Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 515
Filter
1.
J Cell Mol Med ; 24(17): 10027-10041, 2020 09.
Article in English | MEDLINE | ID: mdl-32790210

ABSTRACT

Both SIRT1 and UVA radiation are involved in cellular damage processes such as apoptosis, senescence and ageing. MicroRNAs (miRNAs) have been reported to be closely related to UV radiation, as well as to SIRT1. In this study, we investigated the connections among SIRT1, UVA and miRNA in human skin primary fibroblasts. Our results showed that UVA altered the protein level of SIRT1 in a time point-dependent manner. Using miRNA microarray, bioinformatics analysis, we found that knocking down SIRT1 could cause up-regulation of miR-27a-5p and the latter could down-regulate SMAD2, and these results were verified by qRT-PCR or Western blot. Furthermore, UVA radiation (5 J/cm2 ), knocking down SIRT1 or overexpression of miR-27a-5p led to increased expression of MMP1, and decreased expressions of COL1 and BCL2. We also found additive impacts on MMP1, COL1 and BCL2 under the combination of UVA radiation + Sirtinol (SIRT1 inhibitor), or UVA radiation + miR-27a-5p mimic. SIRT1 activator resveratrol could reverse damage changes caused by UVA radiation. Besides, absent of SIRT1 or overexpression of miR-27a-5p increased cell apoptosis and induced cell arrest in G2/M phase. Taken together, these results demonstrated that UVA could influence a novel SIRT1-miR-27a-5p-SMAD2-MMP1/COL1/BCL2 axis in skin primary fibroblasts, and may provide potential therapeutic targets for UVA-induced skin damage.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/radiation effects , Proteins/metabolism , Signal Transduction/radiation effects , Skin/metabolism , Skin/radiation effects , Ultraviolet Rays/adverse effects , Adolescent , Adult , Apoptosis/radiation effects , Cell Cycle Checkpoints/radiation effects , Cell Division/radiation effects , Cells, Cultured , Down-Regulation/radiation effects , G2 Phase/radiation effects , Humans , Up-Regulation/radiation effects , Young Adult
2.
Radiol Oncol ; 54(2): 168-179, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32229678

ABSTRACT

Background Management of locoregionally recurrent head and neck squamous cell carcinomas (HNSCC) is challenging due to potential radioresistance. Pulsed low-dose rate (PLDR) irradiation exploits phenomena of increased radiosensitivity, low-dose hyperradiosensitivity (LDHRS), and inverse dose-rate effect. The purpose of this study was to evaluate LDHRS and the effect of PLDR irradiation in isogenic HNSCC cells with different radiosensitivity. Materials and methods Cell survival after different irradiation regimens in isogenic parental FaDu and radioresistant FaDu-RR cells was determined by clonogenic assay; post irradiation cell cycle distribution was studied by flow cytometry; the expression of DNA damage signalling genes was assesed by reverse transcription-quantitative PCR. Results Radioresistant Fadu-RR cells displayed LDHRS and were more sensitive to PLDR irradiation than parental FaDu cells. In both cell lines, cell cycle was arrested in G2/M phase 5 hours after irradiation. It was restored 24 hours after irradiation in parental, but not in the radioresistant cells, which were arrested in G1-phase. DNA damage signalling genes were under-expressed in radioresistant compared to parental cells. Irradiation increased DNA damage signalling gene expression in radioresistant cells, while in parental cells only few genes were under-expressed. Conclusions We demonstrated LDHRS in isogenic radioresistant cells, but not in the parental cells. Survival of LDHRS-positive radioresistant cells after PLDR was significantly reduced. This reduction in cell survival is associated with variations in DNA damage signalling gene expression observed in response to PLDR most likely through different regulation of cell cycle checkpoints.


Subject(s)
Head and Neck Neoplasms/radiotherapy , Neoplasm Recurrence, Local/radiotherapy , Radiation Tolerance , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Survival/radiation effects , DNA Damage/genetics , G1 Phase/radiation effects , G2 Phase/radiation effects , Gene Expression , Humans , Mitosis/radiation effects , Radiotherapy Dosage , Time Factors , Tumor Stem Cell Assay/methods
3.
Radiat Res ; 193(5): 451-459, 2020 05.
Article in English | MEDLINE | ID: mdl-32150497

ABSTRACT

Findings from previous studies have suggested that the telomerase system is involved in radiation-induced genomic instability. In this study, we investigated the involvement of telomerase in the development and processing of chromosomal damage at different cell cycle stages after irradiation of human fibroblasts. Several response criteria were investigated, including cell survival, chromosomal damage (using the micronucleus assay), G2-induced chromatid aberrations (using the conventional G2 assay as well as a chemically-induced premature chromosome condensation assay) and DNA double-strand breaks (DSBs; using γ-H2AX, 53BP1 and Rad51) in an isogenic pair of cell lines: BJ human foreskin fibroblasts and BJ1-hTERT, a telomerase-immortalized BJ cell line. To distinguish among G1, S and G2 phase, cells were co-immunostained for CENP-F and cyclin A, which are tightly regulated proteins in the cell cycle. After X-ray irradiation at doses in the range of 0.1-6 Gy, the results showed that for cell survival and micronuclei induction, where the overall effect is dominated by the cells in G1 and S phase, no difference was observed between the two cell types; in contrast, when radiation sensitivity at the G2 stage of the cell cycle was analyzed, a significantly higher number of chromatid-type aberrations (breaks and exchanges), and higher levels of γ-H2AX and of Rad51 foci were observed for the BJ cells compared to the BJ1-hTERT cells. Therefore, it can be concluded that telomerase appears to be involved in DNA DSB repair processes, mainly in the G2 phase. These data, taken overall, reinforce the notion that hTERT or other elements of the telomere/telomerase system may defend chromosome integrity in human fibroblasts by promoting repair in G2 phase of the cell cycle.


Subject(s)
Genomic Instability/radiation effects , Telomerase/metabolism , Cell Line , Cell Survival/radiation effects , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , G2 Phase/radiation effects , Gamma Rays/adverse effects , Humans , Micronucleus Tests , Rad51 Recombinase/metabolism , S Phase/radiation effects
4.
Methods Mol Biol ; 1984: 39-45, 2019.
Article in English | MEDLINE | ID: mdl-31267418

ABSTRACT

The G2 chromosomal radiosensitivity assay or, simply G2 assay, measures the number of chromatid type aberrations induced by radiation in G2 phase. Typically, asynchronous growing cells are irradiated with less than 1 Gy and allowed 0.5-1 h for cells in mitosis, at the time of irradiation, to transit into G1. Later, the G2 phase cells, at the time irradiation, are blocked by colcemid for 1-4 h at metaphase. Cells are collected by standard hypotonic solution and Carnoy solution fixation or directly fixed onto the culture vessels. The G2 assay can detect severe radiosensitivity in ATM homozygous mutated cells and relatively small differences among cellular radiosensitivity such as heterozygous mutation carriers of ATM and BRCA1/2 mutation carriers. The G2 assay also has the capability to detect cancer prone individuals. This assay only requires a conventional cell culture facility and the standard microscopic observation.


Subject(s)
Biological Assay/methods , Chromosomes/radiation effects , G2 Phase/radiation effects , Radiation Tolerance/radiation effects , Animals , CHO Cells , Chromatids/metabolism , Chromatids/radiation effects , Chromosome Aberrations/radiation effects , Cricetinae , Cricetulus , X-Rays
5.
Methods Mol Biol ; 1984: 47-60, 2019.
Article in English | MEDLINE | ID: mdl-31267419

ABSTRACT

Chromosome analysis is a fundamental technique for a wide range of cytogenetic studies. Chromosome aberrations are easily introduced by many kinds of clastogenic agents such as ionizing irradiation, UV, or alkylating agents, and damaged chromosomes may be prone to cancer. Chromosomes are conventionally prepared from mitotic cells arrested by the colcemid block method. However, obtaining of mitotic chromosomes is sometimes hampered under several circumstances, for example after high-dose (over several Gys of γ-rays) ionizing irradiation exposure accident. As a result, cytogenetic analysis will be often difficult or even impossible in such cases. Premature chromosome condensation (PCC) is an alternative technique that has proved to be a unique and useful way in chromosome analysis. Previously, PCC has been achieved following cell fusion mediated either by fusogenic viruses (for example Sendai virus) or by polyethylene glycol (PEG) (cell-fusion PCC), but the cell-fusion PCC has several drawbacks. The novel drug-induced PCC use of specific inhibitors for serine/threonine protein phosphatase was introduced about 20 years ago. This method is much simple and easy even than the conventional mitotic chromosome preparation using colcemid block protocol and the obtained PCC index (equivalent to mitotic index for metaphase chromosome) is much higher. Furthermore, this method allows the interphase chromatin to be condensed and visualized like mitotic chromosomes, and thus has been opening the way for chromosome analysis not only in metaphase chromosomes but also in interphase chromatin. The drug-induced PCC has therefore proven the usefulness in cytogenetics and other many cell biology fields. Since the first version of drug-induced PCC protocol has been published in 2009 (Gotoh, Methods in molecular biology. Humana Press, New York, 2009), many newer applications of drug-induced PCC in radiation biology and chromosome science fields in a wide range of species from animal to plant have been reported (Gotoh et al., Biomed Res 16:63-68, 1995; Lamadrid Boada et al., Mutat Res 757:45-51, 2013; Ravi et al., Biochimie 95:124-33, 2013; Ono et al., J Cell Biol 200:429-41, 2013; Vagnarelli, Exp Cell Res 318:1435-41, 2012; Roukos et al., Nat Protoc 9:2476-92, 2014; Miura and Blakely, Cytometry A 79:1016-22, 2013; Zabka et al., J Plant Physiol 174:62-70, 2015; Samaniego et al., Planta 215:195-204, 2002; Rybaczek et al., Folia Histochem Cytobiol 40:51-9, 2002; Gotoh and Durante J Cell Physiol 209:297-304, 2006). Therefore as a new edition, I will write in this chapter the drug-induced PCC technique with newer findings, in particular focused drug-induced PCC protocols in radiation biology with referring updated articles published recently.


Subject(s)
Chromatin/metabolism , Cytogenetic Analysis/methods , G2 Phase/radiation effects , Interphase/radiation effects , Mitosis/radiation effects , Pharmaceutical Preparations/metabolism , Radiobiology/methods , Animals , Cell Adhesion/radiation effects , Cells, Cultured , Chromosome Aberrations/radiation effects , DNA Repair/radiation effects , Dose-Response Relationship, Radiation , Humans , Kinetics , Radiation, Ionizing
6.
Mutat Res ; 815: 10-19, 2019 05.
Article in English | MEDLINE | ID: mdl-30999232

ABSTRACT

The classical G2-assay is widely used to assess cell-radiosensitivity and cancer phenotype: Cells are exposed to low doses of ionizing-radiation (IR) and collected for cytogenetic- analysis ˜1.5 h later. In this way, chromosome-damage is measured in cells irradiated in G2-phase, without retrieving information regarding kinetics of chromosome-break-repair. Modification of the assay to include analysis at multiple time-points after IR, has enabled kinetic-analysis of chromatid-break-repair and assessment of damage in a larger proportion of G2-phase cells. This modification, however, increases the probability that at later time points not only cells irradiated in G2-phase, but also cells irradiated in S-phase will reach metaphase. However, the response of cells irradiated in G2-phase can be mechanistically different from that of cells irradiated in S-phase. Therefore, indiscriminate analysis may confound the interpretation of experiments designed to elucidate mechanisms of chromosome-break-repair and the contributions of the different DSB-repair-pathways in this response. Here we report an EdU based modification of the assay that enables S- and G2-phase specific analysis of chromatid break repair. Our results show that the majority of metaphases captured during the first 2 h after IR originate from cells irradiated in G2-phase (EdU- metaphases) in both rodent and human cells. Metaphases originating from cells irradiated in S-phase (EdU+ metaphases) start appearing at 2 h and 4 h after IR in rodent and human cells, respectively. The kinetics of chromatid-break-repair are similar in cells irradiated in G2- and S-phase of the cell-cycle, both in rodent and human cells. The protocol is applicable to classical-cytogenetic experiments and allows the cell-cycle specific analysis of chromosomal-aberrations. Finally, the protocol can be applied to the kinetic analysis of chromosome-breaks in prematurely-condensed-chromosomes of G2-phase cells. In summary, the developed protocol provides means to enhance the analysis of IR-induced-cytogenetic-damage by providing information on the cell-cycle phase where DNA damage is inflicted.


Subject(s)
Chromosome Aberrations/radiation effects , Chromosomes/genetics , Metaphase/genetics , Metaphase/radiation effects , Animals , CHO Cells , Cell Line , Cell Line, Tumor , Chromosome Breakage/drug effects , Chromosomes/radiation effects , Cricetulus , DNA Repair/genetics , DNA Repair/radiation effects , G2 Phase/genetics , G2 Phase/radiation effects , HCT116 Cells , Humans , Kinetics , Radiation, Ionizing , S Phase/genetics , S Phase/radiation effects
7.
Int J Radiat Biol ; 95(5): 597-606, 2019 05.
Article in English | MEDLINE | ID: mdl-30507333

ABSTRACT

PURPOSE: Radioresistance is an important factor for unsatisfactory prognosis in Nasopharyngeal carcinoma (NPC) patients. Ubiquitous mitochondrial creatine kinase (CKMT1) is always associated with malignancy in a variety of cancers. However, its significance in NPC progression and radiosensitivity remains unclear. The present study focused on investigating the effects of CKMT1 on NPC cell radiosensitivity. MATERIAL AND METHODS: CKMT1 was overexpressed in NPC cell line CNE-1 or knocked out in CNE-2. Biological changes were detected after cells exposing to different doses of X-ray to determine the role of CKMT1 on NPC cell radiosensitivity. RESULTS: CKMT1 promotes proliferation and migration in NPC cell lines CNE-1 and CNE-2. Overexpression of CKMT1 in CNE-1 cells enhanced colony formation rates, reduced G2/M phase cell cycle arrest, lowered apoptosis rate and c-PARP level, and elevated STAT3 phosphorylation level after radiation treatment. While knocking out CKMT1 using the CRISPR/Cas9 system in CNE-2 cells lowered colony formation rates, increased G2/M phase cell cycle arrest, apoptosis rates, and c-PARP levels, and decreased STAT3 phosphorylation in response to radiation treatment. CONCLUSIONS: NPC cells with higher CKMT1 exhibited lower radiosensitivity through promoting phosphorylation of STAT3. Our findings suggest that CKMT1 may be an alternative radiotherapeutic target in NPC therapy.


Subject(s)
Creatine Kinase/metabolism , Nasopharyngeal Carcinoma/pathology , Radiation Tolerance , Apoptosis/radiation effects , Cell Division/radiation effects , Cell Line, Tumor , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Creatine Kinase/deficiency , Creatine Kinase/genetics , G2 Phase/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , Gene Knockout Techniques , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
PLoS One ; 13(10): e0205691, 2018.
Article in English | MEDLINE | ID: mdl-30352062

ABSTRACT

PURPOSE: Data on the response of chondrocytes differentiated from hiPSCs (hiPSC-DCHs) to ionizing radiation (IR) are lacking. The aim of present study was to assess DNA damage response (DDR) mechanisms of IR-treated hiPSC-DCHs. METHODS AND MATERIALS: The following IR-response characteristics in irradiated hiPSC-DCHs were assessed: 1) the kinetics of DNA DSB formation; 2) activation of major DNA repair mechanisms; 3) cell cycle changes and 4) reactive oxygen species (ROS), level of key markers of apoptosis and senescence. RESULTS: DNA DSBs were observed in 30% of the hiPSC-DCHs overall, and in 60% after high-dose (> 2 Gy) IR. Nevertheless, these cells displayed efficient DNA repair mechanisms, which reduced the DSBs over time until it reached 30% by activating key genes involved in homologous recombination and non-homologous end joining mechanisms. As similar to mature chondrocytes, irradiated hiPSC-DCH cells revealed accumulation of cells in G2 phase. Overall, the hiPSC-DCH cells were characterized by low levels of ROS, cPARP and high levels of senescence. CONCLUSIONS: The chondrocyte-like cells derived from hiPSC demonstrated features characteristic of both mature chondrocytes and "parental" hiPSCs. The main difference between hiPSC-derived chondrocytes and hiPSCs and mature chondrocytes appears to be the more efficient DDR mechanism of hiPSC-DCHs. The unique properties of these cells suggest that they could potentially be used safely in regenerative medicine if these preliminary findings are confirmed in future studies.


Subject(s)
Cell Differentiation/radiation effects , Chondrocytes/physiology , Chondrogenesis/radiation effects , Gamma Rays , Induced Pluripotent Stem Cells/physiology , Cell Line , Chondrocytes/radiation effects , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair/radiation effects , G2 Phase/radiation effects , Humans , Induced Pluripotent Stem Cells/radiation effects , Reactive Oxygen Species/metabolism , Regenerative Medicine/methods
9.
Biochimie ; 154: 86-98, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30096372

ABSTRACT

Histone posttranslational modifications regulate diverse nuclear functions, including DNA repair. Here, we use mass spectrometry, western blotting, immunohistochemistry and advanced confocal microscopy in order to show radiation-specific changes in the histone signature. We studied wild-type mouse embryonic stem cells (mESCs) and mESCs with a depletion of histone deacetylase 1 (HDAC1), which plays a role in DNA repair. Irradiation by γ-rays increased the S139 phosphorylation of histone H2AX but reduced the level of the H3K9-R17 peptide, which contains S10 phosphorylation (H3S10ph). On an individual cellular level, H3S10ph was low in highly γH2AX-positive UV laser-induced DNA lesions, and this nuclear distribution pattern was not changed by HDAC1 depletion. Despite this fact, spontaneous γH2AX-positive DNA lesions colocalized with large H3S10ph-positive nuclear bodies that appear in the G2 phase of the cell cycle. Similarly, by FLIM-FRET analysis, we observed an interaction between H3S10ph and γH2AX in the G2 phase. However, this interaction was reduced when cells were exposed to γ-rays. A mutual link between H3S10ph and γH2AX was not observed in the G1 phase of the cell cycle. Together, our data show that despite the fact that H3S10ph is not directly involved in DNA repair, a decrease in H3S10 phosphorylation and weakened interaction between H3S10ph and γH2AX is a result of radiation-induced damage of the genome. In this case, γ-irradiation also decreased the number of cells in the G1 phase, characterized by no interaction between H3S10ph and γH2AX.


Subject(s)
G2 Phase/radiation effects , Gamma Rays/adverse effects , Histones/metabolism , Animals , G1 Phase/radiation effects , HeLa Cells , Histones/genetics , Humans , Mice , Phosphorylation/radiation effects
10.
Radiat Res ; 190(4): 385-395, 2018 10.
Article in English | MEDLINE | ID: mdl-29979637

ABSTRACT

The goal of this study was to determine whether tetrandrine enhanced radiosensitization in different hepatocellular carcinoma cell lines and to elucidate the potential mechanism. We also tested whether PA28γ was regulated by tetrandrine. The human hepatocellular carcinoma cell lines HepG2 and LM3 were divided into six groups: control; low-dosage (0.5 or 5 µg/ml) tetrandrine alone; high-dosage (1.0 or 10 µg/ml) tetrandrine alone; irradiation alone; irradiation with low-dosage (0.5 µg/ml or 5 µg/ml) tetrandrine; and irradiation with high-dosage (1.0 µg/ml or 10 µg/ml) tetrandrine. Colony-forming assays were performed. Expression of cyclin and apoptosis-related proteins, including cyclin B1, phosphorylated cyclin-dependent kinase 1 [phospho-CDC2 (Tyr15)], Bax and caspase-3, as well as PA28γ expression, were evaluated using Western blot analysis. Apoptosis rate and cell cycle distribution were examined using flow cytometry analysis. Tetrandrine enhanced radiosensitivity in HepG2 and LM3 cells, as characterized by a narrower shoulder area and steeper linear area, and the enhanced radiosensitization increased with tetrandrine dosage. After tetrandrine treatment, the apoptosis rate significantly increased, whereas the proportion of cells in the G2 phase dramatically decreased in dose- and time-dependent manners after irradiation. However, the effect of reverse G2 arrest was weaker in p53-mutant cells (LM3 cells). Finally, we observed that tetrandrine downregulated PA28γ expression. Moreover, when PA28γ was downregulated, apoptosis and cell cycle distribution were also altered; however, the effects were weaker in p53-mutant cells. Therefore, we propose that tetrandrine-mediated apoptosis induction and G2 arrest attenuation are at least partly mediated by PA28γ.


Subject(s)
Benzylisoquinolines/pharmacology , Carcinoma, Hepatocellular/radiotherapy , Liver Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Autoantigens/metabolism , CDC2 Protein Kinase/metabolism , Carcinoma, Hepatocellular/pathology , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cyclin B1/metabolism , Dose-Response Relationship, Drug , G2 Phase/drug effects , G2 Phase/radiation effects , Genes, p53 , Humans , Liver Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , bcl-2-Associated X Protein/metabolism
11.
G3 (Bethesda) ; 8(6): 2099-2106, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29695495

ABSTRACT

Mre11 is a key player for DNA double strand break repair. Previous studies have shown that mammalian Mre11 is methylated at multiple arginines in its C-terminal Glycine-Arginine-Rich motif (GAR) by protein arginine methyltransferase PRMT1. Here, we found that the Drosophila Mre11 is methylated at arginines 559, 563, 565, and 569 in the GAR motif by DART1, the Drosophila homolog of PRMT1. Mre11 interacts with DART1 in S2 cells, and this interaction does not require the GAR motif. Arginines methylated Mre11 localizes exclusively in the nucleus as soluble nuclear protein or chromatin-binding protein. To study the in vivo functions of methylation, we generated the single Arg-Ala and all Arginines mutated flies. We found these mutants were sensitive to ionizing radiation. Furthermore, Arg-Ala mutated flies had no irradiation induced G2/M checkpoint defect in wing disc and eye disc. Thus, we provided evidence that arginines in Drosophila Mre11 are methylated by DART1 methytransferase and flies loss of arginine methylation are sensitive to irradiation.


Subject(s)
Arginine/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/radiation effects , Endodeoxyribonucleases/metabolism , Radiation, Ionizing , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/radiation effects , Cell Line , DNA Damage , Drosophila Proteins/chemistry , Endodeoxyribonucleases/chemistry , Eye/cytology , Eye/metabolism , Eye/radiation effects , G2 Phase/genetics , G2 Phase/radiation effects , Gene Knockdown Techniques , Methylation , Methyltransferases/metabolism , Mitosis/genetics , Mitosis/radiation effects , Mutation/genetics , Survival Analysis , Wings, Animal/metabolism , Wings, Animal/radiation effects
12.
J Int Med Res ; 45(6): 2009-2022, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29239279

ABSTRACT

Objective To evaluate the radiosensitivity effect of CpG oligodeoxyribonucleotide (ODN) 7909 on human epidermoid cancer strain-2 (Hep-2) cells in vitro and discuss the potential for improved radiotherapy treatment in patients with laryngeal squamous cell carcinoma. Methods Toll-like receptor ( TLR) 9 expression was assessed in Hep-2 cells using Western blots and reverse transcription polymerase chain reaction. Cell Counting Kit-8 was used to detect Hep-2 cell viability at 24 and 48 h following treatment with different CpG ODN7909 concentrations. Cellular colonization was evaluated using microscopy. Cell cycle distribution and apoptosis rate was determined with flow cytometry. Interleukin (IL)-12 and tumour necrosis factor (TNF)-α concentrations were detected by enzyme-linked immunosorbent assay. Results Hep-2 cells were found to express TLR9, and CpG ODN7909 treatment suppressed Hep-2 cell viability in a dose- and time-dependent manner. Cell survival curve analyses revealed a sensitivity enhancement ratio of the mean death dose of 1.225 for CpG ODN7909 plus irradiation versus irradiation alone. Furthermore, the population of Gap 2/mitotic-phase cells, apoptosis rate and secreted IL-12 and TNF-α levels were significantly increased in Hep-2 cells treated with CpG ODN7909 plus irradiation versus IR alone. Conclusion CpG ODN7909 enhanced the radiosensitivity of Hep-2 cells in vitro.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Laryngeal Neoplasms/drug therapy , Oligodeoxyribonucleotides/therapeutic use , Radiation Tolerance , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Squamous Cell/pathology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Division/drug effects , Cell Division/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Clone Cells , G2 Phase/drug effects , G2 Phase/radiation effects , Humans , Interleukin-2/metabolism , Laryngeal Neoplasms/pathology , Oligodeoxyribonucleotides/pharmacology , Radiation, Ionizing , Toll-Like Receptor 9/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
Probl Radiac Med Radiobiol ; 22: 208-215, 2017 Dec.
Article in English, Ukrainian | MEDLINE | ID: mdl-29286507

ABSTRACT

OBJECTIVE: To identify the possibility of modification by astaxanthin the level of genome damages induced by gamma quanta in the culture of human peripheral blood lymphocytes exposed in vitro on postsynthetic (G2) phase of the first mitotic cycle. MATERIALS AND METHODS: Peripheral blood lymphocytes from four apparently healthy volunteers 35-51 years old were cultivated using modified micromethod. To obtain genomic damages in G2 phase of the first mitotic cycle the part of cultures was irradiated by γ quanta in dose 1.0 Gy through 46 hours of cultivation. Astaxanthin in final con centration 20 µg/ml was exposed to lymphocytes' cultures before the irradiation. Cytogenetic analysis the uniform ly stained slides of metaphase chromosomes was carried out to determine the frequencies of chromosome and chro matid types of aberrations. Using the method of individual cells electrophoresis (Comet assay) the relative level of DNA damages (Tail Moment index) and the frequency of apoptotic cells with high level of DNA fragmentation were evaluated. RESULTS: Mean group frequencies of chromosome aberrations after gamma irradiation of lymphocytes in vitro exceed ed those without radiation exposure and were 72.35 ± 1.17 and 2.46 ± 0.30 per 100 metaphases, respectively (p < 0.001), mainly due to chromatid type of aberrations (58.32 ± 1.29 per 100 metaphases). Adding of astaxanthin into culture medium before the irradiation did not result in changes as in the frequency of chromosomal damages (71.54 ± 1.34 per 100 metaphases) as in the spectrum of aberrations - also prevailed chromatid type of aberrations (58.47 ± 1.47 per 100 metaphases). The increase of Tail Moment index after radiation exposure (from 3.84 ± 0.36 to 12.06 ± 1.88, respectively, p < 0.001) and lack of significant impact of astaxanthin on this index in the irradiated lym phocytes (8.96 ± 2.39, p > 0.05) was established, ie astaxanthin didn't change the relative level of radiation induced DNA damages. Also apoptogenic effect of astaxanthin was not found: frequency of apoptotic cells were (2.25 ± 1.49) % in cultures of intact lymphocytes, (2.08 ± 1.54) % in irradiated cultures and (1.78 ± 1.25) % under joint action of gamma radiation and astaxanthin (p > 0.05). CONCLUSIONS: Noimpactofastaxanthinongenomicinstabilityinducedbygammairradiation invitroinculturesof human peripheral blood lymphocytes on postsynthetic (G2) phase of first mitotic cycle had been established.


Subject(s)
Antioxidants/pharmacology , Chromosome Aberrations/radiation effects , Gamma Rays/adverse effects , Genome, Human , Lymphocytes/radiation effects , Adult , Apoptosis/radiation effects , Comet Assay , Cytogenetic Analysis , DNA Fragmentation/radiation effects , Female , G2 Phase/radiation effects , Genomic Instability/radiation effects , Humans , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Metaphase , Middle Aged , Primary Cell Culture , Radiation Dosage , Xanthophylls/pharmacology
14.
Stem Cell Res ; 23: 98-104, 2017 08.
Article in English | MEDLINE | ID: mdl-28715716

ABSTRACT

Early mammalian embryonic cells must maintain a particularly robust DNA repair system, as mutations at this developmental point have detrimental consequences for the organism. How the repair system can be tuned to fulfill such elevated requirements is largely unknown, but it may involve transcriptional regulation. Ronin (Thap11) is a transcriptional regulator responsible for vital programs in pluripotent cells. Here, we report that this protein also modulates the DNA damage response of such cells. We show that conditional Ronin knockout sensitizes embryonic stem cells (ESCs) to UV-C-induced DNA damage in association with Atr pathway activation and G2/M arrest. Ronin binds to and regulates the genes encoding several DNA repair factors, including Gtf2h4 and Rad18, providing a potential mechanism for this phenotype. Our results suggest that the unique DNA repair requirements of the early embryo are not met by a static system, but rather via highly regulated processes.


Subject(s)
DNA Damage , Pluripotent Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , Cell Cycle Checkpoints/radiation effects , DNA Damage/genetics , DNA Repair/radiation effects , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/radiation effects , G2 Phase/radiation effects , Mice, Knockout , Mitosis/radiation effects , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/radiation effects , Radiation, Ionizing , Ultraviolet Rays
15.
Cell Cycle ; 16(6): 565-573, 2017 Mar 19.
Article in English | MEDLINE | ID: mdl-28118061

ABSTRACT

Mitochondria play a key role in maintaining cellular homeostasis during stress responses, and mitochondrial dysfunction contributes to carcinogenesis, aging, and neurologic disease. We here investigated ionizing radiation (IR)-induced mitochondrial damage in human neural progenitor stem cells (NSCs), their differentiated counterparts and human normal fibroblasts. Long-term fractionated radiation (FR) with low doses of X-rays for 31 d enhanced mitochondrial activity as evident by elevated mitochondrial membrane potential (ΔΨm) and mitochondrial complex IV (cytochrome c oxidase) activity to fill the energy demands for the chronic DNA damage response in differentiated cells. Subsequent reduction of the antioxidant glutathione via continuous activation of mitochondrial oxidative phosphorylation caused oxidative stress and genomic instability in differentiated cells exposed to long-term FR. In contrast, long-term FR had no effect on the mitochondrial activity in NSCs. This cell type showed efficient DNA repair, no mitochondrial damage, and resistance to long-term FR. After high doses of acute single radiation (SR) (> 5 Gy), cell cycle arrest at the G2 phase was observed in NSCs and human fibroblasts. Under this condition, increase in mitochondria mass, mitochondrial DNA, and intracellular reactive oxygen species (ROS) levels were observed in the absence of enhanced mitochondrial activity. Consequently, cellular senescence was induced by high doses of SR in differentiated cells. In conclusion, we demonstrated that mitochondrial radiation responses differ according to the extent of DNA damage, duration of radiation exposure, and cell differentiation.


Subject(s)
Cell Differentiation/radiation effects , Mitochondria/metabolism , Mitochondria/radiation effects , Neural Stem Cells/metabolism , Neural Stem Cells/radiation effects , Cell Cycle Checkpoints/radiation effects , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Cellular Senescence/radiation effects , DNA Repair/radiation effects , DNA, Mitochondrial/genetics , Dose-Response Relationship, Radiation , Fluorescent Antibody Technique , G2 Phase/radiation effects , Histones/metabolism , Humans , Oxidation-Reduction/radiation effects , Oxidative Phosphorylation/radiation effects , Reactive Oxygen Species/metabolism
16.
Sci Rep ; 6: 37403, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27869214

ABSTRACT

Ultraviolet B (UVB) radiation from the sun may lead to photocarcinogenesis of the skin. Sunscreens were used to protect the skin by reducing UVB irradiance, but sunscreen use did not reduce sunburn episodes. It was shown that UVB-induced erythema depends on surface exposure but not irradiance of UVB. We previously showed that irradiance plays a critical role in UVB-induced cell differentiation. This study investigated the impact of irradiance on UVB-induced photocarcinogenesis. For hairless mice receiving equivalent exposure of UVB radiation, the low irradiance (LI) UVB treated mice showed more rapid tumor development, larger tumor burden, and more keratinocytes harboring mutant p53 in the epidermis as compared to their high irradiance (HI) UVB treated counterpart. Mechanistically, using cell models, we demonstrated that LI UVB radiation allowed more keratinocytes harboring DNA damages to enter cell cycle via ERK-related signaling as compared to its HI UVB counterpart. These results indicated that at equivalent exposure, UVB radiation at LI has higher photocarcinogenic potential as compared to its HI counterpart. Since erythema is the observed sunburn at moderate doses and use of sunscreen was not found to associate with reduced sunburn episodes, the biological significance of sunburn with or without sunscreen use warrants further investigation.


Subject(s)
Carcinogenesis/radiation effects , Ultraviolet Rays , Adult , Animals , Bromodeoxyuridine/metabolism , Butadienes/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Count , Cell Survival/radiation effects , Cells, Cultured , DNA Damage , Dermatitis, Contact/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , G2 Phase/radiation effects , Humans , Immunosuppression Therapy , Keratinocytes/drug effects , Keratinocytes/pathology , Keratinocytes/radiation effects , Mice, Hairless , Mitosis/radiation effects , Mutation/genetics , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidine Dimers/metabolism , Skin Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism
17.
Appl Physiol Nutr Metab ; 41(12): 1248-1254, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27863208

ABSTRACT

The cucurbits (prebiotics) were investigated as novel agents for radio-modification against gastrointestinal injury. The cell-cycle fractions and DNA damage were monitored in HCT-15 cells. A cucurbit extract was added to culture medium 2 h before irradiation (6 Gy) and was substituted by fresh medium at 4 h post-irradiation. The whole extract of the fruits of Lagenaria siceraria, Luffa cylindrica, or Cucurbita pepo extract enhanced G2 fractions (42%, 34%, and 37%, respectively) as compared with control (20%) and irradiated control (31%). With cucurbits, the comet tail length remained shorter (L. siceraria, 28 µm; L. cylindrica, 34.2 µm; C. pepo, 36.75 µm) than irradiated control (41.75 µm). For in vivo studies, L. siceraria extract (2 mg/kg body weight) was administered orally to mice at 2 h before and 4 and 24 h after whole-body irradiation (10 Gy). L. siceraria treatment restored the glutathione contents to 48.8 µmol/gm as compared with control (27.6 µmol/gm) and irradiated control (19.6 µmol/gm). Irradiation reduced the villi height from 379 to 350 µm and width from 54 to 27 µm. L. siceraria administration countered the radiation effects (length, 366 µm; width, 30 µm, respectively) and improved the villi morphology and tight junction integrity. This study reveals the therapeutic potential of cucurbits against radiation-induced gastrointestinal injury.


Subject(s)
Fruit/chemistry , Gastrointestinal Diseases/prevention & control , Lagenidium/chemistry , Plant Extracts/therapeutic use , Prebiotics , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/therapeutic use , Animals , Cell Line, Tumor , Cucurbita/chemistry , DNA Damage , Fruit/economics , G2 Phase/radiation effects , Gastrointestinal Diseases/diet therapy , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/pathology , Glutathione/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/radiation effects , Intestinal Mucosa/ultrastructure , Luffa/chemistry , Male , Mice , Microvilli/metabolism , Microvilli/pathology , Microvilli/radiation effects , Microvilli/ultrastructure , Plant Extracts/metabolism , Radiation Effects , Radiation Injuries, Experimental/diet therapy , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Radiation-Protective Agents/metabolism , Random Allocation , Survival Analysis , Tight Junctions/metabolism , Tight Junctions/pathology , Tight Junctions/radiation effects , Tight Junctions/ultrastructure
18.
Oncotarget ; 7(44): 71660-71672, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27690219

ABSTRACT

Radiotherapy is commonly used to treat a variety of solid tumors but improvements in the therapeutic ratio are sorely needed. The aim of this study was to assess the Chk1 kinase inhibitor, MK-8776, for its ability to radiosensitize human tumor cells. Cells derived from NSCLC and HNSCC cancers were tested for radiosensitization by MK-8776. The ability of MK-8776 to abrogate the radiation-induced G2 block was determined using flow cytometry. Effects on repair of radiation-induced DNA double strand breaks (DSBs) were determined on the basis of rad51, γ-H2AX and 53BP1 foci. Clonogenic survival analyses indicated that MK-8776 radiosensitized p53-defective tumor cells but not lines with wild-type p53. Abrogation of the G2 block was evident in both p53-defective cells and p53 wild-type lines indicating no correlation with radiosensitization. However, only p53-defective cells entered mitosis harboring unrepaired DSBs. MK-8776 appeared to inhibit repair of radiation-induced DSBs at early times after irradiation. A comparison of MK-8776 to the wee1 inhibitor, MK-1775, suggested both similarities and differences in their activities. In conclusion, MK-8776 radiosensitizes tumor cells by mechanisms that include abrogation of the G2 block and inhibition of DSB repair. Our findings support the clinical evaluation of MK-8776 in combination with radiation.


Subject(s)
Checkpoint Kinase 1/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Tumor Suppressor Protein p53/physiology , Cell Line, Tumor , DNA Breaks, Double-Stranded , G2 Phase/radiation effects , Histones/analysis , Humans , Pyrimidinones , Tumor Suppressor Protein p53/genetics
19.
Sci Rep ; 6: 33376, 2016 09 14.
Article in English | MEDLINE | ID: mdl-27624978

ABSTRACT

Integrin α6 (ITGA6), a transmembrane glycoprotein adhesion receptor protein, is widely upregulated in many types of tumors and promotes migration and invasion in cancer cells. However, the role that the ITGA6-associated signaling network plays in radiosensitivity in breast cancer has not been described. The expression of ITGA6 was examined in human breast cancer and normal breast cell lines using western blot analysis. We also explored the role of ITGA6 in the regulation of radiation sensitivity in breast cancer using the colony formation assays, cell cycle analyses, apoptosis assays and immunofluorescence analyses. The results showed that the protein and mRNA expression levels of ITGA6 was higher in breast cancer cells than in normal cells. ITGA6 protectived responses to radiotherapy in breast cancer cells by altering cell apoptosis, DNA damage repair and cell-cycle regulation. Furthermore, ITGA6 enhanced radiation resistance via PI3K/Akt and MEK/Erk signaling. In addition, overexpressing ITGA6 promoted radiation resistance in cells, and this effect was neutralized by the PI3K inhibitor LY294002 and MEK inhibitor U0126. Taken together, these findings indicate that ITGA6 might be involved in a mechanism that underlies radiation resistance and that ITGA6 could be a potential target for therapies aimed at overcoming radiation resistance in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , Extracellular Signal-Regulated MAP Kinases/metabolism , Integrin alpha6/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Apoptosis/genetics , Apoptosis/radiation effects , Breast Neoplasms/genetics , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , DNA Damage , DNA Repair/radiation effects , Female , G2 Phase/genetics , G2 Phase/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Integrin alpha6/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitosis/genetics , Mitosis/radiation effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/radiation effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radiation Tolerance/genetics , Radiation Tolerance/radiation effects , Radiation, Ionizing , Signal Transduction/radiation effects
20.
PLoS One ; 11(5): e0155093, 2016.
Article in English | MEDLINE | ID: mdl-27163610

ABSTRACT

Ionizing radiation (IR) exerts deleterious effects on the developing brain, since proliferative neuronal progenitor cells are highly sensitive to IR-induced DNA damage. Assuming a radiation response that is comparable to mammals, the chick embryo would represent a lower vertebrate model system that allows analysis of the mechanisms underlying this sensitivity, thereby contributing to the reduction, refinement and replacement of animal experiments. Thus, this study aimed to elucidate the radiation response of the embryonic chick retina in three selected embryonic stages. Our studies reveal a lack in the radiation-induced activation of a G1/S checkpoint, but rapid abrogation of G2/M progression after IR in retinal progenitors throughout development. Unlike cell cycle control, radiation-induced apoptosis (RIA) showed strong variations between its extent, dose dependency and temporal occurrence. Whereas the general sensitivity towards RIA declined with ongoing differentiation, its dose dependency constantly increased with age. For all embryonic stages RIA occurred during comparable periods after irradiation, but in older animals its maximum shifted towards earlier post-irradiation time points. In summary, our results are in good agreement with data from the developing rodent retina, strengthening the suitability of the chick embryo for the analysis of the radiation response in the developing central nervous system.


Subject(s)
Apoptosis/radiation effects , G1 Phase/radiation effects , G2 Phase/radiation effects , M Phase Cell Cycle Checkpoints/radiation effects , Retina/radiation effects , Age Factors , Animals , Biomarkers/metabolism , Cell Differentiation/radiation effects , Chick Embryo , Dose-Response Relationship, Radiation , Gene Expression , Histones/genetics , Histones/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Retina/cytology , Retina/metabolism , Time Factors , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...