Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 498
Filter
1.
Pak J Biol Sci ; 27(5): 276-282, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38840468

ABSTRACT

<b>Background and Objective:</b> Gamma irradiation induces genotoxicity, characterized by the formation of extra-nuclear bodies and left behind during the anaphase stage of cell division, often referred to as a micronucleus (MN). The present work aims to monitor exposure to ionizing radiation as a genotoxic agent in the lymphocytes of workers at radiation energy centers. <b>Materials and Methods:</b> The lymphocyte cytokinesis block micronucleus assay used and analyzed the correlation between the Nuclear Division Index (NDI), age, blood type and the number of micronuclei (MN). Blood samples were collected from 20 volunteers in heparin tubes, exposed to 2 Gy gamma rays and cultured <i>in vitro</i>. <b>Results:</b> A significant difference in the number of micronuclei between blood group A and blood groups A, B and AB. The Nuclear Division Index (NDI) value for lymphocytes of radiation energy center workers after gamma radiation was significant (1.74±0.1) but still within the normal range. Neither MN frequency nor NDI values correlated with age, but MN frequency showed a correlation with blood type. <b>Conclusion:</b> The gamma irradiation did not induce a cytostatic effect but proved genotoxic to the lymphocytes of radiation energy center workers. Notably, blood type A demonstrated higher sensitivity to gamma radiation.


Subject(s)
Cytokinesis , Gamma Rays , Lymphocytes , Micronucleus Tests , Occupational Exposure , Humans , Gamma Rays/adverse effects , Lymphocytes/radiation effects , Lymphocytes/metabolism , Micronucleus Tests/methods , Cytokinesis/radiation effects , Occupational Exposure/adverse effects , Adult , Male , Middle Aged , Micronuclei, Chromosome-Defective/radiation effects , Female
2.
Mol Cell ; 84(11): 2203-2213.e5, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38749421

ABSTRACT

The cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway plays a pivotal role in innate immune responses to viral infection and inhibition of autoimmunity. Recent studies have suggested that micronuclei formed by genotoxic stress can activate innate immune signaling via the cGAS-STING pathway. Here, we investigated cGAS localization, activation, and downstream signaling from micronuclei induced by ionizing radiation, replication stress, and chromosome segregation errors. Although cGAS localized to ruptured micronuclei via binding to self-DNA, we failed to observe cGAS activation; cGAMP production; downstream phosphorylation of STING, TBK1, or IRF3; nuclear accumulation of IRF3; or expression of interferon-stimulated genes. Failure to activate the cGAS-STING pathway was observed across primary and immortalized cell lines, which retained the ability to activate the cGAS-STING pathway in response to dsDNA or modified vaccinia virus infection. We provide evidence that micronuclei formed by genotoxic insults contain histone-bound self-DNA, which we show is inhibitory to cGAS activation in cells.


Subject(s)
Chromosome Segregation , Membrane Proteins , Micronuclei, Chromosome-Defective , Nucleotides, Cyclic , Nucleotidyltransferases , Signal Transduction , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Micronuclei, Chromosome-Defective/radiation effects , Nucleotides, Cyclic/metabolism , Phosphorylation , DNA Replication/radiation effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Immunity, Innate/radiation effects , DNA Damage , HEK293 Cells , Animals , Radiation, Ionizing , HeLa Cells
3.
Environ Res ; 251(Pt 1): 118634, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38452915

ABSTRACT

Several human studies indicate that mobile phone specific electromagnetic fields may cause cancer in humans but the underlying molecular mechanisms are currently not known. Studies concerning chromosomal damage (which is causally related to cancer induction) are controversial and those addressing this issue in mobile phone users are based on the use of questionnaires to assess the exposure. We realized the first human intervention trial in which chromosomal damage and acute toxic effects were studied under controlled conditions. The participants were exposed via headsets at one randomly assigned side of the head to low and high doses of a UMTS signal (n = 20, to 0.1 W/kg and n = 21 to 1.6 W/kg Specific Absorption Rate) for 2 h on 5 consecutive days. Before and three weeks after the exposure, buccal cells were collected from both cheeks and micronuclei (MN, which are formed as a consequence of structural and numerical chromosomal aberrations) and other nuclear anomalies reflecting mitotic disturbance and acute cytotoxic effects were scored. We found no evidence for induction of MN and of nuclear buds which are caused by gene amplifications, but a significant increase of binucleated cells which are formed as a consequence of disturbed cell divisions, and of karyolitic cells, which are indicative for cell death. No such effects were seen in cells from the less exposed side. Our findings indicate that mobile phone specific high frequency electromagnetic fields do not cause acute chromosomal damage in oral mucosa cells under the present experimental conditions. However, we found clear evidence for disturbance of the cell cycle and cytotoxicity. These effects may play a causal role in the induction of adverse long term health effects in humans.


Subject(s)
Cell Phone , Cytokinesis , Mouth Mucosa , Humans , Mouth Mucosa/radiation effects , Mouth Mucosa/cytology , Adult , Male , Cytokinesis/radiation effects , Cell Death/radiation effects , Young Adult , Female , Chromosome Aberrations/radiation effects , Micronucleus Tests , Electromagnetic Fields/adverse effects , Micronuclei, Chromosome-Defective/radiation effects
4.
Int J Mol Sci ; 23(3)2022 Jan 29.
Article in English | MEDLINE | ID: mdl-35163494

ABSTRACT

Usher syndrome (USH) is a rare autosomal recessive disease characterized by the combination of hearing loss, visual impairment due to retinitis pigmentosa, and in some cases vestibular dysfunctions. Studies published in the 1980s reported that USH is associated with cellular radiosensitivity. However, the molecular basis of this particular phenotype has not yet been documented. The aim of this study was therefore to document the radiosensitivity of USH1-a subset of USH-by examining the radiation-induced nucleo-shuttling of ATM (RIANS), as well as the functionality of the repair and signaling pathways of the DNA double-strand breaks (DSBs) in three skin fibroblasts derived from USH1 patients. The clonogenic cell survival, the micronuclei, the nuclear foci formed by the phosphorylated forms of the X variant of the H2A histone (É£H2AX), the phosphorylated forms of the ATM protein (pATM), and the meiotic recombination 11 nuclease (MRE11) were used as cellular and molecular endpoints. The interaction between the ATM and USH1 proteins was also examined by proximity ligation assay. The results showed that USH1 fibroblasts were associated with moderate but significant radiosensitivity, high yield of micronuclei, and impaired DSB recognition but normal DSB repair, likely caused by a delayed RIANS, suggesting a possible sequestration of ATM by some USH1 proteins overexpressed in the cytoplasm. To our knowledge, this report is the first radiobiological characterization of cells from USH1 patients at both molecular and cellular scales.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Radiation Tolerance/genetics , Usher Syndromes/enzymology , Usher Syndromes/genetics , Cell Survival/drug effects , Cell Survival/radiation effects , Clone Cells , Diphosphonates/pharmacology , Fibroblasts/drug effects , Fibroblasts/pathology , Fibroblasts/radiation effects , Histones/metabolism , Humans , Kinetics , MRE11 Homologue Protein/metabolism , Micronuclei, Chromosome-Defective/radiation effects , Models, Biological , Phosphorylation/drug effects , Phosphorylation/radiation effects , Radiation Tolerance/drug effects , Radiation Tolerance/radiation effects , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Subcellular Fractions/radiation effects
5.
Cells ; 10(10)2021 10 09.
Article in English | MEDLINE | ID: mdl-34685687

ABSTRACT

Gynaecologic cancers are common among women and treatment includes surgery, radiotherapy or chemotherapy, where the last two methods induce DNA damage in non-targeted cells like peripheral blood lymphocytes (PBL). Damaged normal cells can transform leading to second malignant neoplasms (SMN) but the level of risk and impact of risk modifiers is not well defined. We investigated how radiotherapy alone or in combination with chemotherapy induce DNA damage in PBL of cervix and endometrial cancer patients during therapy. Blood samples were collected from nine endometrial cancer patients (treatment with radiotherapy + chemotherapy-RC) and nine cervical cancer patients (treatment with radiotherapy alone-R) before radiotherapy, 3 weeks after onset of radiotherapy and at the end of radiotherapy. Half of each blood sample was irradiated ex vivo with 2 Gy of gamma radiation in order to check how therapy influenced the sensitivity of PBL to radiation. Analysed endpoints were micronucleus (MN) frequencies, apoptosis frequencies and cell proliferation index. The results were characterised by strong individual variation, especially the MN frequencies and proliferation index. On average, despite higher total dose and larger fields, therapy alone induced the same level of MN in PBL of RC patients as compared to R. This result was accompanied by a higher level of apoptosis and stronger inhibition of cell proliferation in RC patients. The ex vivo dose induced fewer MN, more apoptosis and more strongly inhibited proliferation of PBL of RC as compared to R patients. These results are interpreted as evidence for a sensitizing effect of chemotherapy on radiation cytotoxicity. The possible implications for the risk of second malignant neoplasms are discussed.


Subject(s)
Cisplatin/therapeutic use , Genital Neoplasms, Female/blood , Genital Neoplasms, Female/radiotherapy , Lymphocytes/pathology , Micronuclei, Chromosome-Defective , Neoplasms, Second Primary/blood , Apoptosis/drug effects , Apoptosis/radiation effects , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cisplatin/pharmacology , Female , Genital Neoplasms, Female/drug therapy , Humans , Micronuclei, Chromosome-Defective/drug effects , Micronuclei, Chromosome-Defective/radiation effects , Middle Aged , Risk Factors
6.
Article in English | MEDLINE | ID: mdl-34266630

ABSTRACT

In order to assess the health risk of low-dose radiation to radiation professionals, monitoring is performed through chromosomal aberration analysis and micronuclei (MN) analysis. MN formation has drawbacks for monitoring in the low-dose range. Nucleoplasmic bridge (NPB) analysis, with a lower background level, has good dose-response relationships at both high and relatively low dose ranges. Dicentric and ring chromosomes were analyzed in 199 medical radiation professionals, and NPB/MN yields were analyzed in 205 radiation professionals. The effects of sex, age of donor, types of work, and length of service on these cytogenetic endpoints were also analyzed. The yields of the three cytogenetic endpoints were significantly higher in radiation professionals versus controls. Frequencies of dicentric plus ring chromosomes were affected by length of service. NPB frequencies were influenced by type of work and length of service. MN yields were affected not only by types of work and length of service but also by donor sex and age. In conclusion, dicentric plus ring chromosomes, NPB, and MN can be induced by low-dose radiation in radiation professionals. NPB is a potential biomarker to assess the health risk of occupational low-dose radiation exposure.


Subject(s)
Gamma Rays/adverse effects , Lymphocytes/radiation effects , Occupational Exposure/adverse effects , Radiation Injuries/genetics , Adult , Aged , Cell Nucleus/radiation effects , Chromosome Aberrations/radiation effects , Cytogenetic Analysis/methods , Cytogenetics/methods , DNA Damage/radiation effects , Female , Humans , Male , Micronuclei, Chromosome-Defective/radiation effects , Micronucleus Tests/methods , Middle Aged , Radiation, Ionizing , Young Adult
7.
Environ Mol Mutagen ; 62(7): 422-427, 2021 08.
Article in English | MEDLINE | ID: mdl-34296472

ABSTRACT

It is well-known that the cytotoxicity and mutagenic effects of high dose rate (HDR) ionizing radiation (IR) are increased by increasing the dose but less is known about the effects of chronic low dose rate (LDR). In vitro, we have shown that in addition to the immediate interaction of IR with DNA (the direct and indirect effects), low doses and chronic LDR exposure induce endogenous oxidative stress. During elevated oxidative stress, reactive oxygen species (ROS) react with DNA modifying its structure. Here, BL6 mice were exposed to IR at LDR and HDR and were then sacrificed 3 hours and 3 weeks after exposure to examine early and late effects of IR. The levels of micronuclei, MN, were determined in bone marrow cells. Our data indicate that the effects of 200 mGy on MN-induction are transient, but 500 and 1000 mGy (both HDR and LDR) lead to increased levels of MN up to 3 weeks after the exposure.


Subject(s)
Bone Marrow Cells/pathology , Gamma Rays/adverse effects , Micronuclei, Chromosome-Defective/radiation effects , Whole-Body Irradiation/adverse effects , Animals , Bone Marrow Cells/radiation effects , Dose-Response Relationship, Radiation , Female , Mice , Mice, Inbred C57BL , Micronucleus Tests
8.
Environ Mol Mutagen ; 62(3): 177-184, 2021 03.
Article in English | MEDLINE | ID: mdl-33496960

ABSTRACT

Most women with breast cancer can become pregnant and give birth while undergoing radiation therapy and breastfeeding is generally not contraindicated. The induction of long-lived reactive species in proteins, such as casein by X-ray radiation and DNA damage to unexposed organisms, has been shown when ingesting irradiated cheese. To determine whether exposing lactating rats to X-rays increases the number of micronucleated erythrocytes (MNEs) in peripheral blood of their unexposed or breastfeeding rat pups, 15 female Wistar rats were divided into three groups: Negative control; Experimental group exposed to X-rays, and group exposed to X-rays plus vitamin C. The mothers of groups 2 and 3 were irradiated for three consecutive days after giving birth, returning them to their respective cages each time to continue lactation. A blood sample was taken from the mothers and pups at 0, 24, and 48 hr. Blood smears were stained with acridine orange to analyze MNEs. In mother rats, the frequency of micronucleated polychromatic erythrocytes (MNPCEs) increased significantly at 24 and 48 hr in both study groups exposed to radiation. Likewise, in rat pups the MNPCE and MNE frequencies increased in both groups with radiation and radiation plus vitamin C at 24 and 48 hr, and a protection from vitamin C was observed. In conclusion, the genotoxic damage produced in rat pups that were lactated by mothers irradiated with X-rays is possibly due to the effect of long-lived reactive species that were formed in the breast milk of female Wistar rats during the irradiation process.


Subject(s)
DNA Damage/genetics , Erythrocytes/radiation effects , Lactation/radiation effects , Micronuclei, Chromosome-Defective/radiation effects , Animals , Breast Neoplasms/complications , Breast Neoplasms/radiotherapy , DNA Damage/radiation effects , Erythrocytes/pathology , Female , Lactation/genetics , Male , Micronucleus Tests , Mothers , Pregnancy , Rats , Rats, Wistar , X-Rays/adverse effects
9.
Genes (Basel) ; 11(9)2020 09 17.
Article in English | MEDLINE | ID: mdl-32957448

ABSTRACT

Radioiodine therapy with 131I remains the mainstay of standard treatment for well-differentiated thyroid cancer (DTC). Prognosis is good but concern exists that 131I-emitted ionizing radiation may induce double-strand breaks in extra-thyroidal tissues, increasing the risk of secondary malignancies. We, therefore, sought to evaluate the induction and 2-year persistence of micronuclei (MN) in lymphocytes from 26 131I-treated DTC patients and the potential impact of nine homologous recombination (HR), non-homologous end-joining (NHEJ), and mismatch repair (MMR) polymorphisms on MN levels. MN frequency was determined by the cytokinesis-blocked micronucleus assay while genotyping was performed through pre-designed TaqMan® Assays or conventional PCR-restriction fragment length polymorphism (RFLP). MN levels increased significantly one month after therapy and remained persistently higher than baseline for 2 years. A marked reduction in lymphocyte proliferation capacity was also apparent 2 years after therapy. MLH1 rs1799977 was associated with MN frequency (absolute or net variation) one month after therapy, in two independent groups. Significant associations were also observed for MSH3 rs26279, MSH4 rs5745325, NBN rs1805794, and tumor histotype. Overall, our results suggest that 131I therapy may pose a long-term challenge to cells other than thyrocytes and that the individual genetic profile may influence 131I sensitivity, hence its risk-benefit ratio. Further studies are warranted to confirm the potential utility of these single nucleotide polymorphisms (SNPs) as radiogenomic biomarkers in the personalization of radioiodine therapy.


Subject(s)
Adenocarcinoma, Follicular/pathology , Carcinoma, Papillary/pathology , DNA Repair , Iodine Radioisotopes/therapeutic use , Micronuclei, Chromosome-Defective/radiation effects , Polymorphism, Single Nucleotide , Thyroid Neoplasms/pathology , Adenocarcinoma, Follicular/genetics , Adenocarcinoma, Follicular/radiotherapy , Adult , Aged , Carcinoma, Papillary/genetics , Carcinoma, Papillary/radiotherapy , Female , Follow-Up Studies , Humans , Lymphocytes/pathology , Lymphocytes/radiation effects , Male , Middle Aged , Prognosis , Thyroid Neoplasms/genetics , Thyroid Neoplasms/radiotherapy
10.
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
11.
Genes Genomics ; 42(6): 673-680, 2020 06.
Article in English | MEDLINE | ID: mdl-32314273

ABSTRACT

BACKGROUND: Micronuclei (MN), nuclear bud (NBud), and nucleoplasmic bridge (NPB) are suggested as biomarkers for radiation exposure; however, they have not been extensively studied to understand the underlying mechanisms responsible for their formation. OBJECTIVES: To (1) validate NBud and NPB within the cytokinesis-blocked micronucleus (CBMN) assay as biomarkers for radiation exposure and (2) determine the effects of the DNA repair inhibitors, cytosine arabinoside (Ara C) and 3-aminobenzamide (3-AB) on radiation-induced MN, NBud, and NPB formation. METHODS: Human blood samples were irradiated with 0-3 Gy X-rays and subsequently treated with Ara C and 3-AB. CBMN and chromosome aberration assays were carried out to measure MN, NBud, and NPB and dicentric chromosomes, respectively. RESULTS: The frequency of radiation-induced MN, NBud, and NPB increased in a dose-dependent manner. The frequency of MN, NBud, and NPB was highly and positively correlated with the dicentric chromosome, a standard biomarker for biodosimetry (r > 0.98, p < 0.0001). Furthermore, Ara C increased the frequency of MN, NBud, and NPB, whereas 3-AB increased the frequency of MN and NPB, but not NBud. Further, the potentiating effect of Ara C on the frequency of MN, NBud, and NPB was greater than that of 3-AB. CONCLUSION: Our results validate NBuds and NPBs as independent valuable markers of radiation exposure. Additionally, we suggest that different mechanisms are likely involved in the formation of NBuds and NPBs following X-irradiation; however, additional studies are warranted to better understand the contribution of distinct DNA repair pathways to the formation of radiation-induced damages.


Subject(s)
Benzamides/pharmacology , Cytarabine/pharmacology , Micronuclei, Chromosome-Defective/radiation effects , Adult , Cells, Cultured , DNA Repair/drug effects , Female , Humans , Micronuclei, Chromosome-Defective/drug effects , Radiation Tolerance , X-Rays
12.
Int J Mol Sci ; 21(4)2020 Feb 24.
Article in English | MEDLINE | ID: mdl-32102335

ABSTRACT

During almost 40 years of use, the micronucleus assay (MN) has become one of the most popular methods to assess genotoxicity of different chemical and physical factors, including ionizing radiation-induced DNA damage. In this minireview, we focus on the position of MN among the other genotoxicity tests, its usefulness in different applications and visibility by international organizations, such as International Atomic Energy Agency, Organization for Economic Co-operation and Development and International Organization for Standardization. In addition, the mechanism of micronuclei formation is discussed. Finally, foreseen directions of the MN development are pointed, such as automation, buccal cells MN and chromothripsis phenomenon.


Subject(s)
Lymphocytes/drug effects , Micronuclei, Chromosome-Defective/drug effects , Micronucleus Tests/methods , Mutagenicity Tests/methods , Mutagens/pharmacology , DNA Damage , Forecasting , Humans , Lymphocytes/metabolism , Lymphocytes/radiation effects , Micronuclei, Chromosome-Defective/radiation effects , Micronucleus Tests/trends , Radiation, Ionizing
13.
Article in English | MEDLINE | ID: mdl-32087852

ABSTRACT

We have assessed chromosome-type aberrations and micronuclei in the peripheral lymphocytes of personnel working with C-arm fluoroscopy, multi-slice CT-scan, lithotripsy, and digital radiology medical procedures. The study population comprised of 46 exposed workers and 35 controls matched for age, gender, and other confounding factors. Chromosome-type aberrations and micronuclei were analyzed and compared with occupational dosimetry data. The highest frequency of both chromosome aberrations (1.62 CA/100 cells) and MN (MN = 7.47 ± 2.55) was observed in the operating room group. According to occupational dosimetry, surgeons and medical staff received 0-2.99 mSv over the previous year, well below the limit established by the International Committee on Radiation Protection. An increased level of chromosomal aberrations was observed among workers exposed in the operating rooms. We recommend that operating room radiation safety programs be improved and better supervised, in particular for orthopedic surgeons and personnel performing fluoroscopically guided procedures.


Subject(s)
Chromosome Aberrations/radiation effects , Fluoroscopy/adverse effects , Lithotripsy/adverse effects , Micronuclei, Chromosome-Defective/radiation effects , Occupational Exposure/analysis , Tomography, X-Ray Computed/adverse effects , Adult , Case-Control Studies , Female , Humans , Male , Medical Staff, Hospital , Middle Aged , Operating Rooms , Radiation Dosage
14.
Int J Radiat Biol ; 96(2): 197-205, 2020 02.
Article in English | MEDLINE | ID: mdl-31633434

ABSTRACT

Purpose: To examine the phenomena governing the quantitative relationships between acentric fragments and micronuclei and understand which formulas are useful for curve-fitting of experimental data of micronuclei.Materials and methods: A stochastic model, including the phenomena of inclusion, coalescence and culling out, was developed and applied to experimental data.Results: Probabilities for inclusion/exclusion of acentric fragments into daughter nuclei and for coalescence of many fragments into a single micronucleus were found to be not cell type-specific. The biological basis for this result is explained with the lack of DNA damage checkpoints between metaphase (when acentric fragments are scored) and telophase (when micronuclei are formed). The phenomenon of "culling out" cells with high numbers of acentric fragments is also described, along with its proposed biological mechanism.Conclusions: Apart from complex formulas that describe these phenomena, we discuss which simple formulas can best approximate them and when is the case to use them for curve fitting of micronuclei data.


Subject(s)
Cell Nucleus/metabolism , Chromosome Aberrations/radiation effects , Micronuclei, Chromosome-Defective/radiation effects , Micronucleus Tests , Apoptosis , DNA Damage , Dose-Response Relationship, Radiation , Epithelial Cells/radiation effects , Fibroblasts/radiation effects , Humans , Kinetics , Linear Models , Metaphase , Probability , Telophase
15.
Probl Radiac Med Radiobiol ; 24: 220-234, 2019 Dec.
Article in English, Ukrainian | MEDLINE | ID: mdl-31841469

ABSTRACT

OBJECTIVE: Rat liver stem-like epithelial cells (WB-F344) that under certain conditions may differentiate into hepa- tocyte and biliary lineages were subjected to acute X-irradiation with the aim to examine cell cycle peculiarities dur- ing the course of survival. MATERIALS AND METHODS: Suspensions of WB-F344 cells that grew as a monolayer and reached sub-confluence were irradiated with 1, 5, and 10 Gy of X-rays (2 Gy/min). As an intact control, sham-irradiated cells were used. After irra- diation, cells were plated into 25-cm2 tissue culture flasks to culture them for over several days without reaching contact inhibition. On days 1, 2, 3, and 5 post-irradiation, cells were harvested and examined for nuclear morpholo- gy and DNA ploidy by stoichiometric toluidine blue reaction and image cytometry. On days 7 and 9 post-irradiation, only heavily irradiated (10 Gy) cells were examined. Also, 10 Gy-irradiated cells were chosen for immunofluorescence staining to monitor persistence of DNA lesions (γ-H2AX), cell proliferation (Ki-67), and self-renewal factors charac- teristic for stem cells (OCT4 and NANOG). RESULTS: Radioresistance of WB-F344 cells was evidenced by the findings that they do not undergo rapid and mas- sive cell death that in fact was weakly manifested as apoptotic even in heavily irradiated cells. Instead, there was cell cycle progression delay accompanied by polyploidization (via Ki-67-positive mitotic slippage or via impaired cytokinesis) and micronucleation in a dose-dependent manner, although micronucleation to some extent went ahead of polyploidization. Polyploid cells amenable for recovering from DNA damage can mitotically depolyploidize. Many micronuclei contained γ-H2AX clusters, suggesting isolation of severely damaged DNA fragments. Both factors, OCT4 and NANOG, were expressed in the intact control, but became enhanced after irradiation. CONCLUSIONS: Although the fact of micronucleation is indicative of genotoxic effect, WB-F344 cells can probably escape cell death via sorting of damaged DNA by micronuclei. Induction of polyploidy in these cells can be adaptive to promote cell survival and tissue regeneration with possible involvement of self-renewal mechanism.


Subject(s)
DNA Damage , Epithelial Cells/radiation effects , Liver/radiation effects , Micronuclei, Chromosome-Defective/radiation effects , Polyploidy , Radiation Tolerance , X-Rays/adverse effects , Animals , Cell Culture Techniques , Cell Line , Dose-Response Relationship, Radiation , Epithelial Cells/pathology , Liver/pathology , Radiation Dosage , Rats , Stem Cells/pathology , Stem Cells/radiation effects
16.
Hum Exp Toxicol ; 38(10): 1195-1211, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31256688

ABSTRACT

The present study was premeditated to examine the radioprotective effects of aqueous Aloe vera gel extract against whole-body X-ray irradiation-induced hematological alterations and splenic tissue injury in mice. Healthy male balb/c mice were divided into four groups: group 1, control; group 2, A. vera (50 mg/kg body weight) administered per oral on alternate days for 30 days (15 times); group 3, X-ray exposure of 2 Gy (0.25 Gy twice a day for four consecutive days in the last week of the experimental protocol); and group 4, A. vera + X-ray. X-ray exposure caused alterations in histoarchitecture of spleen along with enhanced clastogenic damage as assessed by micronucleus formation and apoptotic index. Irradiation caused an elevation in proinflammatory cytokines like tumor necrosis factor and interleukin-6, total leucocyte counts, neutrophil counts and decreased platelet counts along with unaltered red blood cell counts and hemoglobin. Irradiation also caused an elevation in reactive oxygen species (ROS), lipid peroxidation (LPO) levels, lactate dehydrogenase activity and alterations in enzymatic and nonenzymatic antioxidant defense mechanism in plasma and spleen. However, administration of A. vera gel extract ameliorated X-ray irradiation-induced elevation in ROS/LPO levels, histopathological and clastogenic damage. It also modulated biochemical indices, inflammatory markers, and hematological parameters. These results collectively indicated that the A. vera gel extract offers protection against whole-body X-ray exposure by virtue of its antioxidant, anti-inflammatory and anti-apoptotic potential.


Subject(s)
Apoptosis/drug effects , Leukocytes/drug effects , Plant Preparations/pharmacology , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/prevention & control , Spleen/drug effects , Administration, Oral , Animals , Antioxidants/metabolism , Apoptosis/radiation effects , Leukocyte Count , Leukocytes/radiation effects , Lipid Peroxidation/drug effects , Lipid Peroxidation/radiation effects , Male , Mice, Inbred BALB C , Micronuclei, Chromosome-Defective/drug effects , Micronuclei, Chromosome-Defective/radiation effects , Platelet Count , Radiation Injuries, Experimental/pathology , Spleen/pathology , Spleen/radiation effects , Whole-Body Irradiation
17.
Methods Mol Biol ; 1984: 23-29, 2019.
Article in English | MEDLINE | ID: mdl-31267416

ABSTRACT

Micronuclei are formed by broken chromosome fragments or chromosomes, which were not appropriately separated into the daughter cells' nuclei after division. The appearance of micronuclei is typically a sign of genotoxic events. Majority of micronuclei are formed by broken acentric fragments, but some micronuclei are formed by centric chromosome fragments which were not appropriately separated to daughter cells' nuclei. Because researchers only need to measure visible micronuclei in binucleated cells, micronuclei analysis is much easier than metaphase chromosome aberration analysis discussed in the previous chapter. This method does not require professional training compared to metaphase chromosome aberration analysis. In addition, one can analyze many samples in a relatively short time. Not only ionizing radiation, but other genotoxic stress also induces micronuclei formation. The background frequency of micronuclei is noticeably higher than chromosome aberrations. But researchers can easily analyze 300-1000 binucleated cells per data point to obtain statistically significant differences of irradiated samples. In this chapter, we will discuss the advantages and preparation of micronuclei samples.


Subject(s)
Micronuclei, Chromosome-Defective/radiation effects , Micronucleus Tests/methods , Radiation, Ionizing , Animals , CHO Cells , Cricetinae , Cricetulus
18.
J Cancer Res Ther ; 15(3): 512-516, 2019.
Article in English | MEDLINE | ID: mdl-31169212

ABSTRACT

INTRODUCTION: Ionizing radiations produce free radicals which are often responsible for DNA damage or cell death. Grape seed extract (GSE) is a natural compound having an antioxidant that protects DNA, lipids, and proteins from free radical damages. In this study, radioprotective effect of the GSE has been investigated in mouse bone marrow cells using micronucleus test. MATERIALS AND METHODS: Four groups of mice were investigated in this study: Mice in Group 1 were subjected to injection of distilled water with no irradiation. Mice in Group 2 were exposed to 3 Gy gamma radiation after the injection of distillated water. Mice in Group 3 were injected with 200 mg/kg of the GSE without any irradiation. In another group, mice were exposed to three gray gamma irradiation after the injection of GSE. Animals were killed, and slides were prepared from the bone marrow cells 24 h after irradiation. The slides were stained with May Grunwald-Giemsa method and analyzed microscopically. The frequency of the micronucleated polychromatic erythrocytes (MnPCEs), micronucleated normochromatic erythrocyte (MnNCEs), and polychromatic erythrocyte/polychromatic erythrocyte + normochromatic erythrocyte (PCE/PCE + NCE) ratios was calculated. RESULTS: Injection of GSE significantly decreased the frequency of MnPCEs (P < 0.0001) and MnNCEs (P < 0.05) and increased the ratio of PCE/PCE + NCE (P < 0.0001) compared to the irradiated control group. DISCUSSION AND CONCLUSIONS: GSE could reduce clastogenic and cytotoxic effects of gamma irradiation in mice bone marrow cells; therefore, it can be concluded that the GSE is a herbal compound with radioprotective effects against gamma irradiation. Free radical scavenging and the antioxidant effects of the GSE probably are responsible mechanisms for the GSE radioprotective effects.


Subject(s)
Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Gamma Rays , Grape Seed Extract/pharmacology , Radiation-Protective Agents/pharmacology , Animals , Bone Marrow Cells/metabolism , Erythrocytes/drug effects , Erythrocytes/radiation effects , Gamma Rays/adverse effects , Grape Seed Extract/chemistry , Male , Mice , Micronuclei, Chromosome-Defective/drug effects , Micronuclei, Chromosome-Defective/radiation effects , Micronucleus Tests , Radiation-Protective Agents/chemistry
19.
Mol Cell ; 75(1): 117-130.e6, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31101499

ABSTRACT

Telomeres are essential for genome stability. Oxidative stress caused by excess reactive oxygen species (ROS) accelerates telomere shortening. Although telomeres are hypersensitive to ROS-mediated 8-oxoguanine (8-oxoG) formation, the biological effect of this common lesion at telomeres is poorly understood because ROS have pleiotropic effects. Here we developed a chemoptogenetic tool that selectively produces 8-oxoG only at telomeres. Acute telomeric 8-oxoG formation increased telomere fragility in cells lacking OGG1, the enzyme that removes 8-oxoG, but did not compromise cell survival. However, chronic telomeric 8-oxoG induction over time shortens telomeres and impairs cell growth. Accumulation of telomeric 8-oxoG in chronically exposed OGG1-deficient cells triggers replication stress, as evidenced by mitotic DNA synthesis at telomeres, and significantly increases telomere losses. These losses generate chromosome fusions, leading to chromatin bridges and micronucleus formation upon cell division. By confining base damage to the telomeres, we show that telomeric 8-oxoG accumulation directly drives telomere crisis.


Subject(s)
Chromosome Aberrations/radiation effects , DNA Glycosylases/genetics , DNA Repair/radiation effects , Genomic Instability/radiation effects , Guanine/analogs & derivatives , Telomere/radiation effects , Cell Division/radiation effects , Cell Line, Tumor , Cell Survival/radiation effects , DNA Damage , DNA Glycosylases/deficiency , DNA Replication/radiation effects , Gene Expression , Guanine/agonists , Guanine/biosynthesis , HeLa Cells , Humans , Light/adverse effects , Micronuclei, Chromosome-Defective/radiation effects , Optogenetics , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoblasts/radiation effects , Oxidative Stress/radiation effects , Singlet Oxygen/agonists , Singlet Oxygen/metabolism , Telomere/metabolism , Telomere Homeostasis/radiation effects
20.
Analyst ; 144(6): 2097-2108, 2019 Mar 11.
Article in English | MEDLINE | ID: mdl-30735207

ABSTRACT

Among different radiotherapy techniques, proton irradiation is an established and effective method for treatment of several types of cancer, because less healthy tissue is exposed with respect to conventional radiotherapy by photons/electrons. Recently, proton therapy has been proposed for the treatment of breast cancer. In vitro studies of proton irradiated normal human breast cells can provide information about cellular radioresponse, particularly as far as healthy tissue is concerned. In this paper, a study of the effects at different time points, following proton irradiation at different doses, of human normal MCF10A breast cells is performed by Raman spectroscopy. The aim of this investigation is to detect the unwanted effects of proton treatment and to investigate the possibility of monitoring them and of making an assessment of the cellular sensitivity by means of such a technique. The obtained results seem to indicate a rather significant sensitivity of MCF10A cells to proton irradiation. In fact, even at doses as low as 0.5 Gy, biological effects are clearly detectable in Raman spectra. In particular, ratiometric analysis of the Raman spectra measured from the nucleoplasm compartment showed that DNA/RNA damage increases with time, suggesting that most cells are unable to repair DNA/RNA broken bonds. The results obtained by the Raman spectroscopy analysis exhibit a similar trend with regard to dose to those obtained by commonly used radiobiological assays (i.e. MTT, clonogenic assay, senescence, apoptosis and necrosis). The results of this study strongly suggest the possibility that the Raman technique can be used to identify molecular markers predicting radiation response.


Subject(s)
Apoptosis/radiation effects , Breast/pathology , Cell Proliferation/radiation effects , DNA Damage , Micronuclei, Chromosome-Defective/radiation effects , Protons/adverse effects , Spectrum Analysis, Raman/methods , Breast/radiation effects , Cells, Cultured , Cellular Senescence , Dose-Response Relationship, Radiation , Female , Humans , Necrosis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...