Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters










Publication year range
1.
J Radiat Res ; 65(3): 263-271, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38461549

ABSTRACT

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.


Subject(s)
Adenosine Triphosphate , BRCA1 Protein , Homologous Recombination , Rad51 Recombinase , Radiation, Ionizing , Humans , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/biosynthesis , Homologous Recombination/radiation effects , Rad51 Recombinase/metabolism , BRCA1 Protein/metabolism , DNA Breaks, Double-Stranded/radiation effects , Replication Protein A/metabolism , Cell Line, Tumor , Intracellular Space/metabolism , Intracellular Space/radiation effects , DNA Repair , Histones/metabolism
2.
Int J Mol Sci ; 22(20)2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34681628

ABSTRACT

The processing of DNA double-strand breaks (DSBs) depends on the dynamic characteristics of chromatin. To investigate how abrupt changes in chromatin compaction alter these dynamics and affect DSB processing and repair, we exposed irradiated cells to hypotonic stress (HypoS). Densitometric and chromosome-length analyses show that HypoS transiently decompacts chromatin without inducing histone modifications known from regulated local chromatin decondensation, or changes in Micrococcal Nuclease (MNase) sensitivity. HypoS leaves undisturbed initial stages of DNA-damage-response (DDR), such as radiation-induced ATM activation and H2AX-phosphorylation. However, detection of ATM-pS1981, γ-H2AX and 53BP1 foci is reduced in a protein, cell cycle phase and cell line dependent manner; likely secondary to chromatin decompaction that disrupts the focal organization of DDR proteins. While HypoS only exerts small effects on classical nonhomologous end-joining (c-NHEJ) and alternative end-joining (alt-EJ), it markedly suppresses homologous recombination (HR) without affecting DNA end-resection at DSBs, and clearly enhances single-strand annealing (SSA). These shifts in pathway engagement are accompanied by decreases in HR-dependent chromatid-break repair in the G2-phase, and by increases in alt-EJ and SSA-dependent chromosomal translocations. Consequently, HypoS sensitizes cells to ionizing radiation (IR)-induced killing. We conclude that HypoS-induced global chromatin decompaction compromises regulated chromatin dynamics and genomic stability by suppressing DSB-processing by HR, and allowing error-prone processing by alt-EJ and SSA.


Subject(s)
Chromatin/metabolism , DNA End-Joining Repair/drug effects , Homologous Recombination/drug effects , Hypotonic Solutions/pharmacology , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Line , Cell Proliferation/drug effects , Chromatin/chemistry , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair/radiation effects , Histones/metabolism , Homologous Recombination/radiation effects , Humans , Hypotonic Solutions/chemistry , RNA Interference , RNA, Small Interfering/metabolism , Rad52 DNA Repair and Recombination Protein/antagonists & inhibitors , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Radiation, Ionizing
3.
J Genet Genomics ; 48(4): 333-340, 2021 04 20.
Article in English | MEDLINE | ID: mdl-34039517

ABSTRACT

Repair of DNA double-strand break (DSB) is critical for the maintenance of genome integrity. A class of DSB-induced small RNAs (diRNAs) has been shown to play an important role in DSB repair. In humans, diRNAs are associated with Ago2 and guide the recruitment of Rad51 to DSB sites to facilitate repair by homologous recombination (HR). Ago2 activity has been reported to be regulated by phosphorylation under normal and hypoxic conditions. However, the role of Ago2 phosphorylation in DNA damage repair is unexplored. Here, we show that S672, S828, T830, and S831 of human Ago2 are phosphorylated in response to ionizing radiation (IR). S672A mutation of Ago2 leads to significant reduction in Rad51 foci formation and HR efficiency. We further show that defective association of Ago2 S672A variant with DSB sites, instead of defects in diRNA and Rad51 binding, may account for decreased Rad51 foci formation and HR efficiency. Our study reveals a novel regulatory mechanism for the function of Ago2 in DNA repair.


Subject(s)
Argonaute Proteins/genetics , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , Rad51 Recombinase/genetics , Amino Acids/genetics , Amino Acids/radiation effects , DNA/genetics , DNA/radiation effects , DNA Damage/genetics , DNA Damage/radiation effects , DNA Repair/genetics , Genome/genetics , Genome/radiation effects , Homologous Recombination/radiation effects , Humans , Phosphorylation/radiation effects , Protein Binding/genetics , RNA/genetics , RNA/radiation effects , Radiation, Ionizing
4.
Radiat Res ; 195(5): 441-451, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33721021

ABSTRACT

We examined lethal damages of X rays induced by direct and indirect actions, in terms of double-strand break (DSB) repair susceptibility using two kinds of repair-deficient Chinese hamster ovary (CHO) cell lines. These CHO mutants (51D1 and xrs6) are genetically deficient in one of the two important DNA repair pathways after genotoxic injury [homologous recombination (HR) and non-homologous end binding (NHEJ) pathways, respectively]. The contribution of indirect action on cell killing can be estimated by applying the maximum level of dimethylsulfoxide (DMSO) to get rid of OH radicals. To control the proportion of direct and indirect actions in lethal damage, we irradiated CHO mutant cells under aerobic and anoxic conditions. The contributions of indirect action on HR-defective 51D1 cells were 76% and 57% under aerobic and anoxic conditions, respectively. Interestingly, these percentages were similar to those of the wild-type cells even if the radiosensitivity was different. However, the contributions of indirect action to cell killing on NHEJ-defective xrs6 cells were 52% and 33% under aerobic and anoxic conditions, respectively. Cell killing by indirect action was significantly affected by the oxygen concentration and the DSB repair pathways but was not correlated with radiosensitivity. These results suggest that the lethal damage induced by direct action is mostly repaired by NHEJ repair pathway since killing of NHEJ-defective cells has significantly higher contribution by the direct action. In other words, the HR repair pathway may not effectively repair the DSB by direct action in place of the NHEJ repair pathway. We conclude that the type of DSB produced by direct action is different from that of DSB induced by indirect action.


Subject(s)
DNA Damage , Oxygen/metabolism , Aerobiosis/genetics , Aerobiosis/radiation effects , Animals , CHO Cells , Cell Death/genetics , Cell Death/radiation effects , Cricetulus , DNA End-Joining Repair/radiation effects , Homologous Recombination/radiation effects , X-Rays/adverse effects
5.
BMC Cancer ; 20(1): 775, 2020 Aug 18.
Article in English | MEDLINE | ID: mdl-32811446

ABSTRACT

BACKGROUND: PARP inhibitors niraparib and talazoparib are FDA approved for special cases of breast cancer. PARP is an interesting repair protein which is frequently affected in cancer cells. We studied the combined action of talazoparib or niraparib with ionizing radiation in melanoma cells and healthy fibroblasts. METHODS: Homologous recombination (HR) status in six different melanoma cell lines and healthy fibroblasts was assessed. Cell cultures were treated with PARP inhibitors talazoparib or niraparib and ionizing radiation (IR). Apoptosis, necrosis and cell cycle distribution was analyzed via flow cytometry. Cell migration was studied by scratch assays. RESULTS: Studied melanoma cell cultures are HR deficient. Studied healthy fibroblasts are HR proficient. Talazoparib and niraparib have congruent effects within the same cell cultures. In all cell cultures, combined treatment increases cell death and G2/M arrest compared to IR. Combined treatment in melanoma cells distinctly increases G2/M arrest. Healthy fibroblasts are less affected by G2/M arrest. Treatment predominantly decelerates or does not modify migration. In two cell cultures migration is enhanced under the inhibitors. CONCLUSIONS: Although the two PARP inhibitors talazoparib and niraparib appear to be suitable for a combination treatment with ionizing radiation in our in vitro studies, a combination treatment cannot generally be recommended. There are clear interindividual differences in the effect of the inhibitors on different melanoma cells. Therefore, the effect on the cancer cells should be studied prior to a combination therapy. Since melanoma cells increase more strongly than fibroblasts in G2/M arrest, the fractional application of combined treatment should be further investigated.


Subject(s)
Chemoradiotherapy/methods , Fibroblasts/drug effects , Melanoma/therapy , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Skin Neoplasms/therapy , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Survival/drug effects , Chemoradiotherapy/adverse effects , Drug Interactions , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , Homologous Recombination/drug effects , Homologous Recombination/radiation effects , Humans , Indazoles/pharmacology , Indazoles/therapeutic use , Melanoma/genetics , Melanoma/pathology , Phthalazines/pharmacology , Phthalazines/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Primary Cell Culture , Skin/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tumor Cells, Cultured
6.
Methods Mol Biol ; 2102: 441-457, 2020.
Article in English | MEDLINE | ID: mdl-31989571

ABSTRACT

A fully optimized staining method for detecting sister chromatid exchanges in cultured cells is presented. The method gives reproducibly robust quantitative results. Sister chromatid exchange is a classic toxicology assay for genotoxicity and for detecting alterations to the biochemistry underlying cellular homologous recombination. Growth of cells in the presence of 5'-bromo-deoxyuridine for two rounds of DNA replication followed by collecting metaphase spreads on glass slides, treatment with the UV-sensitive dye Hoechst 33258, long-wave UV light exposure, and Giemsa staining gives a permanent record of the exchanges.


Subject(s)
Metaphase , Mutagenicity Tests/methods , Sister Chromatid Exchange , Azure Stains , Biological Assay/methods , Bisbenzimidazole , Bromodeoxyuridine/metabolism , Cells, Cultured , Chromatids/drug effects , Chromatids/metabolism , Chromatids/radiation effects , Chromosomes/drug effects , Chromosomes/metabolism , Chromosomes/radiation effects , Homologous Recombination/drug effects , Homologous Recombination/radiation effects , Humans , Metaphase/drug effects , Metaphase/radiation effects , Workflow
7.
Proc Natl Acad Sci U S A ; 116(45): 22609-22618, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31591209

ABSTRACT

A minority of cancers have breast cancer gene (BRCA) mutations that confer sensitivity to poly (ADP-ribose) polymerase (PARP) inhibitors (PARPis), but the role for PARPis in BRCA-proficient cancers is not well established. This suggests the need for novel combination therapies to expand the use of these drugs. Recent reports that low doses of DNA methyltransferase inhibitors (DNMTis) plus PARPis enhance PARPi efficacy in BRCA-proficient AML subtypes, breast, and ovarian cancer open up the possibility that this strategy may apply to other sporadic cancers. We identify a key mechanistic aspect of this combination therapy in nonsmall cell lung cancer (NSCLC): that the DNMTi component creates a BRCAness phenotype through downregulating expression of key homologous recombination and nonhomologous end-joining (NHEJ) genes. Importantly, from a translational perspective, the above changes in DNA repair processes allow our combinatorial PARPi and DNMTi therapy to robustly sensitize NSCLC cells to ionizing radiation in vitro and in vivo. Our combinatorial approach introduces a biomarker strategy and a potential therapy paradigm for treating BRCA-proficient cancers like NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , DNA Modification Methylases/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Animals , Antineoplastic Agents , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Combined Modality Therapy , DNA Modification Methylases/metabolism , DNA Repair/drug effects , DNA Repair/radiation effects , Drug Therapy, Combination , Female , Homologous Recombination/drug effects , Homologous Recombination/radiation effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Phthalazines/administration & dosage , Radiation, Ionizing
8.
Radiother Oncol ; 133: 77-86, 2019 04.
Article in English | MEDLINE | ID: mdl-30935585

ABSTRACT

BACKGROUND AND PURPOSE: Carbon ion radiotherapy is a promising therapeutic option for glioblastoma patients due to its high physical dose conformity and greater biological effectiveness than photons. However, the biological effects of carbon ion radiation are still incompletely understood. Here, we systematically compared the biological effects of clinically used carbon ion radiation to photon radiation with emphasis on DNA repair. MATERIALS AND METHODS: Two human glioblastoma cell lines (U87 and LN229) were irradiated with carbon ions or photons and DNA damage response was systematically analyzed, including clonogenic survival, induction and repair of DNA double-strand breaks (DSBs), cell cycle arrest and apoptosis or autophagy. γH2AX foci were analyzed by flow cytometry, conventional light microscopy and 3D superresolution microscopy. RESULTS: DSBs were repaired delayed and with slower kinetics after carbon ions versus photons. Carbon ions caused stronger and longer-lasting cell cycle delays, predominantly in G2 phase, and a higher rate of apoptosis. Compared to photons, the effectiveness of carbon ions was less cell cycle-dependent. Homologous recombination (HR) appeared to be more important for DSB repair after carbon ions versus photons in phosphatase and tensin homolog (PTEN)-deficient U87 cells, as opposed to PTEN-proficient LN229 cells. CONCLUSION: Carbon ions induced more severe DSB damage than photons, which was repaired less efficiently in both cell lines. Thus, carbon ion radiotherapy may help to overcome resistance mechanisms of glioblastoma associated with DNA repair for example in combination with repair pathway-specific drugs in the context of personalized radiotherapy.


Subject(s)
Brain Neoplasms/radiotherapy , DNA Breaks, Double-Stranded , Glioblastoma/radiotherapy , Heavy Ion Radiotherapy/methods , Photons/therapeutic use , Apoptosis/radiation effects , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , DNA Repair/radiation effects , DNA, Neoplasm/genetics , DNA, Neoplasm/radiation effects , Glioblastoma/genetics , Glioblastoma/pathology , Homologous Recombination/radiation effects , Humans
9.
Radiat Res ; 191(5): 398-412, 2019 05.
Article in English | MEDLINE | ID: mdl-30835165

ABSTRACT

Sirtuin 2 (SIRT2) plays a major role in aging, carcinogenesis and neurodegeneration. While it has been shown that SIRT2 is a mediator of stress-induced cell death, the mechanism remains unclear. In this study, we report the role of SIRT2 in mediating radiation-induced cell death and DNA damage using mouse embryonic fibroblasts (MEFs), progenitor cells and tissues from Sirt2 wild-type and genomic knockout mice, and human tumor and primary cell lines as models. The presence of Sirt2 in cells and tissues significantly enhanced the cell's sensitivity to radiation-induced cytotoxicity by delaying the dispersion of radiation-induced γ-H2AX and 53BP1 foci. This enhanced cellular radiosensitivity correlated with reduced expression of pro-survival and DNA repair proteins, and decreased DNA repair capacities involving both homologous repair and non-homologous end joining DNA repair mechanisms compared to those in Sirt2 knockout (KO) and knockdown (KD) phenotypes. Together, these data suggest SIRT2 plays a critical role in mediating the radiation-induced DNA damage response, thus regulating radiation-induced cell death and survival.


Subject(s)
Radiation Injuries, Experimental/metabolism , Sirtuin 2/metabolism , Animals , Cell Line , Cell Survival/radiation effects , Cognition/radiation effects , DNA Damage , Fibroblasts/radiation effects , Homologous Recombination/radiation effects , Mice , Radiation Injuries, Experimental/genetics , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/physiopathology , Radiation Tolerance
10.
Invest New Drugs ; 37(6): 1158-1165, 2019 12.
Article in English | MEDLINE | ID: mdl-30793218

ABSTRACT

Purpose To elucidate the radiosensitizing effect and underlying mechanism of a new kind of DNA methyltransferase (DNMT) inhibitor with biological availability. Methods A novel non-nucleoside compound, designated as MA-17, was recently derived from a phthalimido alkanamide structure. DNMT expressions were confirmed in cultured human lung cancer (A549) and normal astrocyte (NHA) cells, radiosensitivity was measured using clonogenic assay, and assays of cell cycle alteration, apoptosis, DNA damage repair, and differential gene expression were undertaken. Results MA-17 significantly radiosensitized A549 cells with a mean dose enhancement ratio (DER) of 1.43 at the surviving fraction of 0.2 (p < 0.05 by one-tailed ratio paired t-test). MA-17 did not affect normal astrocytes (mean DER0.2, 1.016; p = 0.420). MA-17 demonstrated a mean half-life of 1.0 h in vivo and a relatively even distribution in various tissues. Pretreatment with MA-17 increased sub-G1 fractions and inhibited the repair of DNA double-strand breaks, which are induced by irradiation. We found that MA-17 also down-regulated DNA homologous recombination and the Fanconi anemia pathway (FANCA, BRCA1, and RAD51C) in A549 cells. This bioinformatics finding was confirmed in validation Western blot to evaluate the expression of vital proteins. Conclusions A novel phthalimido alkanamide derivative, a DNMT inhibitor, possessed both biostability and favorable and substantial radiosensitizing effects by augmenting apoptosis or inhibiting DNA damage repair.


Subject(s)
DNA Modification Methylases/antagonists & inhibitors , Phthalimides/pharmacology , Radiation-Sensitizing Agents/pharmacology , A549 Cells , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage , DNA Modification Methylases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Homologous Recombination/drug effects , Homologous Recombination/radiation effects , Humans , Radiation Tolerance/drug effects , X-Rays
11.
DNA Repair (Amst) ; 74: 70-79, 2019 02.
Article in English | MEDLINE | ID: mdl-30606609

ABSTRACT

DSBs are harmful lesions produced through endogenous metabolism or by exogenous agents such as ionizing radiation, that can trigger genomic rearrangements. We have recently shown that exposure to 2 Gy of X-rays has opposite effects on the induction of Shh-dependent MB in NHEJ- and HR-deficient Ptch1+/- mice. In the current study we provide a comprehensive link on the role of HR/NHEJ at low doses (0.042 and 0.25 Gy) from the early molecular changes through DNA damage processing, up to the late consequences of their inactivation on tumorigenesis. Our data indicate a prominent role for HR in genome stability, by preventing spontaneous and radiation-induced oncogenic damage in neural precursors of the cerebellum, the cell of origin of MB. Instead, loss of DNA-PKcs function increased DSBs and apoptosis in neural precursors of the developing cerebellum, leading to killing of tumor initiating cells, and suppression of MB tumorigenesis in DNA-PKcs-/-/Ptch1+/- mice. Pathway analysis demonstrates that DNA-PKcs genetic inactivation confers a remarkable radiation hypersensitivity, as even extremely low radiation doses may deregulate many DDR genes, also triggering p53 pathway activation and cell cycle arrest. Finally, by showing that DNA-PKcs inhibition by NU7441 radiosensitizes human MB cells, our in vitro findings suggest the inclusion of MB in the list of tumors beneficiating from the combination of radiotherapy and DNA-PKcs targeting, holding promise for clinical translation.


Subject(s)
Cerebellar Neoplasms/genetics , DNA Repair/radiation effects , Medulloblastoma/genetics , Neoplasms, Radiation-Induced/genetics , Patched-1 Receptor/deficiency , Patched-1 Receptor/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/radiation effects , Cell Line, Tumor , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/therapy , DNA Damage , DNA End-Joining Repair/radiation effects , DNA Helicases/genetics , DNA-Activated Protein Kinase/deficiency , DNA-Binding Proteins/deficiency , Dose-Response Relationship, Radiation , Homologous Recombination/radiation effects , Humans , Medulloblastoma/metabolism , Medulloblastoma/pathology , Medulloblastoma/therapy , Mice , Molecular Targeted Therapy , Mutation , Neoplasms, Radiation-Induced/metabolism , Neoplasms, Radiation-Induced/pathology , Neoplasms, Radiation-Induced/therapy , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Risk , X-Rays/adverse effects
12.
New Phytol ; 222(3): 1380-1391, 2019 05.
Article in English | MEDLINE | ID: mdl-30636294

ABSTRACT

Double-stranded breaks can be repaired by different mechanisms such as homologous recombination (HR), classical nonhomologous end joining (C-NHEJ) and alternative end joining (Alt-EJ). Polymerase Q (POLQ) has been proposed to be the main factor involved in Alt-EJ-mediated DNA repair. Here we describe the role of POLQ in DNA repair and gene targeting in Physcomitrella patens. The disruption of the POLQ gene does not influence the genetic stability of P. patens nor its development. The polq mutant shows the same sensitivity as wild-type towards most of the genotoxic agents tested (ultraviolet (UV), methyl methanesulfonate (MMS) and cisplatin) with the notable exception of bleomycin for which it shows less sensitivity than the wild-type. Furthermore, we show that POLQ is involved in the repair of CRISPR-Cas9-induced double-stranded breaks in P. patens. We also demonstrate that POLQ is a potential competitor and/or inhibitor of the HR repair pathway. This finding has a consequence in terms of genetic engineering, as in the absence of POLQ the frequency of gene targeting is significantly increased and the number of clean two-sided HR-mediated insertions is enhanced. Therefore, the control of POLQ activity in plants could be a useful strategy to optimize the tools of genome engineering for plant breeding.


Subject(s)
Bryopsida/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , DNA Breaks, Double-Stranded , DNA Repair , DNA-Directed DNA Polymerase/metabolism , Base Sequence , Bleomycin/pharmacology , Bryopsida/drug effects , Bryopsida/radiation effects , Cisplatin/pharmacology , DNA End-Joining Repair , DNA-Directed DNA Polymerase/genetics , Genomic Instability , Homologous Recombination/drug effects , Homologous Recombination/radiation effects , Methyl Methanesulfonate/pharmacology , Mutation/genetics , Mutation Rate , Phenotype , Ultraviolet Rays , DNA Polymerase theta
13.
J Radiat Res ; 60(1): 37-50, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30423138

ABSTRACT

Radiotherapy is a common modality for treatment of brain cancers, but it can induce long-term physiological and cognitive deficits. The responses of normal human brain cells to radiation is not well understood. Astrocytes have been shown to have a variety of protective mechanisms against oxidative stress and have been shown to protect neurons. We investigated the response of cultured normal human astrocytes (NHAs) to X-ray irradiation. Following exposure to 10 Gy X-irradiation, NHAs exhibited DNA damage as indicated by the formation of γ-H2AX foci. Western blotting showed that NHAs displayed a robust increase in expression of non-homologous end joining DNA repair enzymes within 15 min post-irradiation and increased expression of homologous recombination DNA repair enzymes ~2 h post-irradiation. The cell cycle checkpoint protein p21/waf1 was upregulated from 6-24 h, and then returned to baseline. Levels of DNA repair enzymes returned to basal ~48 h post-irradiation. NHAs re-entered the cell cycle and proliferation was observed at 6 days. In contrast, normal human mesenchymal stem cells (MSCs) failed to upregulate DNA repair enzymes and instead displayed sustained upregulation of p21/waf1, a cell cycle checkpoint marker for senescence. Ectopic overexpression of Ku70 was sufficient to protect MSCs from sustained upregulation of p21/waf1 induced by 10 Gy X-rays. These findings suggest that increased expression of Ku70 may be a key mechanism for the radioresistance of NHAs, preventing their accelerated senescence from high-dose radiation. These results may have implications for the development of novel targets for radiation countermeasure development.


Subject(s)
Astrocytes/radiation effects , DNA End-Joining Repair , Radiation Tolerance , Apoptosis/radiation effects , Astrocytes/cytology , Astrocytes/metabolism , Cell Cycle Checkpoints/radiation effects , Cell Proliferation/radiation effects , Cells, Cultured , Cellular Senescence/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoprotection/radiation effects , DNA End-Joining Repair/radiation effects , HEK293 Cells , Homologous Recombination/radiation effects , Humans , Ku Autoantigen/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/radiation effects , Radiation Exposure , Radiation Tolerance/radiation effects , X-Rays
14.
Sci Rep ; 8(1): 15025, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30302025

ABSTRACT

Genomes are mostly protected from constant DNA-damaging threats, either internal or external, which ultimately sustain the organism. Herein, we report that AIMP3, a previously demonstrated tumour suppressor, plays an essential role in maintaining genome integrity in adult mice. Upon induction of the temporal systemic deletion of AIMP3 by tamoxifen in adult mice, the animals developed an acute radiation syndrome-like phenotype, typified by scleroderma, hypotrophy of haematopoietic cells and organs, and intestinal failure. Induction of γH2AX, an early marker of DNA double-strand breaks, was observed in the spleen, intestine, and the highly replicating embryonic cortex. In addition, sub-lethal irradiation of AIMP3 mKO mice dramatically affected organ damage and survival. Using isolated MEFs from conditional KO mice or AIMP3 knockdown cells, we confirmed the presence of spontaneously occurring DNA double-strand breaks by COMET assay and γH2AX induction. Furthermore, γH2AX removal was delayed, and homologous DNA repair activity was significantly reduced. Reduction of RPA foci formation and subsequent Rad51 foci formation probably underlie the significant reduction in homologous recombination activity in the absence of AIMP3. Together, our data demonstrate that AIMP3 plays a role in genome stability through the DNA repair process.


Subject(s)
Acute Radiation Syndrome/genetics , Histones/genetics , Peptide Elongation Factors/genetics , Rad51 Recombinase/genetics , Acute Radiation Syndrome/pathology , Animals , Comet Assay , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Fibroblasts/radiation effects , Genomic Instability/radiation effects , Homologous Recombination/radiation effects , Humans , Mice , Mice, Knockout , Phenotype , Radiation , Radiation, Ionizing , Tumor Suppressor Proteins/genetics
15.
Sci Rep ; 8(1): 5405, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29599445

ABSTRACT

One of the most important mechanisms for repairing double-strand breaks (DSBs) in model eukaryotes is homologous recombination (HR). Although the genes involved in HR have been found in Trypanosoma brucei and studies have identified some of the proteins that participate in this HR pathway, the recruitment kinetics of the HR machinery onto DNA during DSB repair have not been clearly elucidated in this organism. Using immunofluorescence, protein DNA-bound assays, and DNA content analysis, we established the recruitment kinetics of the HR pathway in response to the DSBs generated by ionizing radiation (IR) in procyclic forms of T. brucei. These kinetics involved the phosphorylation of histone H2A and the sequential recruitment of the essential HR players Exo1, RPA, and Rad51. The process of DSB repair took approximately 5.5 hours. We found that DSBs led to a decline in the G2/M phase after IR treatment, concomitant with cell cycle arrest in the G1/S phase. This finding suggests that HR repairs DSBs faster than the other possible DSB repair processes that act during the G1/S transition. Taken together, these data suggest that the interplay between DNA damage detection and HR machinery recruitment is finely coordinated, allowing these parasites to repair DNA rapidly after DSBs during the late S/G2 proficient phases.


Subject(s)
Homologous Recombination/radiation effects , Radiation, Ionizing , Trypanosoma brucei brucei/metabolism , DNA Fragmentation/radiation effects , G1 Phase Cell Cycle Checkpoints/radiation effects , Histones/metabolism , Phosphorylation/radiation effects , Protozoan Proteins/metabolism , Recombinational DNA Repair/radiation effects , Replication Protein A/genetics , Replication Protein A/metabolism , S Phase Cell Cycle Checkpoints/radiation effects , Trypanosoma brucei brucei/radiation effects
16.
Radiat Environ Biophys ; 57(2): 143-152, 2018 05.
Article in English | MEDLINE | ID: mdl-29453554

ABSTRACT

Boron neutron capture therapy (BNCT) for aggressive tumors is based on nuclear reaction [10B (n, α) 7Li]. Previously, we demonstrated that BNCT could be applied for the treatment of undifferentiated thyroid carcinoma. The aim of the present study was to describe the DNA damage pattern and the repair pathways that are activated by BNCT in thyroid cells. We analyzed γH2AX foci and the expression of Ku70, Rad51 and Rad54, main effector enzymes of non-homologous end joining (NHEJ) and homologous recombination repair (HRR) pathways, respectively, in thyroid follicular carcinoma cells. The studied groups were: (1) C [no irradiation], (2) gamma [60Co source], (3) N [neutron beam alone], (4) BNCT [neutron beam plus 10 µg 10B/ml of boronphenylalanine (10BPA)]. The total absorbed dose was always 3 Gy. The results showed that the number of nuclear γH2AX foci was higher in the gamma group than in the N and BNCT groups (30 min-24 h) (p < 0.001). However, the focus size was significantly larger in BNCT compared to other groups (p < 0.01). The analysis of repair enzymes showed a significant increase in Rad51 and Rad54 mRNA at 4 and 6 h, respectively; in both N and BNCT groups and the expression of Ku70 did not show significant differences between groups. These findings are consistent with an activation of HRR mechanism in thyroid cells. A melanoma cell line showed different DNA damage pattern and activation of both repair pathways. These results will allow us to evaluate different blocking points, to potentiate the damage induced by BNCT.


Subject(s)
Boron Neutron Capture Therapy , DNA Damage , DNA Repair/radiation effects , Thyroid Neoplasms/pathology , Cell Line, Tumor , DNA End-Joining Repair/radiation effects , Dose-Response Relationship, Radiation , Gene Expression Regulation, Neoplastic/radiation effects , Histones/metabolism , Homologous Recombination/radiation effects , Humans
17.
Sci Rep ; 7(1): 16518, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29184099

ABSTRACT

Homologous Recombination (HR) repair is essential for repairing DNA double strand breaks (DSB) in dividing cells and preventing tumorigenesis. BRCA2 plays an important role in HR by recruiting the DNA recombinase RAD51 to the DSB. Despite being a popular model organism in genetic and cancer research, knowledge on the conservation of the HR pathway and function of zebrafish Brca2 is limited. To evaluate this, we developed a Rad51 foci assay in zebrafish embryos. We identified the zebrafish embryonic intestinal tissue as an ideal target for Rad51 immunostaining. After inducing DSB through irradiation, Rad51 foci were present in irradiated embryos but not in unirradiated controls. We present a method for accurate quantification of HR. Both morpholino-induced knockdown and knockout of Brca2 lead to almost complete absence of Rad51 foci in irradiated embryos. These findings indicate conserved function of Brca2 in zebrafish. Interestingly, a statistically significant decrease in Rad51 foci was observed in Brca2 heterozygous carriers compared to wild types, indicative of haploinsufficiency, a hypothesised cause of some tumours in patients with a germline BRCA2 mutation. In conclusion, we demonstrated the suitability of zebrafish as an excellent in vivo model system for studying the HR pathway and its functionality.


Subject(s)
BRCA2 Protein/deficiency , Genetic Testing , Homologous Recombination , Zebrafish Proteins/deficiency , Zebrafish/genetics , Animals , Animals, Genetically Modified , Embryo, Nonmammalian , Gene Knockdown Techniques , Genotyping Techniques , Homologous Recombination/radiation effects , Immunohistochemistry , Rad51 Recombinase/metabolism , Radiation, Ionizing
18.
Radiat Res ; 188(2): 144-155, 2017 08.
Article in English | MEDLINE | ID: mdl-28574327

ABSTRACT

Exposure to ionizing radiation greatly increases the risk of developing papillary thyroid carcinoma (PTC), especially during childhood, mainly due to gradual inactivation of DNA repair genes and DNA damages. Recent molecular characterization of PTC revealed DNA methylation deregulation of several promoters of DNA repair genes. Thus, epigenetic silencing might be a plausible mechanism for the activity loss of tumor suppressor genes in radiation-induced thyroid tumors. Herein, we investigated the impact of ionizing radiation on global methylation and CpG islands within promoter regions of homologous recombination (HR) and non-homologous end joining (NHEJ) genes, as well as its effects on gene expression, using two well-established normal differentiated thyroid cell lines (FRTL5 and PCCL3). Our data reveal that X-ray exposure promoted G2/M arrest in normal thyroid cell lines. The FRTL5 cells displayed a slower kinetics of double-strand breaks (DSB) repair and a lower long interspersed nuclear element-1 (LINE-1) methylation than the PCCL3 cells. Nevertheless, acute X-ray exposure does not alter the expression of genes involved in HR and NHEJ pathways, apart from the downregulation of Brca1 in thyroid cells. On the other hand, HR and NHEJ gene expressions were upregulated in radiation-induced senescent thyroid cells. Taken together, these data suggest that FRTL5 cells intrinsically have less efficient DNA DSB repair machinery than PCCL3 cells, as well as genomic instability, which could predispose the FRTL5 cells to unrepaired DSB lesions and, therefore, gene mutations.


Subject(s)
BRCA1 Protein/genetics , DNA Methylation/radiation effects , DNA Repair/radiation effects , Gene Expression Regulation/radiation effects , Long Interspersed Nucleotide Elements/genetics , Thyroid Gland/cytology , Animals , Cell Line , Cellular Senescence/radiation effects , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair/radiation effects , Homologous Recombination/radiation effects , Kinetics , Phenotype , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Thyroid Gland/metabolism , Thyroid Gland/radiation effects , Up-Regulation/radiation effects
19.
Radiat Res ; 188(1): 82-93, 2017 07.
Article in English | MEDLINE | ID: mdl-28535128

ABSTRACT

Genome instability is a hallmark of cancer cells and dysregulation or defects in DNA repair pathways cause genome instability and are linked to inherited cancer predisposition syndromes. Ionizing radiation can cause immediate effects such as mutation or cell death, observed within hours or a few days after irradiation. Ionizing radiation also induces delayed effects many cell generations after irradiation. Delayed effects include hypermutation, hyper-homologous recombination, chromosome instability and reduced clonogenic survival (delayed death). Delayed hyperrecombination (DHR) is mechanistically distinct from delayed chromosomal instability and delayed death. Using a green fluorescent protein (GFP) direct repeat homologous recombination system, time-lapse microscopy and colony-based assays, we demonstrate that DHR increases several-fold in response to low-LET X rays and high-LET carbon-ion radiation. Time-lapse analyses of DHR revealed two classes of recombinants not detected in colony-based assays, including cells that recombined and then senesced or died. With both low- and high-LET radiation, DHR was evident during the first two weeks postirradiation, but resolved to background levels during the third week. The results indicate that the risk of radiation-induced genome destabilization via DHR is time limited, and suggest that there is little or no additional risk of radiation-induced genome instability mediated by DHR with high-LET radiation compared to low-LET radiation.


Subject(s)
DNA Repair/genetics , Homologous Recombination/genetics , Homologous Recombination/radiation effects , Linear Energy Transfer/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/radiotherapy , Cell Line, Tumor , Dose-Response Relationship, Radiation , Humans , Linear Energy Transfer/radiation effects , Radiotherapy Dosage
20.
Sci Rep ; 7: 44662, 2017 03 20.
Article in English | MEDLINE | ID: mdl-28317934

ABSTRACT

DEK is a highly conserved chromatin-bound protein whose upregulation across cancer types correlates with genotoxic therapy resistance. Loss of DEK induces genome instability and sensitizes cells to DNA double strand breaks (DSBs), suggesting defects in DNA repair. While these DEK-deficiency phenotypes were thought to arise from a moderate attenuation of non-homologous end joining (NHEJ) repair, the role of DEK in DNA repair remains incompletely understood. We present new evidence demonstrating the observed decrease in NHEJ is insufficient to impact immunoglobulin class switching in DEK knockout mice. Furthermore, DEK knockout cells were sensitive to apoptosis with NHEJ inhibition. Thus, we hypothesized DEK plays additional roles in homologous recombination (HR). Using episomal and integrated reporters, we demonstrate that HR repair of conventional DSBs is severely compromised in DEK-deficient cells. To define responsible mechanisms, we tested the role of DEK in the HR repair cascade. DEK-deficient cells were impaired for γH2AX phosphorylation and attenuated for RAD51 filament formation. Additionally, DEK formed a complex with RAD51, but not BRCA1, suggesting a potential role regarding RAD51 filament formation, stability, or function. These findings define DEK as an important and multifunctional mediator of HR, and establish a synthetic lethal relationship between DEK loss and NHEJ inhibition.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Homologous Recombination , Oncogene Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Female , HeLa Cells , Histones/metabolism , Homologous Recombination/drug effects , Homologous Recombination/radiation effects , Humans , Male , Mice, Knockout , Protein Binding/drug effects , Protein Binding/radiation effects , Protein Kinase Inhibitors/pharmacology , Rad51 Recombinase/metabolism , Radiation, Ionizing , Replication Protein A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...