Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Am Chem Soc ; 146(30): 20742-20749, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39037865

RESUMO

5-Formyl-2'-deoxycytidine, an intermediate during the erasure of epigenetic marker 5-methyl-2'-deoxycytidine, and 5-formyl-2'-deoxyuridine, an oxidative lesion of thymidine, are naturally occurring DNA modifications. The carbonyl groups of these DNA modifications are the smallest possible photosensitizers and have the potential to generate cyclobutane pyrimidine dimers upon irradiation with UV light. To evidence this damaging potential, ternary DNA architectures were used, in which the photosensitizer and the damage site were located at well-defined positions in the sequences. The quantitative and time-dependent analysis revealed not only the high photodamaging potential of both natural DNA modifications but also the mechanisms for this new pathway to photodamage. 5-Formyl-2'-deoxycytidine is more efficiently generating cyclobutane pyrimidine dimers than 5-formyl-2'-deoxyuridine because the latter is also photochemically converted to 5-carboxy-2'-deoxyuridine. This demonstrates for the first time that epigenetic DNA modifications regulating gene expression interact with sunlight and can induce DNA photodamages.


Assuntos
Dano ao DNA , DNA , Epigênese Genética , Raios Ultravioleta , DNA/química , DNA/efeitos da radiação , Epigênese Genética/efeitos da radiação , Dano ao DNA/efeitos da radiação , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Desoxicitidina/análogos & derivados , Desoxicitidina/química , Dímeros de Pirimidina/química , Desoxiuridina/análogos & derivados , Desoxiuridina/química
2.
Int J Mol Sci ; 25(15)2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39126012

RESUMO

Exposure to ionizing radiation can result in the development of a number of diseases, including cancer, cataracts and neurodegenerative pathologies. Certain occupational groups are exposed to both natural and artificial sources of radiation as a consequence of their professional activities. The development of non-invasive biomarkers to assess the risk of exposure to ionizing radiation for these groups is of great importance. In this context, our objective was to identify epigenetic and molecular biomarkers that could be used to monitor exposure to ionizing radiation. The impact of X-ray exposure on the miRNAs profile and the level of cf mtDNA were evaluated using the RT-PCR method. The levels of pro-inflammatory cytokines in their blood were quantified using the ELISA method. A significant decrease in miR-19a-3p, miR-125b-5p and significant increase in miR-29a-3p was observed in the blood plasma of individuals exposed to X-ray. High levels of pro-inflammatory cytokines and cf mtDNA were also detected. In silico identification of potential targets of these miRNAs was conducted using MIENTURNET. VDAC1 and ALOX5 were identified as possible targets. Our study identified promising biomarkers such as miRNAs and cf mtDNA that showed a dose-dependent effect of X-ray exposure.


Assuntos
Biomarcadores , DNA Mitocondrial , Epigênese Genética , MicroRNAs , Humanos , MicroRNAs/sangue , MicroRNAs/genética , DNA Mitocondrial/sangue , DNA Mitocondrial/genética , Epigênese Genética/efeitos da radiação , Biomarcadores/sangue , Raios X/efeitos adversos , Masculino , Exposição Ocupacional/efeitos adversos , Adulto , Pessoa de Meia-Idade , Citocinas/sangue , Citocinas/genética , Feminino
3.
Int J Mol Sci ; 22(20)2021 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-34681703

RESUMO

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.


Assuntos
Epigênese Genética/efeitos da radiação , Mitocôndrias/metabolismo , Radiação Ionizante , Transdução de Sinais/efeitos da radiação , Transição Epitelial-Mesenquimal/efeitos da radiação , Instabilidade Genômica/efeitos da radiação , Humanos , Mitocôndrias/genética , Mitocôndrias/efeitos da radiação , Dinâmica Mitocondrial/efeitos da radiação , Estresse Oxidativo/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo
4.
Int J Mol Sci ; 22(16)2021 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-34445758

RESUMO

Radiotherapy is still a long way from personalizing cancer treatment plans, and its effectiveness depends on the radiosensitivity of tumor cells. Indeed, therapies that are efficient and successful for some patients may be relatively ineffective for others. Based on this, radiobiological research is focusing on the ability of some reagents to make cancer cells more responsive to ionizing radiation, as well as to protect the surrounding healthy tissues from possible side effects. In this scenario, zebrafish emerged as an effective model system to test for radiation modifiers that can potentially be used for radiotherapeutic purposes in humans. The adoption of this experimental organism is fully justified and supported by the high similarity between fish and humans in both their genome sequences and the effects provoked in them by ionizing radiation. This review aims to provide the literature state of the art of zebrafish in vivo model for radiobiological studies, particularly focusing on the epigenetic and radiomodifying effects produced during fish embryos' and larvae's exposure to radiotherapy treatments.


Assuntos
Epigênese Genética/efeitos da radiação , Radiossensibilizantes/efeitos adversos , Radioterapia/efeitos adversos , Peixe-Zebra , Animais , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/efeitos da radiação , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos da radiação , Modelos Animais
5.
Molecules ; 26(16)2021 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-34443661

RESUMO

Protein methyltransferases are vital to the epigenetic modification of gene expression. Thus, obtaining a better understanding of and control over the regulation of these crucial proteins has significant implications for the study and treatment of numerous diseases. One ideal mechanism of protein regulation is the specific installation of a photolabile-protecting group through the use of photocaged non-canonical amino acids. Consequently, PRMT1 was caged at a key tyrosine residue with a nitrobenzyl-protected Schultz amino acid to modulate protein function. Subsequent irradiation with UV light removes the caging group and restores normal methyltransferase activity, facilitating the spatial and temporal control of PRMT1 activity. Ultimately, this caged PRMT1 affords the ability to better understand the protein's mechanism of action and potentially regulate the epigenetic impacts of this vital protein.


Assuntos
Epigênese Genética/efeitos da radiação , Proteínas Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/genética , Proteínas Repressoras/genética , Sequência de Aminoácidos/genética , Aminoácidos , Epigênese Genética/genética , Expressão Gênica/efeitos da radiação , Humanos , Metilação/efeitos da radiação , Proteínas Metiltransferases/efeitos da radiação , Proteína-Arginina N-Metiltransferases/efeitos da radiação , Proteínas Repressoras/efeitos da radiação , Fatores de Transcrição/genética , Tirosina/química , Raios Ultravioleta
6.
Chromosome Res ; 27(1-2): 41-55, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30610403

RESUMO

Repair of ribosomal DNA (rDNA) is a very important nuclear process due to the most active transcription of ribosomal genes. Proper repair of rDNA is required for physiological biogenesis of ribosomes. Here, we analyzed the epigenetics of the DNA damage response in a nucleolar compartment, thus in the ribosomal genes studied in nonirradiated and UVA-irradiated mouse embryonic fibroblasts (MEFs). We found that the promoter of ribosomal genes is not abundant on H4K20me2, but it is densely occupied by H4K20me3. Ribosomal genes, regulated via UBF1/2 proteins, were characterized by an interaction between UBF1/2 and H4K20me2/me3. This interaction was strengthened by UVA irradiation that additionally causes a focal accumulation of H4K20me3 in the nucleolus. No interaction has been found between UBF1/2 and H3K9me3. Interestingly, UVA irradiation decreases the levels of H3K9me3 and H4K20me3 at 28S rDNA. Altogether, the UVA light affects the epigenetic status of ribosomal genes at 28S rDNA and strengthens an interaction between UBF1/2 proteins and H4K20me2/me3.


Assuntos
DNA Ribossômico/genética , Histonas/metabolismo , Proteínas Pol1 do Complexo de Iniciação de Transcrição/metabolismo , Raios Ultravioleta , Animais , Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA , Epigênese Genética/efeitos da radiação , Imunofluorescência , Regulação da Expressão Gênica/efeitos da radiação , Sequenciamento de Nucleotídeos em Larga Escala , Metilação , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica
7.
Lasers Med Sci ; 35(2): 299-306, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31494789

RESUMO

Differentiation potential of stem cells into various lineages makes these cells as promising sources to treat multiple diseases. In this regard, the use of different strategies and protocols to increase differentiation capacity is highly demanded. Low-level laser therapy, a relatively noninvasive technique, has the capacity to accelerate the healing of numerous injuries and a portion of restorative capacity could be correlated with the stem cell activation and differentiation. Several mechanisms have been diagnosed to participate in orientation of stem cells to functional mature cells. Among them, the status of DNA methylation orchestrates the maintenance of tissue-specific gene expression during the differentiation procedure. DNA methylation is a momentous event in embryogenesis and functional maturation. This review article highlighted the potency of laser irradiation (low-level intensities) in the differentiation of stem cells by modulation of methylation. The analysis of these modalities could help us to understand the underlying mechanisms participating in the therapeutic effects of photobiomodulation.


Assuntos
Diferenciação Celular/efeitos da radiação , Epigênese Genética/efeitos da radiação , Terapia com Luz de Baixa Intensidade , Células-Tronco/citologia , Células-Tronco/efeitos da radiação , Animais , Metilação de DNA/genética , Metilação de DNA/efeitos da radiação , Desmetilação/efeitos da radiação , Humanos , Células-Tronco/metabolismo
8.
Int J Mol Sci ; 21(17)2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32825382

RESUMO

The present system of radiation protection assumes that exposure at low doses and/or low dose-rates leads to health risks linearly related to the dose. They are evaluated by a combination of epidemiological data and radiobiological models. The latter imply that radiation induces deleterious effects via genetic mutation caused by DNA damage with a linear dose-dependence. This picture is challenged by the observation of radiation-induced epigenetic effects (changes in gene expression without altering the DNA sequence) and of non-linear responses, such as non-targeted and adaptive responses, that in turn can be controlled by gene expression networks. Here, we review important aspects of the biological response to ionizing radiation in which epigenetic mechanisms are, or could be, involved, focusing on the possible implications to the low dose issue in radiation protection. We examine in particular radiation-induced cancer, non-cancer diseases and transgenerational (hereditary) effects. We conclude that more realistic models of radiation-induced cancer should include epigenetic contribution, particularly in the initiation and progression phases, while the impact on hereditary risk evaluation is expected to be low. Epigenetic effects are also relevant in the dispute about possible "beneficial" effects at low dose and/or low dose-rate exposures, including those given by the natural background radiation.


Assuntos
Epigênese Genética/efeitos da radiação , Lesões por Radiação/genética , Radiação Ionizante , Animais , Metilação de DNA/efeitos dos fármacos , Relação Dose-Resposta à Radiação , Regulação da Expressão Gênica/efeitos da radiação , Histonas/genética , Histonas/metabolismo , Histonas/efeitos da radiação , Humanos , Neoplasias/etiologia , RNA não Traduzido , Lesões por Radiação/complicações , Lesões por Radiação/etiologia , Proteção Radiológica
9.
Nature ; 500(7463): 472-476, 2013 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-23877069

RESUMO

The dynamic nature of gene expression enables cellular programming, homeostasis and environmental adaptation in living systems. Dissection of causal gene functions in cellular and organismal processes therefore necessitates approaches that enable spatially and temporally precise modulation of gene expression. Recently, a variety of microbial and plant-derived light-sensitive proteins have been engineered as optogenetic actuators, enabling high-precision spatiotemporal control of many cellular functions. However, versatile and robust technologies that enable optical modulation of transcription in the mammalian endogenous genome remain elusive. Here we describe the development of light-inducible transcriptional effectors (LITEs), an optogenetic two-hybrid system integrating the customizable TALE DNA-binding domain with the light-sensitive cryptochrome 2 protein and its interacting partner CIB1 from Arabidopsis thaliana. LITEs do not require additional exogenous chemical cofactors, are easily customized to target many endogenous genomic loci, and can be activated within minutes with reversibility. LITEs can be packaged into viral vectors and genetically targeted to probe specific cell populations. We have applied this system in primary mouse neurons, as well as in the brain of freely behaving mice in vivo to mediate reversible modulation of mammalian endogenous gene expression as well as targeted epigenetic chromatin modifications. The LITE system establishes a novel mode of optogenetic control of endogenous cellular processes and enables direct testing of the causal roles of genetic and epigenetic regulation in normal biological processes and disease states.


Assuntos
Epigênese Genética/genética , Epigênese Genética/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Luz , Optogenética/métodos , Transcrição Gênica/efeitos da radiação , Animais , Proteínas de Arabidopsis/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Cultivadas , Cromatina/genética , Cromatina/efeitos da radiação , Criptocromos/metabolismo , Regulação da Expressão Gênica/genética , Vetores Genéticos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Neurônios/efeitos da radiação , Fatores de Tempo , Transcrição Gênica/genética , Técnicas do Sistema de Duplo-Híbrido , Vigília
10.
Lasers Med Sci ; 34(7): 1465-1472, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30820776

RESUMO

The aim of this study was to evaluate the effect of photobiomodulation therapy (PBMT) on histone 3 acetylation (acH3) and NF-κB expression during oral ulcer healing. A total of 48 male Wistar rats were divided into control group (CG) and PBMT group (n = 24 each). Traumatic ulcers were created in the dorsum of the rats' tongue with a punch tool. Irradiation with InGaAlP laser, 660 nm, 40 mW, 0.04 cm2 spot size, 4 J/cm2, 4 s, and 0.16 J per spot was performed once a day in close contact for 10 consecutive days. CG received only daily handling. Rats were euthanized on days 3, 5, and 10 (n = 8) and were monitored daily to assess wound status. Immunohistochemical analysis for acH3 and NF-κB detection was performed. One thousand epithelial cells were counted, and mean acH3- and NF-κB-positive cells were calculated and compared between the groups. PBMT accelerated the repair of oral ulcers. On day 3, PBMT showed significantly higher means for acH3- and NF-κB-positive cells than CG. On day 5, no difference was observed between the groups concerning both markers. On day 10, PBMT presented lower acH3 and NF-κB means than the control group. We concluded that PBMT stimulates keratinocyte migration in the early stage of oral wound healing and keratinocyte differentiation at the final stage by modulating histone acetylation and NF-κB expression.


Assuntos
Epigênese Genética/efeitos da radiação , Terapia com Luz de Baixa Intensidade , Mucosa Bucal/efeitos da radiação , NF-kappa B/metabolismo , Cicatrização/efeitos da radiação , Acetilação/efeitos da radiação , Animais , Histonas/metabolismo , Masculino , Mucosa Bucal/patologia , Ratos Wistar , Reepitelização/efeitos da radiação
11.
Int J Cancer ; 143(2): 343-354, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29435983

RESUMO

With the increase in the number of long-term cancer survivors worldwide, there is a growing concern about the risk of secondary cancers induced by radiotherapy. Epigenetic modifications of genes associated with carcinogenesis are attractive targets for the prevention of cancer owing to their reversible nature. To identify genes with possible changes in functionally relevant DNA methylation patterns in mammary carcinomas induced by radiation exposure, we performed microarray-based global DNA methylation and expression profiling in γ-ray-induced rat mammary carcinomas and normal mammary glands. The gene expression profiling identified dysregulation of developmentally related genes, including the downstream targets of polycomb repressive complex 2 (PRC2) and overexpression of enhancer of zeste homolog 2, a component of PRC2, in the carcinomas. By integrating expression and DNA methylation profiles, we identified ten hypermethylated and three hypomethylated genes that possibly act as tumor-suppressor genes and oncogenes dysregulated by aberrant DNA methylation; half of these genes encode developmental transcription factors. Bisulfite sequencing and quantitative PCR confirmed the dysregulation of the polycomb-regulated developmentally related transcription-factor genes Dmrt2, Hoxa7, Foxb1, Sox17, Lhx8, Gata3 and Runx1. Silencing of Hoxa7 was further verified by immunohistochemistry. These results suggest that, in radiation-induced mammary gland carcinomas, PRC2-mediated aberrant DNA methylation leads to dysregulation of developmentally related transcription-factor genes. Our findings provide clues to molecular mechanisms linking epigenetic regulation and radiation-induced breast carcinogenesis and underscore the potential of such epigenetic mechanisms as targets for cancer prevention.


Assuntos
Metilação de DNA/efeitos da radiação , Perfilação da Expressão Gênica/métodos , Neoplasias Mamárias Experimentais/genética , Neoplasias Induzidas por Radiação/genética , Análise de Sequência de DNA/métodos , Animais , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Epigênese Genética/efeitos da radiação , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Complexo Repressor Polycomb 2/genética , Ratos , Fatores de Transcrição/genética
12.
Nat Chem Biol ; 12(6): 399-401, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27089030

RESUMO

We engineered a photoactivatable system for rapidly and reversibly exporting proteins from the nucleus by embedding a nuclear export signal in the LOV2 domain from phototropin 1. Fusing the chromatin modifier Bre1 to the photoswitch, we achieved light-dependent control of histone H2B monoubiquitylation in yeast, revealing fast turnover of the ubiquitin mark. Moreover, this inducible system allowed us to dynamically monitor the status of epigenetic modifications dependent on H2B ubiquitylation.


Assuntos
Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Epigênese Genética/efeitos da radiação , Flavoproteínas/metabolismo , Histonas/metabolismo , Luz , Ubiquitinação/efeitos da radiação , Flavoproteínas/química , Flavoproteínas/genética , Sinais de Exportação Nuclear , Fototropinas/química , Domínios Proteicos , Transporte Proteico/efeitos da radiação , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efeitos da radiação , Proteínas de Saccharomyces cerevisiae/metabolismo , Ubiquitina/metabolismo
13.
PLoS Comput Biol ; 13(2): e1005391, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28196079

RESUMO

There is compelling biological data to suggest that cancer arises from a series of mutations in single target cells, resulting in defects in cell renewal and differentiation processes which lead to malignancy. Because much mutagenic damage is expressed following cell division, more-rapidly renewing tissues could be at higher risk because of the larger number of cell replications. Cairns suggested that renewing tissues may reduce cancer risk by partitioning the dividing cell populations into lineages comprising infrequently-dividing long-lived stem cells and frequently-dividing short-lived daughter transit cells. We develop generalizations of three recent cancer-induction models that account for the joint maintenance and renewal of stem and transit cells, also competing processes of partially transformed cell proliferation and differentiation/apoptosis. We are particularly interested in using these models to separately assess the probabilities of mutation and development of cancer associated with "spontaneous" processes and with those linked to a specific environmental mutagen, specifically ionizing radiation or cigarette smoking. All three models demonstrate substantial variation in cancer risks, by at least 20 orders of magnitude, depending on the assumed number of critical mutations required for cancer, and the stem-cell and transition-cell mutation rates. However, in most cases the conditional probabilities of cancer being mutagen-induced range between 7-96%. The relative risks associated with mutagen exposure compared to background rates are also stable, ranging from 1.0-16.0. Very few cancers, generally <0.5%, arise from mutations occurring solely in stem cells rather than in a combination of stem and transit cells. However, for cancers with 2 or 3 critical mutations, a substantial proportion of cancers, in some cases 100%, have at least one mutation derived from a mutated stem cell. Little difference is made to relative risks if competing processes of proliferation and differentiation in the partially transformed stem and transit cell population are allowed for, nor is any difference made if one assumes that transit cells require an extra mutation to confer malignancy from the number required by stem cells. The probability of a cancer being mutagen-induced correlates across cancer sites with the estimated cumulative number of stem cell divisions in the associated tissue (p<0.05), although in some cases there is sensitivity of findings to removal of high-leverage outliers and in some cases only modest variation in probability, but these issues do not affect the validity of the findings. There are no significant correlations (p>0.3) between lifetime cancer-site specific radiation risk and the probability of that cancer being mutagen-induced. These results do not depend on the assumed critical number of mutations leading to cancer, or on the assumed mutagen-associated mutation rate, within the generally-accepted ranges tested. However, there are borderline significant negative correlations (p = 0.08) between the smoking-associated mortality rate difference (current vs former smokers) and the probability of cancer being mutagen-induced. This is only the case where values of the critical number of mutations leading to cancer, k, is 3 or 4 and not for smaller values (1 or 2), but does not strongly depend on the assumed mutagen-associated mutation rate.


Assuntos
Modelos Genéticos , Neoplasias Induzidas por Radiação/epidemiologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos da radiação , Nicotiana/toxicidade , Fumaça , Fumar , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/efeitos da radiação , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/efeitos da radiação , Simulação por Computador , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/genética , Epigênese Genética/efeitos da radiação , Humanos , Mutação , Neoplasias Induzidas por Radiação/genética
14.
Cancer Control ; 25(1): 1073274818812908, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30477310

RESUMO

Lighting technology is rapidly advancing toward shorter wavelength illuminations that offer energy-efficient properties. Along with this advantage, the increased use of such illuminations also poses some health challenges, particularly breast cancer progression. Here, we evaluated the effects of artificial light at night (ALAN) of 4 different spectral compositions (500-595 nm) at 350 Lux on melatonin suppression by measuring its urine metabolite 6-sulfatoxymelatonin, global DNA methylation, tumor growth, metastases formation, and urinary corticosterone levels in 4T1 breast cancer cell-inoculated female BALB/c mice. The results revealed an inverse dose-dependent relationship between wavelength and melatonin suppression. Short wavelength increased tumor growth, promoted lung metastases formation, and advanced DNA hypomethylation, while long wavelength lessened these effects. Melatonin treatment counteracted these effects and resulted in reduced cancer burden. The wavelength suppression threshold for melatonin-induced tumor growth was 500 nm. These results suggest that short wavelength increases cancer burden by inducing aberrant DNA methylation mediated by the suppression of melatonin. Additionally, melatonin suppression and global DNA methylation are suggested as promising biomarkers for early diagnosis and therapy of breast cancer. Finally, ALAN may manifest other physiological responses such as stress responses that may challenge the survival fitness of the animal under natural environments.


Assuntos
Epigênese Genética/efeitos da radiação , Iluminação/efeitos adversos , Neoplasias Pulmonares/epidemiologia , Neoplasias Mamárias Experimentais/etiologia , Melatonina/metabolismo , Animais , Linhagem Celular Tumoral/transplante , Corticosterona/urina , Metilação de DNA/efeitos da radiação , Feminino , Humanos , Neoplasias Pulmonares/secundário , Neoplasias Pulmonares/urina , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/urina , Melatonina/administração & dosagem , Melatonina/análogos & derivados , Melatonina/urina , Camundongos , Camundongos Endogâmicos BALB C , Fotoperíodo
15.
Child Dev ; 89(1): 129-136, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28504324

RESUMO

Mobile phones and other wireless devices that produce electromagnetic fields (EMF) and pulsed radiofrequency radiation (RFR) are widely documented to cause potentially harmful health impacts that can be detrimental to young people. New epigenetic studies are profiled in this review to account for some neurodevelopmental and neurobehavioral changes due to exposure to wireless technologies. Symptoms of retarded memory, learning, cognition, attention, and behavioral problems have been reported in numerous studies and are similarly manifested in autism and attention deficit hyperactivity disorders, as a result of EMF and RFR exposures where both epigenetic drivers and genetic (DNA) damage are likely contributors. Technology benefits can be realized by adopting wired devices for education to avoid health risk and promote academic achievement.


Assuntos
Desempenho Acadêmico , Desenvolvimento do Adolescente/efeitos da radiação , Telefone Celular , Desenvolvimento Infantil/efeitos da radiação , Campos Eletromagnéticos/efeitos adversos , Epigênese Genética/efeitos da radiação , Transtornos Mentais/etiologia , Ondas de Rádio/efeitos adversos , Tecnologia sem Fio , Adolescente , Criança , Humanos
16.
Bioelectromagnetics ; 39(1): 3-14, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28990199

RESUMO

Modulations of epigenetic machinery, namely DNA methylation pattern, histone modification, and non-coding RNAs expression, have been recently included among the key determinants contributing to Parkinson's Disease (PD) aetiopathogenesis and response to therapy. Along this line of reasoning, a set of experimental findings are highlighting the epigenetic-based response to electromagnetic (EM) therapies used to alleviate PD symptomatology, mainly Deep Brain Stimulation (DBS) and Transcranial Magnetic Stimulation (TMS). Notwithstanding the proven efficacy of EM therapies, the precise molecular mechanisms underlying the brain response to these types of stimulations are still far from being elucidated. In this review we provide an overview of the epigenetic changes triggered by DBS and TMS in both PD patients and neurons from different experimental animal models. Furthermore, we also propose a critical overview of the exposure modalities currently applied, in order to evaluate the technical robustness and dosimetric control of the stimulation, which are key issues to be carefully assessed when new molecular findings emerge from experimental studies. Bioelectromagnetics. 39:3-14, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Encéfalo/metabolismo , Encéfalo/efeitos da radiação , Campos Eletromagnéticos , Epigênese Genética/efeitos da radiação , Magnetoterapia/métodos , Doença de Parkinson/genética , Doença de Parkinson/terapia , Animais , Estimulação Encefálica Profunda , Humanos
17.
J Cell Sci ; 128(23): 4380-94, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26446258

RESUMO

Epigenetic mechanisms determine the access of regulatory factors to DNA during events such as transcription and the DNA damage response. However, the global response of histone modifications and chromatin accessibility to UV exposure remains poorly understood. Here, we report that UV exposure results in a genome-wide reduction in chromatin accessibility, while the distribution of the active regulatory mark H3K27ac undergoes massive reorganization. Genomic loci subjected to epigenetic reprogramming upon UV exposure represent target sites for sequence-specific transcription factors. Most of these are distal regulatory regions, highlighting their importance in the cellular response to UV exposure. Furthermore, UV exposure results in an extensive reorganization of super-enhancers, accompanied by expression changes of associated genes, which may in part contribute to the stress response. Taken together, our study provides the first comprehensive resource for genome-wide chromatin changes upon UV irradiation in relation to gene expression and elucidates new aspects of this relationship.


Assuntos
Montagem e Desmontagem da Cromatina/efeitos da radiação , Cromatina/metabolismo , Dano ao DNA , Epigênese Genética/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Animais , Cromatina/genética , Cromatina/patologia , Camundongos , Células NIH 3T3
18.
Proc Natl Acad Sci U S A ; 111(35): 12811-6, 2014 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-25136098

RESUMO

Histone H2B monoubiquitylation plays an important role in RNA polymerase II (RNAPII) elongation. Whether this modification responds to RNAPII stalling is not yet known. We report that both yeast and human cells undergo a rapid and significant H2B deubiquitylation after exposure to UV irradiation. This deubiquitylation occurs concurrently with UV-induced transcription arrest and is significantly reduced in a DNA damage-bypassing RNAPII yeast mutant. Consistent with these results, yeast deubiquitylases Ubp8 and Ubp10 are associated with the RNAPII complex. Moreover, simultaneous deletion of Ubp8 and Ubp10 leads to a lack of H2B deubiquitylation after UV exposure. Consequently, nucleotide excision repair at an actively transcribed gene locus is decreased, whereas UV-induced RNAPII degradation is increased in ubp8Δubp10Δ mutant cells. These results indicate that eukaryotic cells respond to RNAPII arrest by deubiquitylating H2B to coordinate DNA repair and RNAPII degradation.


Assuntos
Dano ao DNA/fisiologia , Reparo do DNA/fisiologia , Histonas/metabolismo , RNA Polimerase II/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Células Cultivadas , Síndrome de Cockayne/genética , Síndrome de Cockayne/metabolismo , Endopeptidases/metabolismo , Epigênese Genética/genética , Epigênese Genética/efeitos da radiação , Humanos , Proteínas Nucleares/metabolismo , Nucleossomos/metabolismo , RNA Polimerase II/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Ubiquitina/metabolismo , Ubiquitina Tiolesterase/metabolismo , Raios Ultravioleta/efeitos adversos
19.
BMC Genomics ; 17(1): 825, 2016 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-27776477

RESUMO

BACKGROUND: Astronauts are exposed to 56Fe ions that may pose a significant health hazard during and following prolonged missions in deep space. We showed previously that object recognition requiring the hippocampus, a structure critical for cognitive function, is affected in 2-month-old mice irradiated with 56Fe ions. Here we examined object recognition in 6-month-old mice irradiated with 56Fe ions, a biological age more relevant to the typical ages of astronauts. Moreover, because the mechanisms mediating the detrimental effects of 56Fe ions on hippocampal function are unclear, we examined changes in hippocampal networks involved in synaptic plasticity and memory, gene expression, and epigenetic changes in cytosine methylation (5mC) and hydroxymethylation (5hmC) that could accompany changes in gene expression. We assessed the effects of whole body 56Fe ion irradiation at early (2 weeks) and late (20 weeks) time points on hippocampus-dependent memory and hippocampal network stability, and whether these effects are associated with epigenetic changes in hippocampal DNA methylation (both 5mC and 5hmC) and gene expression. RESULTS: At the two-week time point, object recognition and network stability were impaired following irradiation at the 0.1 and 0.4 Gy dose, but not following irradiation at the 0.2 Gy dose. No impairments in object recognition or network stability were seen at the 20-week time point at any irradiation dose used. Consistent with this pattern, the significance of pathways for gene categories for 5hmC was lower, though not eliminated, at the 20-week time point compared to the 2-week time point. Similarly, significant changes were observed for 5mC gene pathways at the 2-week time point, but no significant gene categories were observed at the 20-week time point. Only the 5hmC changes tracked with gene expression changes. CONCLUSIONS: Dose- and time-dependent epigenomic remodeling in the hippocampus following 56Fe ion exposure correlates with behavioral changes.


Assuntos
Cognição/efeitos da radiação , Metilação de DNA/efeitos da radiação , Epigênese Genética/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Hipocampo/metabolismo , Hipocampo/efeitos da radiação , Ferro , Radiação Ionizante , Animais , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/efeitos da radiação , Análise por Conglomerados , Perfilação da Expressão Gênica , Ontologia Genética , Imuno-Histoquímica , Masculino , Aprendizagem em Labirinto , Camundongos , Desempenho Psicomotor/efeitos da radiação
20.
Genome Res ; 23(2): 248-59, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23080539

RESUMO

Pluripotent stem cells evade replicative senescence, whereas other primary cells lose their proliferation and differentiation potential after a limited number of cell divisions, and this is accompanied by specific senescence-associated DNA methylation (SA-DNAm) changes. Here, we investigate SA-DNAm changes in mesenchymal stromal cells (MSC) upon long-term culture, irradiation-induced senescence, immortalization, and reprogramming into induced pluripotent stem cells (iPSC) using high-density HumanMethylation450 BeadChips. SA-DNAm changes are highly reproducible and they are enriched in intergenic and nonpromoter regions of developmental genes. Furthermore, SA-hypomethylation in particular appears to be associated with H3K9me3, H3K27me3, and Polycomb-group 2 target genes. We demonstrate that ionizing irradiation, although associated with a senescence phenotype, does not affect SA-DNAm. Furthermore, overexpression of the catalytic subunit of the human telomerase (TERT) or conditional immortalization with a doxycycline-inducible system (TERT and SV40-TAg) result in telomere extension, but do not prevent SA-DNAm. In contrast, we demonstrate that reprogramming into iPSC prevents almost the entire set of SA-DNAm changes. Our results indicate that long-term culture is associated with an epigenetically controlled process that stalls cells in a particular functional state, whereas irradiation-induced senescence and immortalization are not causally related to this process. Absence of SA-DNAm in pluripotent cells may play a central role for their escape from cellular senescence.


Assuntos
Senescência Celular/genética , Metilação de DNA , Células-Tronco Pluripotentes/metabolismo , Adulto , Idoso , Linhagem Celular Transformada , Células Cultivadas , Senescência Celular/efeitos da radiação , Metilação de DNA/efeitos da radiação , Epigênese Genética/efeitos da radiação , Raios gama/efeitos adversos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/efeitos da radiação , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/efeitos da radiação , Pessoa de Meia-Idade , Modelos Biológicos , Células-Tronco Pluripotentes/efeitos da radiação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA