their notion of a "safe dose." These findings suggest that the perception of radiation risk varies widely even for radiation professionals and that the legal exposure dose limits derived from regulatory science may act as an anchor of safety. The different levels of risk perception for different target groups among radiation professionals appear similar to those in the general population. The gap between these characteristics of radiation professionals and the generally accepted picture of radiation professionals might have played a role in the state of confusion after the radiological accident.


Asunto(s)
Accidente Nuclear de Fukushima , Dosis Máxima Tolerada , Plantas de Energía Nuclear , Opinión Pública , Traumatismos por Radiación/psicología , Protección Radiológica/estadística & datos numéricos , Adulto , Actitud del Personal de Salud , Testimonio de Experto , Femenino , Humanos , Japón , Masculino , Persona de Mediana Edad , Dosis de Radiación , Medición de Riesgo/estadística & datos numéricos , Recursos Humanos
19.
Genes Chromosomes Cancer ; 55(8): 650-60, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27113385

RESUMEN

Chromosomal translocations arise from misrejoining of DNA double strand breaks (DSBs) between loci located on two chromosomes. One current model suggests that spatial proximity of potential chromosomal translocation partners influences translocation probability. Ionizing radiation (IR) is a potent inducer of translocations. Accumulating evidence demonstrates that particle irradiation more frequently causes translocations compared with X-ray irradiation. This observation has led to the hypothesis that the high frequency of translocations after particle irradiation may be due to the formation of DSBs at chromosome boundaries along the particle track, because such DSBs can be misrejoined between distinct chromosomes. In this study, we simultaneously visualized the site of IR-induced DSBs and chromosome position by combining Immunofluorescence and fluorescence in situ hybridization. Importantly, the frequency of γH2AX foci at the chromosome boundary of chromosome 1 after carbon-ion irradiation was >4-fold higher than that after X-ray irradiation. This observation is consistent with the idea that particle irradiation generates DSBs at the boundaries of two chromosomes along the track. Further, we showed that resolution of γH2AX foci at chromosome boundaries is prevented by inhibition of DNA-PKcs activity, indicating that the DSB repair is NHEJ-dependent. Finally, we found that γH2AX foci at chromosome boundaries after carbon-ion irradiation contain DSBs undergoing DNA-end resection, which promotes repair utilizing microhomology mediated end-joining during translocation. Taken together, our study suggests that the frequency of DSB formation at chromosome boundaries is associated with the incidence of chromosomal translocations, supporting the notion that the spatial proximity between breaks is an important factor in translocation formation. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN por Unión de Extremidades/efectos de la radiación , Histonas/genética , Translocación Genética/efectos de la radiación , Radioisótopos de Carbono , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 1/efectos de la radiación , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Fibroblastos/efectos de la radiación , Humanos , Hibridación Fluorescente in Situ , Radiación Ionizante , Rayos X
20.
PLoS One ; 9(12): e115121, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25531293

RESUMEN

BACKGROUND AND PURPOSE: To understand the mechanisms involved in the strong killing effect of carbon-ion beam irradiation on cancer cells with TP53 tumor suppressor gene deficiencies. MATERIALS AND METHODS: DNA damage responses after carbon-ion beam or X-ray irradiation in isogenic HCT116 colorectal cancer cell lines with and without TP53 (p53+/+ and p53-/-, respectively) were analyzed as follows: cell survival by clonogenic assay, cell death modes by morphologic observation of DAPI-stained nuclei, DNA double-strand breaks (DSBs) by immunostaining of phosphorylated H2AX (γH2AX), and cell cycle by flow cytometry and immunostaining of Ser10-phosphorylated histone H3. RESULTS: The p53-/- cells were more resistant than the p53+/+ cells to X-ray irradiation, while the sensitivities of the p53+/+ and p53-/- cells to carbon-ion beam irradiation were comparable. X-ray and carbon-ion beam irradiations predominantly induced apoptosis of the p53+/+ cells but not the p53-/- cells. In the p53-/- cells, carbon-ion beam irradiation, but not X-ray irradiation, markedly induced mitotic catastrophe that was associated with premature mitotic entry with harboring long-retained DSBs at 24 h post-irradiation. CONCLUSIONS: Efficient induction of mitotic catastrophe in apoptosis-resistant p53-deficient cells implies a strong cancer cell-killing effect of carbon-ion beam irradiation that is independent of the p53 status, suggesting its biological advantage over X-ray treatment.


Asunto(s)
Apoptosis/efectos de la radiación , Radiación Ionizante , Proteína p53 Supresora de Tumor/genética , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de la radiación , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Células HCT116 , Radioterapia de Iones Pesados , Histonas/metabolismo , Humanos , Puntos de Control de la Fase M del Ciclo Celular/efectos de la radiación , Fosforilación , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...