Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Mutat Res ; 823: 111758, 2021.
Article in English | MEDLINE | ID: mdl-34333390

ABSTRACT

Exposure to the ultraviolet (UV) radiation in sunlight creates DNA lesions, which if left unrepaired can induce mutations and contribute to skin cancer. The two most common UV-induced DNA lesions are the cis-syn cyclobutane pyrimidine dimers (CPDs) and pyrimidine (6-4) pyrimidone photoproducts (6-4PPs), both of which can initiate mutations. Interestingly, mutation frequency across the genomes of many cancers is heterogenous with significant increases in heterochromatin. Corresponding increases in UV lesion susceptibility and decreases in repair are observed in heterochromatin versus euchromatin. However, the individual contributions of CPDs and 6-4PPs to mutagenesis have not been systematically examined in specific genomic and epigenomic contexts. In this study, we compared genome-wide maps of 6-4PP and CPD lesion abundances in primary cells and conducted comprehensive analyses to determine the genetic and epigenetic features associated with susceptibility. Overall, we found a high degree of similarity between 6-4PP and CPD formation, with an enrichment of both in heterochromatin regions. However, when examining the relative levels of the two UV lesions, we found that bivalent and Polycomb-repressed chromatin states were uniquely more susceptible to 6-4PPs. Interestingly, when comparing UV susceptibility and repair with melanoma mutation frequency in these regions, disparate patterns were observed in that susceptibility was not always inversely associated with repair and mutation frequency. Functional enrichment analysis hint at mechanisms of negative selection for these regions that are essential for cell viability, immune function and induce cell death when mutated. Ultimately, these results reveal both the similarities and differences between UV-induced lesions that contribute to melanoma.


Subject(s)
DNA Repair , Epigenesis, Genetic/radiation effects , Melanoma/genetics , Mutation , Skin Neoplasms/genetics , Ultraviolet Rays/adverse effects , DNA Damage , Databases, Genetic , Euchromatin/chemistry , Euchromatin/metabolism , Euchromatin/radiation effects , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Genome, Human/radiation effects , Heterochromatin/chemistry , Heterochromatin/metabolism , Heterochromatin/radiation effects , Histones/genetics , Histones/metabolism , Humans , Melanoma/etiology , Melanoma/metabolism , Melanoma/pathology , Mutagenesis , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Primary Cell Culture , Pyrimidine Dimers/agonists , Pyrimidine Dimers/metabolism , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
2.
Int J Mol Sci ; 22(14)2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34299263

ABSTRACT

BACKGROUND: Charged-particle radiotherapy is an emerging treatment modality for radioresistant tumors. The enhanced effectiveness of high-energy particles (such as heavy ions) has been related to the spatial clustering of DNA lesions due to highly localized energy deposition. Here, DNA damage patterns induced by single and multiple carbon ions were analyzed in the nuclear chromatin environment by different high-resolution microscopy approaches. MATERIAL AND METHODS: Using the heavy-ion microbeam SNAKE, fibroblast monolayers were irradiated with defined numbers of carbon ions (1/10/100 ions per pulse, ipp) focused to micrometer-sized stripes or spots. Radiation-induced lesions were visualized as DNA damage foci (γH2AX, 53BP1) by conventional fluorescence and stimulated emission depletion (STED) microscopy. At micro- and nanoscale level, DNA double-strand breaks (DSBs) were visualized within their chromatin context by labeling the Ku heterodimer. Single and clustered pKu70-labeled DSBs were quantified in euchromatic and heterochromatic regions at 0.1 h, 5 h and 24 h post-IR by transmission electron microscopy (TEM). RESULTS: Increasing numbers of carbon ions per beam spot enhanced spatial clustering of DNA lesions and increased damage complexity with two or more DSBs in close proximity. This effect was detectable in euchromatin, but was much more pronounced in heterochromatin. Analyzing the dynamics of damage processing, our findings indicate that euchromatic DSBs were processed efficiently and repaired in a timely manner. In heterochromatin, by contrast, the number of clustered DSBs continuously increased further over the first hours following IR exposure, indicating the challenging task for the cell to process highly clustered DSBs appropriately. CONCLUSION: Increasing numbers of carbon ions applied to sub-nuclear chromatin regions enhanced the spatial clustering of DSBs and increased damage complexity, this being more pronounced in heterochromatic regions. Inefficient processing of clustered DSBs may explain the enhanced therapeutic efficacy of particle-based radiotherapy in cancer treatment.


Subject(s)
DNA Breaks, Double-Stranded/radiation effects , DNA/radiation effects , Heavy Ion Radiotherapy/adverse effects , Cell Culture Techniques , Cluster Analysis , DNA Damage/radiation effects , DNA Repair/radiation effects , Euchromatin/genetics , Euchromatin/radiation effects , Fibroblasts , Heavy Ion Radiotherapy/methods , Heavy Ions/adverse effects , Heterochromatin/genetics , Heterochromatin/radiation effects , Humans , Ku Autoantigen/genetics , Ku Autoantigen/radiation effects , Linear Energy Transfer/radiation effects , Microscopy, Electron/methods , Radiation, Ionizing
3.
Nat Commun ; 12(1): 2428, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33893291

ABSTRACT

Heterochromatin is a critical chromatin compartment, whose integrity governs genome stability and cell fate transitions. How heterochromatin features, including higher-order chromatin folding and histone modifications associated with transcriptional silencing, are maintained following a genotoxic stress challenge is unknown. Here, we establish a system for targeting UV damage to pericentric heterochromatin in mammalian cells and for tracking the heterochromatin response to UV in real time. We uncover profound heterochromatin compaction changes during repair, orchestrated by the UV damage sensor DDB2, which stimulates linker histone displacement from chromatin. Despite massive heterochromatin unfolding, heterochromatin-specific histone modifications and transcriptional silencing are maintained. We unveil a central role for the methyltransferase SETDB1 in the maintenance of heterochromatic histone marks after UV. SETDB1 coordinates histone methylation with new histone deposition in damaged heterochromatin, thus protecting cells from genome instability. Our data shed light on fundamental molecular mechanisms safeguarding higher-order chromatin integrity following DNA damage.


Subject(s)
DNA Damage , DNA Repair , DNA/genetics , Heterochromatin/genetics , Animals , Cell Line, Tumor , Chromatin Assembly and Disassembly/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heterochromatin/radiation effects , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , MCF-7 Cells , Methylation , Mice , NIH 3T3 Cells , Ultraviolet Rays
4.
Cells ; 9(5)2020 05 08.
Article in English | MEDLINE | ID: mdl-32397212

ABSTRACT

Compact chromatin is linked to a poor tumour prognosis and resistance to radiotherapy from photons. We investigated DNA damage induction and repair in the context of chromatin structure for densely ionising alpha radiation as well as its therapeutic potential. Chromatin opening by histone deacetylase inhibitor trichostatin A (TSA) pretreatment reduced clonogenic survival and increased γH2AX foci in MDA-MB-231 cells, indicative of increased damage induction by free radicals using gamma radiation. In contrast, TSA pretreatment tended to improve survival after alpha radiation while γH2AX foci were similar or lower; therefore, an increased DNA repair is suggested due to increased access of repair proteins. MDA-MB-231 cells exposed to fractionated gamma radiation (2 Gy × 6) expressed high levels of stem cell markers, elevated heterochromatin H3K9me3 marker, and a trend towards reduced clonogenic survival in response to alpha radiation. There was a higher level of H3K9me3 at baseline, and the ratio of DNA damage induced by alpha vs. gamma radiation was higher in the aggressive MDA-MB-231 cells compared to hormone receptor-positive MCF7 cells. We demonstrate that heterochromatin structure and stemness properties are induced by fractionated radiation exposure. Gamma radiation-exposed cells may be targeted using alpha radiation, and we provide a mechanistic basis for the involvement of chromatin in these effects.


Subject(s)
Alpha Particles , Breast Neoplasms/metabolism , Gamma Rays , Heterochromatin/radiation effects , Acetylation , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Clone Cells , Female , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Lysine/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Radiation Exposure , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Spheroids, Cellular/radiation effects
5.
Biochemistry ; 57(19): 2756-2761, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29658277

ABSTRACT

One of the most sensitive, time-consuming, and variable steps of chromatin immunoprecipitation (ChIP) is chromatin sonication. Traditionally, this process can take hours to properly sonicate enough chromatin for multiple ChIP assays. Further, the length of sheared DNA is often inconsistent. In order to faithfully measure chemical and structural changes at the chromatin level, sonication needs to be reliable. Thus, chromatin fragmentation by sonication represents a significant bottleneck to downstream quantitative analysis. To improve the consistency and efficiency of chromatin sonication, we developed and tested a cavitation enhancing reagent based on sonically active nanodroplets. Here, we show that nanodroplets increase sonication efficiency by 16-fold and provide more consistent levels of chromatin fragmentation. Using the previously characterized chromatin in vivo assay (CiA) platform, we generated two distinct chromatin states in order to test nanodroplet-assisted sonication sensitivity in measuring post-translational chromatin marks. By comparing euchromatin to chemically induced heterochromatin at the same CiA:Oct4 locus, we quantitatively measure the capability of our new sonication technique to resolve differences in chromatin structure. We confirm that nanodroplet-assisted sonication results are indistinguishable from those of samples processed with traditional sonication in downstream applications. While the processing time for each sample was reduced from 38.4 to 2.3 min, DNA fragment distribution sizes were significantly more consistent with a coefficient of variation 2.7 times lower for samples sonicated in the presence of nanodroplets. In conclusion, sonication utilizing the nanodroplet cavitation enhancement reagent drastically reduces the amount of processing time and provides consistently fragmented chromatin of high quality for downstream applications.


Subject(s)
Chromatin Immunoprecipitation/methods , DNA Fragmentation/radiation effects , DNA/genetics , Sonication/methods , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Chromatin/radiation effects , DNA/chemistry , DNA/radiation effects , Euchromatin/radiation effects , Heterochromatin/radiation effects , Mice , Nanoparticles/chemistry , Octamer Transcription Factor-3/chemistry , Octamer Transcription Factor-3/genetics
6.
J Invest Dermatol ; 138(2): 405-412, 2018 02.
Article in English | MEDLINE | ID: mdl-28951242

ABSTRACT

Exposure to solar UVB radiation leads to the formation of the highly mutagenic cyclobutane pyrimidine dimers (CPDs), the DNA damage responsible for mutations found in skin cancer. The frequency of CPD formation and the repair rate of those lesions are two important parameters to determine the probability of UVR-induced mutations. Previous work has shown that chronic irradiation with sublethal doses of UVB radiation (chronic low-dose UVB radiation) leads to the accumulation of residual CPD that persists over time. We have thus investigated the persistence, localization, and consequences on genome stability of those chronic low-dose UVB radiation-induced residual CPDs. We show that chronic low-dose UVB radiation-induced residual CPDs persist on DNA and are diluted via semiconservative replication. They are overrepresented in the heterochromatin and at the TT dipyrimidine sites, and they catalyze the incidence of sister chromatin exchange. Our results shed some light on the impact of chronic UVB radiation exposure on DNA, with a focus on residual CPDs, their distribution, and consequences.


Subject(s)
DNA Damage/radiation effects , DNA Repair/radiation effects , Genome, Human/radiation effects , Genomic Instability/radiation effects , Skin Neoplasms/genetics , Ultraviolet Rays/adverse effects , Adult , Biopsy , DNA Repair/genetics , DNA Replication/radiation effects , Dose-Response Relationship, Radiation , Female , Fibroblasts , Genome, Human/genetics , Genomic Instability/genetics , Heterochromatin/genetics , Heterochromatin/radiation effects , Humans , Primary Cell Culture , Pyrimidine Dimers/radiation effects , Skin/cytology , Skin/pathology , Skin/radiation effects , Young Adult
7.
Biochem J ; 474(2): 281-300, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27760841

ABSTRACT

Cellular transition to senescence is associated with extensive chromatin reorganization and changes in gene expression. Recent studies appear to imply an association of lamin B1 (LB1) reduction with chromatin rearrangement in human fibroblasts promoted to senescence, while the mechanisms and structural features of these relationships have not yet been clarified. In this work, we examined the functions of LB1 and the lamin B receptor (LBR) in human cancer cells. We found that both LB1 and LBR tend to deplete during cancer cell transfer to senescence by γ-irradiation. A functional study employing silencing of LBR by small hairpin ribonucleic acid (shRNA) constructs revealed reduced LB1 levels suggesting that the regulation of both proteins is interrelated. The reduced expression of LBR resulted in the relocation of centromeric heterochromatin (CSH) from the inner nuclear membrane (INM) to the nucleoplasm and is associated with its unfolding. This indicates that LBR tethers heterochromatin to INM in cycling cancer cells and that LB1 is an integral part of this tethering. Down-regulation of LBR and LB1 at the onset of senescence are thus necessary for the release of heterochromatin binding to lamina, resulting in changes in chromatin architecture and gene expression. However, the senescence phenotype was not manifested in cell lines with reduced LBR and LB1 expression suggesting that other factors, such as deoxyribonucleic acid (DNA) damage, are needed to trigger senescence. We conclude that the primary response of cells to various stresses leading to senescence consists of the down-regulation of LBR and LB1 to attain reversal of the chromatin architecture.


Subject(s)
Gene Expression Regulation, Neoplastic , Heterochromatin/metabolism , Lamin Type B/genetics , Osteoblasts/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Cell Line, Tumor , Cellular Senescence/radiation effects , Centromere/metabolism , Centromere/radiation effects , Centromere/ultrastructure , Gamma Rays , Heterochromatin/radiation effects , Heterochromatin/ultrastructure , Humans , Lamin Type B/metabolism , MCF-7 Cells , Nuclear Envelope/metabolism , Nuclear Envelope/radiation effects , Nuclear Envelope/ultrastructure , Osteoblasts/pathology , Osteoblasts/radiation effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Lamin B Receptor
8.
Int J Mol Sci ; 17(1)2016 Jan 02.
Article in English | MEDLINE | ID: mdl-26729112

ABSTRACT

Chromatin structure affects the extent of DNA damage and repair. Thus, it has been shown that heterochromatin is more protective against DNA double strand breaks (DSB) formation by ionizing radiation (IR); and that DNA DSB repair may proceed differently in hetero- and euchromatin regions. Human embryonic stem cells (hESC) have a more open chromatin structure than differentiated cells. Here, we study the effect of chromatin structure in hESC on initial DSB formation and subsequent DSB repair. DSB were scored by comet assay; and DSB repair was assessed by repair foci formation via 53BP1 antibody staining. We found that in hESC, heterochromatin is confined to distinct regions, while in differentiated cells it is distributed more evenly within the nuclei. The same dose of ionizing radiation produced considerably more DSB in hESC than in differentiated derivatives, normal human fibroblasts; and one cancer cell line. At the same time, the number of DNA repair foci were not statistically different among these cells. We showed that in hESC, DNA repair foci localized almost exclusively outside the heterochromatin regions. We also noticed that exposure to ionizing radiation resulted in an increase in heterochromatin marker H3K9me3 in cancer HT1080 cells, and to a lesser extent in IMR90 normal fibroblasts, but not in hESCs. These results demonstrate the importance of chromatin conformation for DNA protection and DNA damage repair; and indicate the difference of these processes in hESC.


Subject(s)
Chromatin/ultrastructure , DNA Breaks, Double-Stranded/radiation effects , Human Embryonic Stem Cells/radiation effects , Cell Differentiation , Cell Line , Chromatin/radiation effects , Heterochromatin/radiation effects , Heterochromatin/ultrastructure , Human Embryonic Stem Cells/cytology , Humans , Radiation, Ionizing
9.
Genetics ; 201(2): 563-72, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26294667

ABSTRACT

We designed a system to determine whether dicentric chromosomes in Drosophila melanogaster break at random or at preferred sites. Sister chromatid exchange in a Ring-X chromosome produced dicentric chromosomes with two bridging arms connecting segregating centromeres as cells divide. This double bridge can break in mitosis. A genetic screen recovered chromosomes that were linearized by breakage in the male germline. Because the screen required viability of males with this X chromosome, the breakpoints in each arm of the double bridge must be closely matched to produce a nearly euploid chromosome. We expected that most linear chromosomes would be broken in heterochromatin because there are no vital genes in heterochromatin, and breakpoint distribution would be relatively unconstrained. Surprisingly, approximately half the breakpoints are found in euchromatin, and the breakpoints are clustered in just a few regions of the chromosome that closely match regions identified as intercalary heterochromatin. The results support the Laird hypothesis that intercalary heterochromatin can explain fragile sites in mitotic chromosomes, including fragile X. Opened rings also were recovered after male larvae were exposed to X-rays. This method was much less efficient and produced chromosomes with a strikingly different array of breakpoints, with almost all located in heterochromatin. A series of circularly permuted linear X chromosomes was generated that may be useful for investigating aspects of chromosome behavior, such as crossover distribution and interference in meiosis, or questions of nuclear organization and function.


Subject(s)
Centromere/genetics , Chromosome Aberrations/radiation effects , Heterochromatin/genetics , X Chromosome/genetics , Animals , Drosophila melanogaster , Heterochromatin/radiation effects , Larva/genetics , Larva/radiation effects , Male , Meiosis/genetics , Mitosis/genetics , X Chromosome/radiation effects
10.
Mutat Res Rev Mutat Res ; 764: 108-17, 2015.
Article in English | MEDLINE | ID: mdl-26041270

ABSTRACT

Non-coding DNA comprises a very large proportion of the total genomic content in higher organisms, but its function remains largely unclear. Non-coding DNA sequences constitute the majority of peripheral heterochromatin, which has been hypothesized to be the genome's 'bodyguard' against DNA damage from chemicals and radiation for almost four decades. The bodyguard protective function of peripheral heterochromatin in genome defense has been strengthened by the results from numerous recent studies, which are summarized in this review. These data have suggested that cells and/or organisms with a higher level of heterochromatin and more non-coding DNA sequences, including longer telomeric DNA and rDNAs, exhibit a lower frequency of DNA damage, higher radioresistance and longer lifespan after IR exposure. In addition, the majority of heterochromatin is peripherally located in the three-dimensional structure of genome organization. Therefore, the peripheral heterochromatin with non-coding DNA could play a protective role in genome defense against DNA damage from ionizing radiation by both absorbing the radicals from water radiolysis in the cytosol and reducing the energy of IR. However, the bodyguard protection by heterochromatin has been challenged by the observation that DNA damage is less frequently detected in peripheral heterochromatin than in euchromatin, which is inconsistent with the expectation and simulation results. Previous studies have also shown that the DNA damage in peripheral heterochromatin is rarely repaired and moves more quickly, broadly and outwardly to approach the nuclear pore complex (NPC). Additionally, it has been shown that extrachromosomal circular DNAs (eccDNAs) are formed in the nucleus, highly detectable in the cytoplasm (particularly under stress conditions) and shuttle between the nucleus and the cytoplasm. Based on these studies, this review speculates that the sites of DNA damage in peripheral heterochromatin could occur more frequently and may be removed by repetitive elements in non-coding DNA through the formation of eccDNAs and expelled out of the nucleus to the cytoplasm via the NPC. Therefore, this review proposes that the genome and central protein-coding sequences are doubly protected by non-coding DNA in peripheral heterochromatin against DNA damage from radiation, which may be a novel protective role of non-coding DNA in genome defense.


Subject(s)
DNA/metabolism , Heterochromatin/genetics , Heterochromatin/radiation effects , Cell Nucleus/genetics , DNA/classification , DNA Damage , Euchromatin/genetics , Euchromatin/radiation effects , Genome , Radiation, Ionizing
11.
Proc Natl Acad Sci U S A ; 112(21): E2836-44, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25964332

ABSTRACT

The spatial organization of chromatin can be subject to extensive remodeling in plant somatic cells in response to developmental and environmental signals. However, the mechanisms controlling these dynamic changes and their functional impact on nuclear activity are poorly understood. Here, we determined that light perception triggers a switch between two different nuclear architectural schemes during Arabidopsis postembryonic development. Whereas progressive nucleus expansion and heterochromatin rearrangements in cotyledon cells are achieved similarly under light and dark conditions during germination, the later steps that lead to mature nuclear phenotypes are intimately associated with the photomorphogenic transition in an organ-specific manner. The light signaling integrators DE-ETIOLATED 1 and CONSTITUTIVE PHOTOMORPHOGENIC 1 maintain heterochromatin in a decondensed state in etiolated cotyledons. In contrast, under light conditions cryptochrome-mediated photoperception releases nuclear expansion and heterochromatin compaction within conspicuous chromocenters. For all tested loci, chromatin condensation during photomorphogenesis does not detectably rely on DNA methylation-based processes. Notwithstanding, the efficiency of transcriptional gene silencing may be impacted during the transition, as based on the reactivation of transposable element-driven reporter genes. Finally, we report that global engagement of RNA polymerase II in transcription is highly increased under light conditions, suggesting that cotyledon photomorphogenesis involves a transition from globally quiescent to more active transcriptional states. Given these findings, we propose that light-triggered changes in nuclear architecture underlie interplays between heterochromatin reorganization and transcriptional reprogramming associated with the establishment of photosynthesis.


Subject(s)
Arabidopsis/growth & development , Arabidopsis/radiation effects , Light Signal Transduction , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/radiation effects , Cotyledon/growth & development , Cotyledon/metabolism , Cotyledon/radiation effects , DNA Methylation , Gene Silencing , Genes, Plant , Heterochromatin/genetics , Heterochromatin/radiation effects , Intracellular Signaling Peptides and Proteins , Light Signal Transduction/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Plants, Genetically Modified , RNA Polymerase II/metabolism , Seedlings/growth & development , Seedlings/metabolism , Seedlings/radiation effects , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
12.
Acta Biochim Pol ; 62(2): 173-6, 2015.
Article in English | MEDLINE | ID: mdl-26015993

ABSTRACT

Histone modifications are involved in the DNA damage response (DDR). Here, by utilizing an ELISA immunoassay we assessed the methylation at H3K9 (H3K9me2 and H3K9me3) in two cell lines with differential sensitivity to radiation-induced apoptosis, HeLa (sensitive) and MCF-7 (resistant). We found that DNA damage induction by γ-irradiation leads to considerable accumulation (up to 5-fold) of H3K9me2 and H3K9me3, but not of H4K20me3 (control modification) in MCF-7 cells (p<0.05). Interestingly, a lower dose (2 Gy) was more effective than 5 Gy. In HeLa cells a smaller effect (approx. 1.5-1.8-fold) was evident only at 5 Gy. In conclusion, our findings reveal that DNA damage leads to specific accumulation of H3K9me2 and H3K9me3 in a cell-type specific manner.


Subject(s)
Heterochromatin/metabolism , Histones/metabolism , Radiation, Ionizing , DNA Damage/physiology , DNA Damage/radiation effects , Dose-Response Relationship, Radiation , HeLa Cells/metabolism , HeLa Cells/radiation effects , Heterochromatin/radiation effects , Humans , Lysine/metabolism , MCF-7 Cells/metabolism , MCF-7 Cells/radiation effects , Methylation , Radiation Tolerance , Tumor Suppressor Protein p53/metabolism
13.
Article in English | MEDLINE | ID: mdl-25813721

ABSTRACT

The purpose of this work is to test the hypothesis that kinetics of double strand breaks (DSB) repair is governed by complexity of DSB. To test the hypothesis we used our recent published mechanistic mathematical model of DSB repair for DSB induced by selected protons, deuterons, and helium ions of different energies representing radiations of different qualities. In light of recent advances in experimental and computational techniques, the most appropriate method to study cellular responses in radiation therapy, and exposures to low doses of ionizing radiations is using mechanistic approaches. To this end, we proposed a 'bottom-up' approach to study cellular response that starts with the DNA damage. Monte Carlo track structure method was employed to simulate initial damage induced in the genomic DNA by direct and indirect effects. Among the different types of DNA damage, DSB are known to be induced in simple and complex forms. The DSB repair model in G1 and early S phases of the cell cycle was employed to calculate the repair kinetics. The model considers the repair of simple and complex DSB, and the DSB produced in the heterochromatin. The inverse sampling method was used to calculate the repair kinetics for each individual DSB. The overall repair kinetics for 500 DSB induced by single tracks of the radiation under test were compared with experimental results. The results show that the model is capable of predicting the repair kinetics for the DSB induced by radiations of different qualities within an accepted range of uncertainty.


Subject(s)
Cell Cycle/radiation effects , DNA Breaks, Double-Stranded/radiation effects , G1 Phase/radiation effects , Radiation, Ionizing , S Phase/radiation effects , Animals , Cell Line , Cricetinae , DNA Repair/radiation effects , Heterochromatin/metabolism , Heterochromatin/radiation effects , Models, Molecular , Monte Carlo Method
14.
Mutat Res ; 750(1-2): 56-66, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23958412

ABSTRACT

Repair of double strand breaks (DSBs) is essential for cell survival and genome integrity. While much is known about the molecular mechanisms involved in DSB repair and checkpoint activation, the roles of nuclear dynamics of radiation-induced foci (RIF) in DNA repair are just beginning to emerge. Here, we summarize results from recent studies that point to distinct features of these dynamics in two different chromatin environments: heterochromatin and euchromatin. We also discuss how nuclear architecture and chromatin components might control these dynamics, and the need of novel quantification methods for a better description and interpretation of these phenomena. These studies are expected to provide new biomarkers for radiation risk and new strategies for cancer detection and treatment.


Subject(s)
Cell Nucleus/radiation effects , Euchromatin/radiation effects , Heterochromatin/radiation effects , Neoplasms, Radiation-Induced/genetics , Animals , Cell Nucleus/genetics , DNA Breaks, Double-Stranded , DNA Repair/physiology , Euchromatin/genetics , Heterochromatin/genetics , Humans , Kinetics , Neoplasms, Radiation-Induced/pathology
15.
Int J Radiat Biol ; 89(11): 898-906, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23692433

ABSTRACT

PURPOSE: With widespread use of ionizing radiation, more attention has been attracted to low-dose radiation (LDR); however, the mechanisms of long-term LDR-induced bio-effects are unclear. Here, we applied human B lymphoblast cell line HMy2.CIR to monitor the effects of long-term LDR and the potential involvement of DNA methylation. MATERIALS AND METHODS: HMy2.CIR cells were irradiated with 0.032 Gy γ-rays three times per week for 1-4 weeks. Some of these primed cells were further challenged with 2 Gy γ-rays. Cell proliferation, micronuclei formation, gene expression of DNA methyltransferases (DNMT), levels of global genomic DNA methylation and protein expression of methyl CpG binding protein 2 (MeCP2) and heterochromatin protein-1 (HP1) were measured. RESULTS: Long-term LDR enhanced cell proliferation and clonogenicity and triggered a cellular adaptive response (AR). Furthermore, global genomic DNA methylation was increased in HMy2.CIR cells after long-term LDR, accompanied with an increase of gene expression of DNMT1 and protein expression of MeCP2 and HP1. After treatment with 5-aza-2'-deoxycytidine (5-aza-dC), a DNA methyltransferase inhibitor, the long-term LDR-induced global genomic DNA hypermethylation was decreased and the AR was eliminated. CONCLUSION: Global genomic DNA hypermethylation accompanied with increases of DNMT1 and MeCP2 expression and heterochromatin formation might be involved in long-term LDR-induced adaptive response.


Subject(s)
Adaptation, Physiological/radiation effects , B-Lymphocytes/radiation effects , DNA Methylation/radiation effects , Gamma Rays/adverse effects , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , Decitabine , Dose-Response Relationship, Radiation , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Heterochromatin/drug effects , Heterochromatin/metabolism , Heterochromatin/radiation effects , Humans , Radiation Tolerance/drug effects
16.
Proc Natl Acad Sci U S A ; 110(3): E212-20, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23267107

ABSTRACT

Specific mutations in the XPD subunit of transcription factor IIH result in combined xeroderma pigmentosum (XP)/Cockayne syndrome (CS), a severe DNA repair disorder characterized at the cellular level by a transcriptional arrest following UV irradiation. This transcriptional arrest has always been thought to be the result of faulty transcription-coupled repair. In the present study, we showed that, following UV irradiation, XP-D/CS cells displayed a gross transcriptional dysregulation compared with "pure" XP-D cells or WT cells. Furthermore, global RNA-sequencing analysis showed that XP-D/CS cells repressed the majority of genes after UV, whereas pure XP-D cells did not. By using housekeeping genes as a model, we demonstrated that XP-D/CS cells were unable to reassemble these gene promoters and thus to restart transcription after UV irradiation. Furthermore, we found that the repression of these promoters in XP-D/CS cells was not a simple consequence of deficient repair but rather an active heterochromatinization process mediated by the histone deacetylase Sirt1. Indeed, RNA-sequencing analysis showed that inhibition of and/or silencing of Sirt1 changed the chromatin environment at these promoters and restored the transcription of a large portion of the repressed genes in XP-D/CS cells after UV irradiation. Our work demonstrates that a significant part of the transcriptional arrest displayed by XP-D/CS cells arises as a result of an active repression process and not simply as a result of a DNA repair deficiency. This dysregulation of Sirt1 function that results in transcriptional repression may be the cause of various severe clinical features in patients with XP-D/CS that cannot be explained by a DNA repair defect.


Subject(s)
Cockayne Syndrome/genetics , Cockayne Syndrome/metabolism , RNA/biosynthesis , Sirtuin 1/metabolism , Xeroderma Pigmentosum Group D Protein/genetics , Xeroderma Pigmentosum Group D Protein/metabolism , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism , Cells, Cultured , Cockayne Syndrome/complications , DNA Repair , Heterochromatin/genetics , Heterochromatin/metabolism , Heterochromatin/radiation effects , Humans , Mutation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Transcription Factor TFIIH/chemistry , Transcription Factor TFIIH/genetics , Transcription Factor TFIIH/metabolism , Transcription, Genetic/radiation effects , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/complications , Xeroderma Pigmentosum Group D Protein/chemistry
17.
PLoS One ; 7(5): e38165, 2012.
Article in English | MEDLINE | ID: mdl-22666473

ABSTRACT

PURPOSE: DNA double-strand breaks (DSBs) generated by ionizing radiation pose a serious threat to the preservation of genetic and epigenetic information. The known importance of local chromatin configuration in DSB repair raises the question of whether breaks in different chromatin environments are recognized and repaired by the same repair machinery and with similar efficiency. An essential step in DSB processing by non-homologous end joining is the high-affinity binding of Ku70-Ku80 and DNA-PKcs to double-stranded DNA ends that holds the ends in physical proximity for subsequent repair. METHODS AND MATERIALS: Using transmission electron microscopy to localize gold-labeled pKu70 and pDNA-PKcs within nuclear ultrastructure, we monitored the formation and repair of actual DSBs within euchromatin (electron-lucent) and heterochromatin (electron-dense) in cortical neurons of irradiated mouse brain. RESULTS: While DNA lesions in euchromatin (characterized by two pKu70-gold beads, reflecting the Ku70-Ku80 heterodimer) are promptly sensed and rejoined, DNA packaging in heterochromatin appears to retard DSB processing, due to the time needed to unravel higher-order chromatin structures. Complex pKu70-clusters formed in heterochromatin (consisting of 4 or ≥ 6 gold beads) may represent multiple breaks in close proximity caused by ionizing radiation of highly-compacted DNA. All pKu70-clusters disappeared within 72 hours post-irradiation, indicating efficient DSB rejoining. However, persistent 53BP1 clusters in heterochromatin (comprising ≥ 10 gold beads), occasionally co-localizing with γH2AX, but not pKu70 or pDNA-PKcs, may reflect incomplete or incorrect restoration of chromatin structure rather than persistently unrepaired DNA damage. DISCUSSION: Higher-order organization of chromatin determines the accessibility of DNA lesions to repair complexes, defining how readily DSBs are detected and processed. DNA lesions in heterochromatin appear to be more complex, with multiple breaks in spatial vicinity inducing severe chromatin disruptions. Imperfect restoration of chromatin configurations may leave DSB-induced epigenetic memory of damage with potentially pathological repercussions.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Euchromatin/genetics , Euchromatin/ultrastructure , Heterochromatin/genetics , Heterochromatin/ultrastructure , Microscopy, Electron, Transmission , Animals , Cerebral Cortex/cytology , Chromosomal Proteins, Non-Histone/metabolism , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins/metabolism , Euchromatin/metabolism , Euchromatin/radiation effects , Heterochromatin/metabolism , Heterochromatin/radiation effects , Mice , Neurons/cytology , Neurons/radiation effects , Neurons/ultrastructure , Tumor Suppressor p53-Binding Protein 1
18.
Proc Natl Acad Sci U S A ; 108(29): E314-22, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21715654

ABSTRACT

Prep1 is a homeodomain transcription factor that is essential in embryonic development and functions in the adult as a tumor suppressor. We show here that Prep1 is involved in maintaining genomic stability and preventing neoplastic transformation. Hypomorphic homozygous Prep1(i/i) fetal liver cells and mouse embryonic fibroblasts (MEFs) exhibit increased basal DNA damage and normal DNA damage response after γ-irradiation compared with WT. Cytogenetic analysis shows the presence of numerous chromosomal aberrations and aneuploidy in very early-passage Prep1(i/i) MEFs. In human fibroblasts, acute Prep1 down-regulation by siRNA induces DNA damage response, like in Prep1(i/i) MEFs, together with an increase in heterochromatin-associated modifications: rapid increase of histone methylation and decreased transcription of satellite DNA. Ectopic expression of Prep1 rescues DNA damage and heterochromatin methylation. Inhibition of Suv39 activity blocks the chromatin but not the DNA damage phenotype. Finally, Prep1 deficiency facilitates cell immortalization, escape from oncogene-induced senescence, and H-Ras(V12)-dependent transformation. Importantly, the latter can be partially rescued by restoration of Prep1 level. The results show that the tumor suppressor role of Prep1 is associated with the maintenance of genomic stability.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation/physiology , Genomic Instability/physiology , Homeodomain Proteins/physiology , Transcription Factors/physiology , Animals , Chromatin Immunoprecipitation , Comet Assay , Cytogenetic Analysis , DNA Damage/genetics , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Gamma Rays , Gene Expression Regulation/genetics , Heterochromatin/genetics , Heterochromatin/radiation effects , Homeodomain Proteins/metabolism , Humans , Mice , Oligonucleotides/genetics , Transcription Factors/metabolism
19.
DNA Repair (Amst) ; 10(2): 119-25, 2011 Feb 07.
Article in English | MEDLINE | ID: mdl-21130713

ABSTRACT

Due to its link with human pathologies, including cancer, the mechanism of Nucleotide Excision Repair (NER) has been extensively studied. Most of the pathway and players have been defined using in vitro reconstitution experiments. However, in vivo, the NER machinery must deal with the presence of organized chromatin, which in some regions, such as heterochromatin, is highly condensed but still susceptible to DNA damage. A series of events involving different chromatin-remodeling factors and histone-modifying enzymes target chromatin regions that contain DNA lesions. CPDs change the structure of the nucleosome, allowing access to factors that can recognize the lesion. Next, DDB1-DDB2 protein complexes, which mono-ubiquitinate histones H2A, H3, and H4, recognize nucleosomes containing DNA lesions. The ubiquitinated nucleosome facilitates the recruitment of ATP-dependent chromatin-remodeling factors and the XPC-HR23B-Centrin 2 complex to the target region. Different ATP-dependent chromatin-remodeling factors, such as SWI/SNF and INO80, have been identified as having roles in the UV irradiation response prior to the action of the NER machinery. Subsequently, remodeling of the nucleosome allows enzymatic reactions by histone-modifying factors that may acetylate, methylate or demethylate specific histone residues. Intriguingly, some of these histone modifications are dependent on p53. These histone modifications and the remodeling of the nucleosome allow the entrance of TFIIH, XPC and other NER factors that remove the damaged strand; then, gap-filling DNA synthesis and ligation reactions are carried out after excision of the oligonucleotide with the lesion. Finally, after DNA repair, the initial chromatin structure has to be reestablished. Therefore, factors that modulate chromatin dynamics contribute to the NER mechanism, and they are significant in the future design of treatments for human pathologies related to genome instability and the appearance of drug-resistant tumors.


Subject(s)
Chromatin Assembly and Disassembly/drug effects , Chromatin/genetics , Chromatin/radiation effects , DNA Damage , DNA Repair , Ultraviolet Rays , Chromatin/chemistry , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Deoxyribodipyrimidine Photo-Lyase/metabolism , Genes, p53 , Heterochromatin/chemistry , Heterochromatin/genetics , Heterochromatin/radiation effects , Histones/chemistry , Histones/genetics , Histones/radiation effects , Humans , Nucleosomes/genetics , Nucleosomes/radiation effects , Pyrimidine Dimers/chemistry , Pyrimidine Dimers/metabolism
20.
J Zhejiang Univ Sci B ; 11(10): 801-5, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20872988

ABSTRACT

To investigate the influence of microwave radiation on the human fibroblast nuclei, the effects of three variants of electromagnetic wave polarization, linear and left-handed and right-handed elliptically polarized, were examined. Experimental conditions were: frequency (f) 36.65 GHz, power density (P) at the surface of exposed object 1, 10, 30, and 100 µW/cm(2), exposure time 10 s. Human fibroblasts growing in a monolayer on a cover slide were exposed to microwave electromagnetic radiation. The layer of medium that covered cells during microwave exposure was about 1 mm thick. Cells were stained immediately after irradiation by 2% (w/v) orcein solution in 45% (w/v) acetic acid. Experiments were made at room temperature (25 °C), and control cell samples were processed in the same conditions. We assessed heterochromatin granule quantity (HGQ) at 600× magnification. Microwave irradiation at the intensity of 1 µW/cm(2) produced no effect, and irradiation at the intensities of 10 and 100 µW/cm(2) induced an increase in HGQ. More intense irradiation induced more chromatin condensation. The right-handed elliptically polarized radiation revealed more biological activity than the left-handed polarized one.


Subject(s)
Cell Nucleus/radiation effects , Fibroblasts/radiation effects , Microwaves , Cell Nucleus/ultrastructure , Cells, Cultured , Fibroblasts/ultrastructure , Heterochromatin/radiation effects , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...