Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 340
Filtrar
1.
Nature ; 613(7943): 365-374, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36544019

RESUMEN

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.


Asunto(s)
Caenorhabditis elegans , Daño del ADN , Reparación del ADN , Histonas , Animales , Humanos , Masculino , Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Caenorhabditis elegans/efectos de la radiación , Daño del ADN/efectos de la radiación , Inestabilidad Genómica/efectos de la radiación , Histonas/metabolismo , Mutación , Radiación Ionizante , Pérdida del Embrión/genética , Femenino , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN por Unión de Extremidades , ADN Polimerasa theta
2.
Nat Commun ; 13(1): 701, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-35121747

RESUMEN

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.


Asunto(s)
Aneuploidia , Apoptosis/genética , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Células Germinativas/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Estrés Fisiológico/genética , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/genética , Regulación de la Expresión Génica , Inestabilidad Genómica/efectos de la radiación , Calor , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Radiación Ionizante , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Biomed Res Int ; 2021: 2888393, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34926683

RESUMEN

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.


Asunto(s)
Inestabilidad Genómica/efectos de la radiación , Hierro/efectos adversos , Animales , Núcleo Celular/efectos de la radiación , Dietoterapia/métodos , Ingestión de Alimentos/fisiología , Eritrocitos/efectos de la radiación , Femenino , Ratones , Ratones Endogámicos C57BL , Pruebas de Mutagenicidad/métodos , Radiación Ionizante
4.
Sci Rep ; 11(1): 21364, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34725419

RESUMEN

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.


Asunto(s)
Daño del ADN/efectos de la radiación , ADN Polimerasa Dirigida por ADN/genética , Nucleotidiltransferasas/genética , Rayos Ultravioleta/efectos adversos , ADN Polimerasa Dirigida por ADN/metabolismo , Inestabilidad Genómica/efectos de la radiación , Células HEK293 , Humanos , Mutación/efectos de la radiación , Nucleotidiltransferasas/metabolismo , Mapas de Interacción de Proteínas/efectos de la radiación
5.
Int J Mol Sci ; 22(20)2021 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-34681703

RESUMEN

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.


Asunto(s)
Epigénesis Genética/efectos de la radiación , Mitocondrias/metabolismo , Radiación Ionizante , Transducción de Señal/efectos de la radiación , Transición Epitelial-Mesenquimal/efectos de la radiación , Inestabilidad Genómica/efectos de la radiación , Humanos , Mitocondrias/genética , Mitocondrias/efectos de la radiación , Dinámicas Mitocondriales/efectos de la radiación , Estrés Oxidativo/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo
6.
Cell Rep ; 36(5): 109492, 2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34348144

RESUMEN

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.


Asunto(s)
Movimiento Celular , Melanoma/patología , Nevo/patología , Células Madre/patología , Animales , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Epidermis/patología , Epidermis/efectos de la radiación , Amplificación de Genes , Inestabilidad Genómica/efectos de la radiación , Melanocitos/patología , Melanocitos/efectos de la radiación , Melanoma/diagnóstico , Ratones Endogámicos C57BL , Factores de Riesgo , Piel/patología , Piel/efectos de la radiación , Pigmentación de la Piel/efectos de la radiación , Glándulas Sudoríparas/efectos de la radiación , Rayos Ultravioleta
7.
PLoS Genet ; 17(1): e1009302, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33444353

RESUMEN

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.


Asunto(s)
Daño del ADN/efectos de la radiación , Metilación de ADN/genética , Replicación del ADN/genética , Piel/efectos de la radiación , Metilación de ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Replicación del ADN/efectos de la radiación , Fibroblastos/efectos de la radiación , Genoma Humano/genética , Genoma Humano/efectos de la radiación , Inestabilidad Genómica/efectos de la radiación , Genómica/métodos , Humanos , Mutación INDEL/efectos de la radiación , Melanocitos/efectos de la radiación , Mutagénesis/genética , Mutagénesis/efectos de la radiación , Piel/metabolismo , Rayos Ultravioleta/efectos adversos
8.
Plast Reconstr Surg ; 147(1S-2): 7S-14S, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33347069

RESUMEN

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.


Asunto(s)
Envejecimiento/genética , Vuelo Espacial , Acortamiento del Telómero/fisiología , Telómero/metabolismo , Gemelos Monocigóticos/genética , Envejecimiento/efectos de la radiación , Radiación Cósmica/efectos adversos , Inestabilidad Genómica/efectos de la radiación , Humanos , Estudios Longitudinales , Masculino , Telomerasa/metabolismo , Acortamiento del Telómero/efectos de la radiación , Factores de Tiempo
9.
J Invest Dermatol ; 141(4S): 1104-1110, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33358021

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Envejecimiento de la Piel/efectos de la radiación , Neoplasias Cutáneas/etiología , Factor de Crecimiento Transformador beta/metabolismo , Rayos Ultravioleta/efectos adversos , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Inestabilidad Genómica/efectos de la radiación , Humanos , Inhibidores de Puntos de Control Inmunológico , Queratinocitos/efectos de los fármacos , Queratinocitos/patología , Queratinocitos/efectos de la radiación , Ratones , Mutación/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/efectos de la radiación , Piel/efectos de los fármacos , Piel/patología , Piel/efectos de la radiación , Envejecimiento de la Piel/efectos de los fármacos , Envejecimiento de la Piel/genética , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Escape del Tumor/genética , Escape del Tumor/efectos de la radiación , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
10.
PLoS One ; 15(11): e0235998, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33253193

RESUMEN

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.


Asunto(s)
Bronquios/patología , Transformación Celular Neoplásica/patología , Replicación del ADN , Células Epiteliales/patología , Proteína Forkhead Box M1/metabolismo , Inestabilidad Genómica/efectos de la radiación , Estrés Fisiológico , Bronquios/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Daño del ADN , Células Epiteliales/metabolismo , Proteína Forkhead Box M1/genética , Humanos , Radiación Ionizante
11.
Sci Rep ; 10(1): 19422, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33173044

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacología , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Inestabilidad Genómica/efectos de los fármacos , Inestabilidad Genómica/genética , Daño del ADN/efectos de la radiación , Ensayo de Cambio de Movilidad Electroforética , Escherichia coli/efectos de la radiación , Citometría de Flujo , Inestabilidad Genómica/efectos de la radiación , Concentración de Iones de Hidrógeno , Viabilidad Microbiana/efectos de los fármacos , Viabilidad Microbiana/efectos de la radiación , Ácido Nalidíxico/farmacología , Propidio/farmacología , Rifampin/farmacología , Rayos Ultravioleta
12.
Nucleic Acids Res ; 48(19): e111, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-33010172

RESUMEN

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.


Asunto(s)
Partículas alfa/efectos adversos , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Inestabilidad Genómica/efectos de la radiación , Genética de Radiación/instrumentación , Células A549 , Ciclo Celular/efectos de la radiación , Células HeLa , Humanos , Estrés Oxidativo/efectos de la radiación , Saccharomyces cerevisiae , Transducción de Señal/efectos de la radiación
13.
Cell Cycle ; 19(12): 1545-1561, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32380926

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Reparación del ADN/genética , Fosfatasa 3 de Especificidad Dual/metabolismo , Inestabilidad Genómica , Ciclo Celular/efectos de la radiación , Proliferación Celular/genética , Proliferación Celular/efectos de la radiación , Daño del ADN , Reparación del ADN/efectos de la radiación , Silenciador del Gen/efectos de la radiación , Inestabilidad Genómica/efectos de la radiación , Humanos , Dímeros de Pirimidina/metabolismo , Estrés Fisiológico/efectos de la radiación , Rayos Ultravioleta
14.
Biomed Res Int ; 2020: 4703286, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32337251

RESUMEN

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.


Asunto(s)
Radiación Cósmica/efectos adversos , Vuelo Espacial , Rayos Ultravioleta , Animales , Astronautas , Carcinogénesis/efectos de la radiación , Sistema Nervioso Central/efectos de la radiación , Aberraciones Cromosómicas/efectos de la radiación , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Medio Ambiente Extraterrestre , Inestabilidad Genómica/efectos de la radiación , Humanos , Micronúcleos con Defecto Cromosómico/efectos de la radiación , Sustancias Protectoras/farmacología , Dosis de Radiación , Exposición a la Radiación/efectos adversos , Exposición a la Radiación/prevención & control , Estrés Psicológico , Ingravidez
15.
Radiat Res ; 193(5): 451-459, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32150497

RESUMEN

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.


Asunto(s)
Inestabilidad Genómica/efectos de la radiación , Telomerasa/metabolismo , Línea Celular , Supervivencia Celular/efectos de la radiación , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Fase G2/efectos de la radiación , Rayos gamma/efectos adversos , Humanos , Pruebas de Micronúcleos , Recombinasa Rad51/metabolismo , Fase S/efectos de la radiación
16.
Cells ; 9(2)2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-32033081

RESUMEN

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.


Asunto(s)
Adenosina/análogos & derivados , ARN no Traducido/metabolismo , ARN/metabolismo , Rayos Ultravioleta , Adenosina/metabolismo , Animales , Línea Celular Tumoral , Cromatina/metabolismo , Daño del ADN , Desmetilación del ADN/efectos de la radiación , Metilación de ADN/genética , Metilación de ADN/efectos de la radiación , Inestabilidad Genómica/efectos de la radiación , Guanosina/análogos & derivados , Guanosina/metabolismo , Metilación/efectos de la radiación , Ratones , Estrés Fisiológico/efectos de la radiación
17.
Radiat Environ Biophys ; 59(2): 221-236, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32076810

RESUMEN

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.


Asunto(s)
Drosophila melanogaster/efectos de la radiación , Inestabilidad Genómica/efectos de la radiación , Radio (Elemento) , Animales , Ensayo Cometa , Daño del ADN , Drosophila melanogaster/genética , Genoma de los Insectos , Células Germinativas/efectos de la radiación , Masculino , Mutación
18.
Nat Commun ; 11(1): 394, 2020 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-31959748

RESUMEN

Ionising radiation (IR) is a recognised carcinogen responsible for cancer development in patients previously treated using radiotherapy, and in individuals exposed as a result of accidents at nuclear energy plants. However, the mutational signatures induced by distinct types and doses of radiation are unknown. Here, we analyse the genetic architecture of mammary tumours, lymphomas and sarcomas induced by high (56Fe-ions) or low (gamma) energy radiation in mice carrying Trp53 loss of function alleles. In mammary tumours, high-energy radiation is associated with induction of focal structural variants, leading to genomic instability and Met amplification. Gamma-radiation is linked to large-scale structural variants and a point mutation signature associated with oxidative stress. The genomic architecture of carcinomas, sarcomas and lymphomas arising in the same animals are significantly different. Our study illustrates the complex interactions between radiation quality, germline Trp53 deficiency and tissue/cell of origin in shaping the genomic landscape of IR-induced tumours.


Asunto(s)
Carcinogénesis/efectos de la radiación , Inestabilidad Genómica/efectos de la radiación , Neoplasias Inducidas por Radiación/genética , Traumatismos Experimentales por Radiación/genética , Proteína p53 Supresora de Tumor/genética , Animales , Carcinogénesis/genética , Daño del ADN/efectos de la radiación , Análisis Mutacional de ADN , Relación Dosis-Respuesta en la Radiación , Femenino , Amplificación de Genes/efectos de la radiación , Mutación de Línea Germinal , Humanos , Masculino , Ratones , Ratones Noqueados , Neoplasias Inducidas por Radiación/patología , Mutación Puntual/efectos de la radiación , Proteínas Proto-Oncogénicas c-met/genética , Traumatismos Experimentales por Radiación/patología , Secuenciación Completa del Genoma
20.
Nucleic Acids Res ; 48(4): 1652-1668, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31930303

RESUMEN

The excision of mutagenic DNA adducts by the nucleotide excision repair (NER) pathway is essential for genome stability, which is key to avoiding genetic diseases, premature aging, cancer and neurologic disorders. Due to the need to process an extraordinarily high damage density embedded in the nucleosome landscape of chromatin, NER activity provides a unique functional caliper to understand how histone modifiers modulate DNA damage responses. At least three distinct lysine methyltransferases (KMTs) targeting histones have been shown to facilitate the detection of ultraviolet (UV) light-induced DNA lesions in the difficult to access DNA wrapped around histones in nucleosomes. By methylating core histones, these KMTs generate docking sites for DNA damage recognition factors before the chromatin structure is ultimately relaxed and the offending lesions are effectively excised. In view of their function in priming nucleosomes for DNA repair, mutations of genes coding for these KMTs are expected to cause the accumulation of DNA damage promoting cancer and other chronic diseases. Research on the question of how KMTs modulate DNA repair might pave the way to the development of pharmacologic agents for novel therapeutic strategies.


Asunto(s)
Cromatina/genética , Daño del ADN/genética , Histona Metiltransferasas/genética , Histonas/genética , Cromatina/efectos de la radiación , Daño del ADN/efectos de la radiación , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , Inestabilidad Genómica/genética , Inestabilidad Genómica/efectos de la radiación , Histona Metiltransferasas/química , Metilación/efectos de la radiación , Nucleosomas/genética , Nucleosomas/efectos de la radiación , Saccharomyces cerevisiae/genética , Transducción de Señal/efectos de la radiación , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...