Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 342
Filter
1.
Ecotoxicol Environ Saf ; 279: 116486, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38820877

ABSTRACT

Human exposure to radiofrequency electromagnetic fields (RF-EMF) is restricted to prevent thermal effects in the tissue. However, at very low intensity exposure "non-thermal" biological effects, like oxidative stress, DNA or chromosomal aberrations, etc. collectively termed genomic-instability can occur after few hours. Little is known about chronic (years long) exposure with non-thermal RF-EMF. We identified two neighboring housing estates in a rural region with residents exposed to either relatively low (control-group) or relatively high (exposed-group) RF-EMF emitted from nearby mobile phone base stations (MPBS). 24 healthy adults that lived in their homes at least for 5 years volunteered. The homes were surveyed for common types of EMF, blood samples were tested for oxidative status, transient DNA alterations, permanent chromosomal damage, and specific cancer related genetic markers, like MLL gene rearrangements. We documented possible confounders, like age, sex, nutrition, life-exposure to ionizing radiation (X-rays), occupational exposures, etc. The groups matched well, age, sex, lifestyle and occupational risk factors were similar. The years long exposure had no measurable effect on MLL gene rearrangements and c-Abl-gene transcription modification. Associated with higher exposure, we found higher levels of lipid oxidation and oxidative DNA-lesions, though not statistically significant. DNA double strand breaks, micronuclei, ring chromosomes, and acentric chromosomes were not significantly different between the groups. Chromosomal aberrations like dicentric chromosomes (p=0.007), chromatid gaps (p=0.019), chromosomal fragments (p<0.001) and the total of chromosomal aberrations (p<0.001) were significantly higher in the exposed group. No potential confounder interfered with these findings. Increased rates of chromosomal aberrations as linked to excess exposure with ionizing radiation may also occur with non-ionizing radiation exposure. Biological endpoints can be informative for designing exposure limitation strategies. Further research is warranted to investigate the dose-effect-relationship between both, exposure intensity and exposure time, to account for endpoint accumulations after years of exposure. As established for ionizing radiation, chromosomal aberrations could contribute to the definition of protection thresholds, as their rate reflects exposure intensity and exposure time.


Subject(s)
Cell Phone , Electromagnetic Fields , Genomic Instability , Oxidative Stress , Humans , Male , Female , Electromagnetic Fields/adverse effects , Germany , Adult , Middle Aged , Genomic Instability/radiation effects , Chromosome Aberrations , Environmental Exposure , Radio Waves/adverse effects , DNA Damage
2.
Int J Radiat Biol ; 100(7): 1072-1084, 2024.
Article in English | MEDLINE | ID: mdl-38683196

ABSTRACT

PURPOSE: Gamma rays are the most widely exploited physical mutagen in plant mutation breeding. They are known to be involved in the development of more than 60% of global cowpea (Vigna unguiculata (L.) Walp.) mutant varieties. Nevertheless, the nature and type of genome-wide mutations induced by gamma rays have not been studied in cowpea and therefore, the present investigation was undertaken. MATERIALS AND METHODS: Genomic DNAs from three stable gamma rays-induced mutants (large seed size, small seed size and disease resistant mutant) of cowpea cultivar 'CPD103' in M6 generation along with its progenitor were used for Illumina-based whole-genome resequencing. RESULTS: Gamma rays induced a relatively higher frequency (88.9%) of single base substitutions (SBSs) with an average transition to transversion ratio (Ti/Tv) of 3.51 in M6 generation. A > G transitions, including its complementary T > C transitions, predominated the transition mutations, while all four types of transversion mutations were detected with frequencies over 6.5%. Indels (small insertions and deletions) constituted about 11% of the total induced variations, wherein small insertions (6.3%) were relatively more prominent than small deletions (4.8%). Among the indels, single-base indels and, in particular, those involving A/T bases showed a preponderance, albeit indels of up to three bases were detected in low proportions. Distributed across all 11 chromosomes, only a fraction of SBSs (19.45%) and indels (20.2%) potentially altered the encoded amino acids/peptides. The inherent mutation rate induced by gamma rays in cowpea was observed to be in the order of 1.4 × 10-7 per base pair in M6 generation. CONCLUSION: Gamma-rays with a greater tendency to induce SBSs and, to a lesser extent, indels could be efficiently and effectively exploited in cowpea mutation breeding.


Subject(s)
Gamma Rays , Genome, Plant , Mutation , Vigna , Whole Genome Sequencing , Vigna/genetics , Vigna/radiation effects , Genomic Instability/radiation effects
3.
Nature ; 613(7943): 365-374, 2023 01.
Article in English | MEDLINE | ID: mdl-36544019

ABSTRACT

How paternal exposure to ionizing radiation affects genetic inheritance and disease risk in the offspring has been a long-standing question in radiation biology. In humans, nearly 80% of transmitted mutations arise in the paternal germline1, but the transgenerational effects of ionizing radiation exposure has remained controversial and the mechanisms are unknown. Here we show that in sex-separated Caenorhabditis elegans strains, paternal, but not maternal, exposure to ionizing radiation leads to transgenerational embryonic lethality. The offspring of irradiated males displayed various genome instability phenotypes, including DNA fragmentation, chromosomal rearrangement and aneuploidy. Paternal DNA double strand breaks were repaired by maternally provided error-prone polymerase theta-mediated end joining. Mechanistically, we show that depletion of an orthologue of human histone H1.0, HIS-24, or the heterochromatin protein HPL-1, could significantly reverse the transgenerational embryonic lethality. Removal of HIS-24 or HPL-1 reduced histone 3 lysine 9 dimethylation and enabled error-free homologous recombination repair in the germline of the F1 generation from ionizing radiation-treated P0 males, consequently improving the viability of the F2 generation. This work establishes the mechanistic underpinnings of the heritable consequences of paternal radiation exposure on the health of offspring, which may lead to congenital disorders and cancer in humans.


Subject(s)
Caenorhabditis elegans , DNA Damage , DNA Repair , Histones , Animals , Humans , Male , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/radiation effects , DNA Damage/radiation effects , Genomic Instability/radiation effects , Histones/metabolism , Mutation , Radiation, Ionizing , Embryo Loss/genetics , Female , DNA Breaks, Double-Stranded/radiation effects , DNA End-Joining Repair , DNA Polymerase theta
4.
Nat Commun ; 13(1): 701, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35121747

ABSTRACT

Inheritance of stable and euploid genomes is a prerequisite for species maintenance. The DNA damage response in germ cells controls the integrity of heritable genomes. Whether and how somatic stress responses impact the quality control of germline genomes has remained unclear. Here, we show that PMK-1/p38-mediated stress signaling in intestinal cells is required for germ cell apoptosis amid ionizing radiation (IR)-induced or meiotic DNA double strand breaks (DSBs) in C. elegans. We demonstrate that intestinal PMK-1/p38 signaling regulates the germ cell death in response to environmental stress. The PMK-1/p38 target SYSM-1 is secreted from the intestine into the germline to trigger apoptosis of meiotic pachytene cells. Compromised PMK-1/p38 signaling in intestinal cells leads to stress-induced aneuploidy in the consequent generation. Our data suggest that somatic stress surveillance controls heritable genome integrity and euploidy.


Subject(s)
Aneuploidy , Apoptosis/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Germ Cells/metabolism , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinases/genetics , Stress, Physiological/genetics , Animals , Animals, Genetically Modified , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/genetics , Gene Expression Regulation , Genomic Instability/radiation effects , Hot Temperature , Mitogen-Activated Protein Kinases/metabolism , Radiation, Ionizing , Reverse Transcriptase Polymerase Chain Reaction
5.
Biomed Res Int ; 2021: 2888393, 2021.
Article in English | MEDLINE | ID: mdl-34926683

ABSTRACT

Background and Purpose. Postexposure onset of dietary restriction (DR) is expected to provide therapeutic nutritional approaches to reduce health risk from exposure to ionizing radiation (IR) due to such as manned space exploration, radiotherapy, or nuclear accidents as IR could alleviate radiocarcinogenesis in animal models. However, the underlying mechanisms remain largely unknown. This study is aimed at investigating the effect from postexposure onset of DR on genotoxicity and genomic instability (GI) induced by total body irradiation (TBI) in mice. Materials and Methods. Mice were exposed to 2.0 Gy of accelerated iron particles with an initial energy of 500 MeV/nucleon and a linear energy transfer (LET) value of about 200 keV/µm. After TBI, mice were either allowed to free access to a standard laboratory chow or treated under DR (25% cut in diet). Using micronucleus frequency (MNF) in bone marrow erythrocytes, induction of acute genotoxicity and GI in the hematopoietic system was, respectively, determined 1 and 2 months after TBI. Results and Conclusions. TBI alone caused a significant increase in MNF while DR alone did not markedly influence the MNF. DR induced a significant decrease in MNF compared to the treatment by TBI alone. Results demonstrated that postexposure onset of DR could relieve the elevated MNF induced by TBI with high-LET iron particles. These findings indicated that reduction in acute genotoxicity and late GI may be at least a part of the mechanisms underlying decreased radiocarcinogenesis by DR.


Subject(s)
Genomic Instability/radiation effects , Iron/adverse effects , Animals , Cell Nucleus/radiation effects , Diet Therapy/methods , Eating/physiology , Erythrocytes/radiation effects , Female , Mice , Mice, Inbred C57BL , Mutagenicity Tests/methods , Radiation, Ionizing
6.
Sci Rep ; 11(1): 21364, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34725419

ABSTRACT

In response to UV irradiation, translesion DNA synthesis (TLS) utilizes specialized DNA polymerases to bypass replication-blocking lesions. In a well-established polymerase switch model, Polη is thought to be a preferred TLS polymerase to insert correct nucleotides across from the thymine dimer, and Rev1 plays a scaffold role through physical interaction with Polη and the Rev7 subunit of Polζ for continual DNA synthesis. Defective Polη causes a variant form of xeroderma pigmentosum (XPV), a disease with predisposition to sunlight-induced skin cancer. Previous studies revealed that expression of Rev1 alone is sufficient to confer enhanced UV damage tolerance in mammalian cells, which depends on its physical interaction with Polζ but is independent of Polη, a conclusion that appears to contradict current literature on the critical roles of Polη in TLS. To test a hypothesis that the Rev1 catalytic activity is required to backup Polη in TLS, we found that the Rev1 polymerase-dead mutation is synergistic with either Polη mutation or the Polη-interaction mutation in response to UV-induced DNA damage. On the other hand, functional complementation of polH cells by Polη relies on its physical interaction with Rev1. Hence, our studies reveal critical interactions between Rev1 and Polη in response to UV damage.


Subject(s)
DNA Damage/radiation effects , DNA-Directed DNA Polymerase/genetics , Nucleotidyltransferases/genetics , Ultraviolet Rays/adverse effects , DNA-Directed DNA Polymerase/metabolism , Genomic Instability/radiation effects , HEK293 Cells , Humans , Mutation/radiation effects , Nucleotidyltransferases/metabolism , Protein Interaction Maps/radiation effects
7.
Int J Mol Sci ; 22(20)2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34681703

ABSTRACT

Until recently, radiation effects have been considered to be mainly due to nuclear DNA damage and their management by repair mechanisms. However, molecular biology studies reveal that the outcomes of exposures to ionizing radiation (IR) highly depend on activation and regulation through other molecular components of organelles that determine cell survival and proliferation capacities. As typical epigenetic-regulated organelles and central power stations of cells, mitochondria play an important pivotal role in those responses. They direct cellular metabolism, energy supply and homeostasis as well as radiation-induced signaling, cell death, and immunological responses. This review is focused on how energy, dose and quality of IR affect mitochondria-dependent epigenetic and functional control at the cellular and tissue level. Low-dose radiation effects on mitochondria appear to be associated with epigenetic and non-targeted effects involved in genomic instability and adaptive responses, whereas high-dose radiation effects (>1 Gy) concern therapeutic effects of radiation and long-term outcomes involving mitochondria-mediated innate and adaptive immune responses. Both effects depend on radiation quality. For example, the increased efficacy of high linear energy transfer particle radiotherapy, e.g., C-ion radiotherapy, relies on the reduction of anastasis, enhanced mitochondria-mediated apoptosis and immunogenic (antitumor) responses.


Subject(s)
Epigenesis, Genetic/radiation effects , Mitochondria/metabolism , Radiation, Ionizing , Signal Transduction/radiation effects , Epithelial-Mesenchymal Transition/radiation effects , Genomic Instability/radiation effects , Humans , Mitochondria/genetics , Mitochondria/radiation effects , Mitochondrial Dynamics/radiation effects , Oxidative Stress/radiation effects , Reactive Oxygen Species/metabolism
8.
Cell Rep ; 36(5): 109492, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34348144

ABSTRACT

Early differential diagnosis between malignant and benign tumors and their underlying intrinsic differences are the most critical issues for life-threatening cancers. To study whether human acral melanomas, deadly cancers that occur on non-hair-bearing skin, have distinct origins that underlie their invasive capability, we develop fate-tracing technologies of melanocyte stem cells in sweat glands (glandular McSCs) and in melanoma models in mice and compare the cellular dynamics with human melanoma. Herein, we report that glandular McSCs self-renew to expand their migratory progeny in response to genotoxic stress and trauma to generate invasive melanomas in mice that mimic human acral melanomas. The analysis of melanocytic lesions in human volar skin reveals that genetically unstable McSCs expand in sweat glands and in the surrounding epidermis in melanomas but not in nevi. The detection of such cell spreading dynamics provides an innovative method for an early differential diagnosis of acral melanomas from nevi.


Subject(s)
Cell Movement , Melanoma/pathology , Nevus/pathology , Stem Cells/pathology , Animals , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Cyclin D1/metabolism , Disease Models, Animal , Epidermis/pathology , Epidermis/radiation effects , Gene Amplification , Genomic Instability/radiation effects , Melanocytes/pathology , Melanocytes/radiation effects , Melanoma/diagnosis , Mice, Inbred C57BL , Risk Factors , Skin/pathology , Skin/radiation effects , Skin Pigmentation/radiation effects , Sweat Glands/radiation effects , Ultraviolet Rays
9.
PLoS Genet ; 17(1): e1009302, 2021 01.
Article in English | MEDLINE | ID: mdl-33444353

ABSTRACT

Human skin is continuously exposed to environmental DNA damage leading to the accumulation of somatic mutations over the lifetime of an individual. Mutagenesis in human skin cells can be also caused by endogenous DNA damage and by DNA replication errors. The contributions of these processes to the somatic mutation load in the skin of healthy humans has so far not been accurately assessed because the low numbers of mutations from current sequencing methodologies preclude the distinction between sequencing errors and true somatic genome changes. In this work, we sequenced genomes of single cell-derived clonal lineages obtained from primary skin cells of a large cohort of healthy individuals across a wide range of ages. We report here the range of mutation load and a comprehensive view of the various somatic genome changes that accumulate in skin cells. We demonstrate that UV-induced base substitutions, insertions and deletions are prominent even in sun-shielded skin. In addition, we detect accumulation of mutations due to spontaneous deamination of methylated cytosines as well as insertions and deletions characteristic of DNA replication errors in these cells. The endogenously induced somatic mutations and indels also demonstrate a linear increase with age, while UV-induced mutation load is age-independent. Finally, we show that DNA replication stalling at common fragile sites are potent sources of gross chromosomal rearrangements in human cells. Thus, somatic mutations in skin of healthy individuals reflect the interplay of environmental and endogenous factors in facilitating genome instability and carcinogenesis.


Subject(s)
DNA Damage/radiation effects , DNA Methylation/genetics , DNA Replication/genetics , Skin/radiation effects , DNA Methylation/radiation effects , DNA Repair/radiation effects , DNA Replication/radiation effects , Fibroblasts/radiation effects , Genome, Human/genetics , Genome, Human/radiation effects , Genomic Instability/radiation effects , Genomics/methods , Humans , INDEL Mutation/radiation effects , Melanocytes/radiation effects , Mutagenesis/genetics , Mutagenesis/radiation effects , Skin/metabolism , Ultraviolet Rays/adverse effects
10.
Plast Reconstr Surg ; 147(1S-2): 7S-14S, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33347069

ABSTRACT

BACKGROUND: The landmark National Aeronautics and Space Administration Twins Study represented an integrated effort to launch human space life science research into the modern age of molecular- and "omics"-based studies. As part of the first One-Year Mission aboard the International Space Station, identical twin astronauts Scott and Mark Kelly were the subjects of this "out of this world" research opportunity. Telomeres, the natural ends of chromosomes that shorten with cell division and a host of lifestyle factors and stresses, are key molecular determinants of aging and aging trajectories. METHODS: We proposed that telomere length dynamics (changes over time) represent a particularly relevant and integrative biomarker for astronauts, as they reflect the combined experiences and environmental exposures encountered during spaceflight. Telomere length (quantitative polymerase chain reaction and telomere fluorescence in situ hybridization) and telomerase activity (quantitative polymerase chain reaction -telomere repeat amplification protocol) were longitudinally assessed in the space- and earth-bound twins. Chromosome aberrations (directional genomic hybridization), signatures of radiation exposure, were also evaluated. RESULTS: The twins had relatively similar telomere lengths before spaceflight, and the earth-bound twins' telomeres remained relatively stable over the course of the study. Surprisingly, the space twins' telomeres were longer during spaceflight, and upon return to Earth shortened rapidly, resulting in many more short telomeres after spaceflight than before. Chromosomal signatures of space radiation exposure were also elevated during spaceflight, and increased inversion frequencies persisted after spaceflight, suggestive of ongoing genome instability. CONCLUSION: Although the definitive mechanisms underlying such dramatic spaceflight-associated shifts in telomere length remain unclear, improved maintenance of telomere length has important implications for aging science and improving healthspan for those on Earth, as well.


Subject(s)
Aging/genetics , Space Flight , Telomere Shortening/physiology , Telomere/metabolism , Twins, Monozygotic/genetics , Aging/radiation effects , Cosmic Radiation/adverse effects , Genomic Instability/radiation effects , Humans , Longitudinal Studies , Male , Telomerase/metabolism , Telomere Shortening/radiation effects , Time Factors
11.
J Invest Dermatol ; 141(4S): 1104-1110, 2021 04.
Article in English | MEDLINE | ID: mdl-33358021

ABSTRACT

UVR is a major etiology for premature skin aging that leads to photoaging and UV-induced skin cancers. In the skin, TGFß signaling is a growth inhibitor for keratinocytes and a profibrotic factor in the dermis. It exerts context-dependent effects on tumor progression. Chronic UV exposure likely causes TGFß1/SMAD3 signaling activation and contributes to metalloproteinase-induced collagen degradation and photoinflammation in photoaging. UV irradiation also causes gene mutations in key elements of the TGFß pathway, including TGFßRI, TGFßRII, SMAD2, and SMAD4. These mutations enable tumor cells to escape from TGFß-induced growth inhibition and induce genomic instability and cancer stem cells, leading to the initiation, progression, invasion, and metastasis of cutaneous squamous cell carcinoma (cSCC). Furthermore, UV-induced mutations cause TGFß overexpression in the tumor microenvironment (TME) of cSCC, basal cell carcinoma (BCC), and cutaneous melanoma, resulting in inflammation, angiogenesis, cancer-associated fibroblasts, and immune inhibition, supporting cancer survival, immune evasion, and metastasis. The pleiotropic effects of TGFß provide possible treatment options for photoaging and skin cancer. Given the high UV-induced mutational burden and immune-repressive TME seen in cSCC, BCC, and cutaneous melanoma, treatment with the combination of a TGFß signaling inhibitor and immune checkpoint blockade could reverse immune evasion to reduce tumor growth.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Skin Aging/radiation effects , Skin Neoplasms/etiology , Transforming Growth Factor beta/metabolism , Ultraviolet Rays/adverse effects , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Genomic Instability/radiation effects , Humans , Immune Checkpoint Inhibitors , Keratinocytes/drug effects , Keratinocytes/pathology , Keratinocytes/radiation effects , Mice , Mutation/radiation effects , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/radiation effects , Skin/drug effects , Skin/pathology , Skin/radiation effects , Skin Aging/drug effects , Skin Aging/genetics , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Transforming Growth Factor beta/antagonists & inhibitors , Tumor Escape/genetics , Tumor Escape/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/radiation effects , Xenograft Model Antitumor Assays
12.
Sci Rep ; 10(1): 19422, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33173044

ABSTRACT

Extracellular pH has been assumed to play little if any role in how bacteria respond to antibiotics and antibiotic resistance development. Here, we show that the intracellular pH of Escherichia coli equilibrates to the environmental pH following treatment with the DNA damaging antibiotic nalidixic acid. We demonstrate that this allows the environmental pH to influence the transcription of various DNA damage response genes and physiological processes such as filamentation. Using purified RecA and a known pH-sensitive mutant variant RecA K250R we show how pH can affect the biochemical activity of a protein central to control of the bacterial DNA damage response system. Finally, two different mutagenesis assays indicate that environmental pH affects antibiotic resistance development. Specifically, at environmental pH's greater than six we find that mutagenesis plays a significant role in producing antibiotic resistant mutants. At pH's less than or equal to 6 the genome appears more stable but extensive filamentation is observed, a phenomenon that has previously been linked to increased survival in the presence of macrophages.


Subject(s)
Anti-Bacterial Agents/pharmacology , DNA Damage/drug effects , DNA Damage/genetics , Escherichia coli/drug effects , Escherichia coli/genetics , Genomic Instability/drug effects , Genomic Instability/genetics , DNA Damage/radiation effects , Electrophoretic Mobility Shift Assay , Escherichia coli/radiation effects , Flow Cytometry , Genomic Instability/radiation effects , Hydrogen-Ion Concentration , Microbial Viability/drug effects , Microbial Viability/radiation effects , Nalidixic Acid/pharmacology , Propidium/pharmacology , Rifampin/pharmacology , Ultraviolet Rays
13.
PLoS One ; 15(11): e0235998, 2020.
Article in English | MEDLINE | ID: mdl-33253193

ABSTRACT

In contrast to the vast majority of research that has focused on the immediate effects of ionizing radiation, this work concentrates on the molecular mechanism driving delayed effects that emerge in the progeny of the exposed cells. We employed functional protein arrays to identify molecular changes induced in a human bronchial epithelial cell line (HBEC3-KT) and osteosarcoma cell line (U2OS) and evaluated their impact on outcomes associated with radiation induced genomic instability (RIGI) at day 5 and 7 post-exposure to a 2Gy X-ray dose, which revealed replication stress in the context of increased FOXM1b expression. Irradiated cells had reduced DNA replication rate detected by the DNA fiber assay and increased DNA resection detected by RPA foci and phosphorylation. Irradiated cells increased utilization of homologous recombination-dependent repair detected by a gene conversion assay and DNA damage at mitosis reflected by RPA positive chromosomal bridges, micronuclei formation and 53BP1 positive bodies in G1, all known outcomes of replication stress. Interference with the function of FOXM1, a transcription factor widely expressed in cancer, employing an aptamer, decreased radiation-induced micronuclei formation and cell transformation while plasmid-driven overexpression of FOXM1b was sufficient to induce replication stress, micronuclei formation and cell transformation.


Subject(s)
Bronchi/pathology , Cell Transformation, Neoplastic/pathology , DNA Replication , Epithelial Cells/pathology , Forkhead Box Protein M1/metabolism , Genomic Instability/radiation effects , Stress, Physiological , Bronchi/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , DNA Damage , Epithelial Cells/metabolism , Forkhead Box Protein M1/genetics , Humans , Radiation, Ionizing
14.
Nucleic Acids Res ; 48(19): e111, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33010172

ABSTRACT

Ionizing radiation (IR) is environmentally prevalent and, depending on dose and linear energy transfer (LET), can elicit serious health effects by damaging DNA. Relative to low LET photon radiation (X-rays, gamma rays), higher LET particle radiation produces more disease causing, complex DNA damage that is substantially more challenging to resolve quickly or accurately. Despite the majority of human lifetime IR exposure involving long-term, repetitive, low doses of high LET alpha particles (e.g. radon gas inhalation), technological limitations to deliver alpha particles in the laboratory conveniently, repeatedly, over a prolonged period, in low doses and in an affordable, high-throughput manner have constrained DNA damage and repair research on this topic. To resolve this, we developed an inexpensive, high capacity, 96-well plate-compatible alpha particle irradiator capable of delivering adjustable, low mGy/s particle radiation doses in multiple model systems and on the benchtop of a standard laboratory. The system enables monitoring alpha particle effects on DNA damage repair and signalling, genome stability pathways, oxidative stress, cell cycle phase distribution, cell viability and clonogenic survival using numerous microscopy-based and physical techniques. Most importantly, this method is foundational for high-throughput genetic screening and small molecule testing in mammalian and yeast cells.


Subject(s)
Alpha Particles/adverse effects , DNA Damage/radiation effects , DNA Repair/radiation effects , Genomic Instability/radiation effects , Radiation Genetics/instrumentation , A549 Cells , Cell Cycle/radiation effects , HeLa Cells , Humans , Oxidative Stress/radiation effects , Saccharomyces cerevisiae , Signal Transduction/radiation effects
15.
Cell Cycle ; 19(12): 1545-1561, 2020 06.
Article in English | MEDLINE | ID: mdl-32380926

ABSTRACT

The DUSP3 phosphatase regulates cell cycle, proliferation, apoptosis and senescence of different cell types, lately shown as a mediator of DNA repair processes. This work evaluated the impact of DUSP3 loss of function (lof) on DNA repair-proficient fibroblasts (MRC-5), NER-deficient cell lines (XPA and XPC) and translesion DNA synthesis (TLS)-deficient cells (XPV), after UV-radiation stress. The levels of DNA strand breaks, CPDs and 6-4-PPs have accumulated over time in all cells under DUSP3 lof, with a significant increase in NER-deficient lines. The inefficient repair of these lesions increased sub-G1 population of XPA and XPC cells 24 hours after UV treatment, notably marked by DUSP3 lof, which is associated with a reduced cell population in G1, S and G2/M phases. It was also detected an increase in S and G2/M populations of XPV and MRC-5 cells after UV-radiation exposure, which was slightly attenuated by DUSP3 lof due to a discrete increase in sub-G1 cells. The cell cycle progression was accompanied by changes in the levels of the main Cyclins (A1, B1, D1 or E1), CDKs (1, 2, 4 or 6), and the p21 Cip1 inhibitor, in a DUSP3-dependent manner. DUSP3 lof affected the proliferation of MRC-5 and XPA cells, with marked worsening of the XP phenotype after UV radiation. This work highlights the roles of DUSP3 in DNA repair fitness and in the fine control of regulatory proteins of cell cycle, essential mechanisms to maintenance of genomic stability.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Repair/genetics , Dual Specificity Phosphatase 3/metabolism , Genomic Instability , Cell Cycle/radiation effects , Cell Proliferation/genetics , Cell Proliferation/radiation effects , DNA Damage , DNA Repair/radiation effects , Gene Silencing/radiation effects , Genomic Instability/radiation effects , Humans , Pyrimidine Dimers/metabolism , Stress, Physiological/radiation effects , Ultraviolet Rays
16.
Biomed Res Int ; 2020: 4703286, 2020.
Article in English | MEDLINE | ID: mdl-32337251

ABSTRACT

Space travel has advanced significantly over the last six decades with astronauts spending up to 6 months at the International Space Station. Nonetheless, the living environment while in outer space is extremely challenging to astronauts. In particular, exposure to space radiation represents a serious potential long-term threat to the health of astronauts because the amount of radiation exposure accumulates during their time in space. Therefore, health risks associated with exposure to space radiation are an important topic in space travel, and characterizing space radiation in detail is essential for improving the safety of space missions. In the first part of this review, we provide an overview of the space radiation environment and briefly present current and future endeavors that monitor different space radiation environments. We then present research evaluating adverse biological effects caused by exposure to various space radiation environments and how these can be reduced. We especially consider the deleterious effects on cellular DNA and how cells activate DNA repair mechanisms. The latest technologies being developed, e.g., a fluorescent ubiquitination-based cell cycle indicator, to measure real-time cell cycle progression and DNA damage caused by exposure to ultraviolet radiation are presented. Progress in examining the combined effects of microgravity and radiation to animals and plants are summarized, and our current understanding of the relationship between psychological stress and radiation is presented. Finally, we provide details about protective agents and the study of organisms that are highly resistant to radiation and how their biological mechanisms may aid developing novel technologies that alleviate biological damage caused by radiation. Future research that furthers our understanding of the effects of space radiation on human health will facilitate risk-mitigating strategies to enable long-term space and planetary exploration.


Subject(s)
Cosmic Radiation/adverse effects , Space Flight , Ultraviolet Rays , Animals , Astronauts , Carcinogenesis/radiation effects , Central Nervous System/radiation effects , Chromosome Aberrations/radiation effects , DNA Damage/radiation effects , DNA Repair/radiation effects , Extraterrestrial Environment , Genomic Instability/radiation effects , Humans , Micronuclei, Chromosome-Defective/radiation effects , Protective Agents/pharmacology , Radiation Dosage , Radiation Exposure/adverse effects , Radiation Exposure/prevention & control , Stress, Psychological , Weightlessness
17.
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
18.
Cells ; 9(2)2020 02 04.
Article in English | MEDLINE | ID: mdl-32033081

ABSTRACT

The DNA damage response is mediated by both DNA repair proteins and epigenetic markers. Here, we observe that N6-methyladenosine (m6A), a mark of the epitranscriptome, was common in RNAs accumulated at UV-damaged chromatin; however, inhibitors of RNA polymerases I and II did not affect the m6A RNA level at the irradiated genomic regions. After genome injury, m6A RNAs either diffused to the damaged chromatin or appeared at the lesions enzymatically. DNA damage did not change the levels of METTL3 and METTL14 methyltransferases. In a subset of irradiated cells, only the METTL16 enzyme, responsible for m6A in non-coding RNAs as well as for splicing regulation, was recruited to microirradiated sites. Importantly, the levels of the studied splicing factors were not changed by UVA light. Overall, if the appearance of m6A RNAs at DNA lesions is regulated enzymatically, this process must be mediated via the coregulatory function of METTL-like enzymes. This event is additionally accompanied by radiation-induced depletion of 2,2,7-methylguanosine (m3G/TMG) in RNA. Moreover, UV-irradiation also decreases the global cellular level of N1-methyladenosine (m1A) in RNAs. Based on these results, we prefer a model in which m6A RNAs rapidly respond to radiation-induced stress and diffuse to the damaged sites. The level of both (m1A) RNAs and m3G/TMG in RNAs is reduced as a consequence of DNA damage, recognized by the nucleotide excision repair mechanism.


Subject(s)
Adenosine/analogs & derivatives , RNA, Untranslated/metabolism , RNA/metabolism , Ultraviolet Rays , Adenosine/metabolism , Animals , Cell Line, Tumor , Chromatin/metabolism , DNA Damage , DNA Demethylation/radiation effects , DNA Methylation/genetics , DNA Methylation/radiation effects , Genomic Instability/radiation effects , Guanosine/analogs & derivatives , Guanosine/metabolism , Methylation/radiation effects , Mice , Stress, Physiological/radiation effects
19.
Radiat Environ Biophys ; 59(2): 221-236, 2020 05.
Article in English | MEDLINE | ID: mdl-32076810

ABSTRACT

The article is devoted to the study of the role of intracellular mechanisms in the formation of radiation-induced genetic instability and its transgenerational effect in cells of different tissues of the descendants of Drosophila melanogaster mutant strains whose parents were exposed to chronic radiation (0.42 and 3.5 mGy/h). The level of DNA damage (alkali-labile sites (ALS), single-strand (SSB) and double-strand (DSB) breaks) in cells of somatic (nerve ganglia, imaginal discs) and generative (testis) tissues from directly irradiated animals and their unirradiated offspring was evaluated. Confident transgenerational instability (on the level of ALSs and SSBs), observed only in somatic tissues and only at the higher dose rate, is characteristic for mus209 mutant strains defective in excision repair and, less often, for mus205 and mus210 mutant strains. The greatest manifestation of radiation-induced genetic instability was found in evaluating the DSBs. Dysfunction of the genes mus205, mus304, mei-9 and mei-41, which are responsible for postreplicative repair, excision repair, recombination and control of the cell cycle, affects transgenerational changes in the somatic tissues of the offspring of parents irradiated in both low and high dose rates. In germ cells, the key role in maintaining genetic stability under chronic irradiation is played by the non-recombination postreplication repair mus101 gene. We revealed the tissue specificity of the radiation-induced effects, transgenerational transmission and accumulation of DNA damage to descendants of chronically irradiated animals.


Subject(s)
Drosophila melanogaster/radiation effects , Genomic Instability/radiation effects , Radium , Animals , Comet Assay , DNA Damage , Drosophila melanogaster/genetics , Genome, Insect , Germ Cells/radiation effects , Male , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...