Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
1.
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
2.
BMC Cancer ; 22(1): 49, 2022 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-34998382

RESUMEN

BACKGROUND: Colorectal cancer (CRC) represents a common malignancy in gastrointestinal tract. Iodine-125 (125I) seed implantation is an emerging treatment technology for unresectable tumors. This study investigated the mechanism of 125I seed in the function of CRC cells. METHODS: The CRC cells were irradiated with different doses of 125I seed (0.4, 0.6 and 0.8 mCi). miR-615 expression in CRC tissues and adjacent tissues was detected by RT-qPCR. miR-615 expression was intervened with miR-615 mimic or miR-615 inhibitor, and then the CRC cells were treated with 5-AZA (methylation inhibitor). The CRC cell growth, invasion and apoptosis were measured. The methylation level of miR-615 promoter region was detected. The xenograft tumor model irradiated by 125I seed was established in nude mice. The methylation of miR-615, Ki67 expression and CRC cell apoptosis were detected. RESULTS: 125I seed irradiation repressed the growth and facilitated apoptosis of CRC cells in a dose-dependent manner. Compared with adjacent tissues, miR-615 expression in CRC tissues was downregulated and miR-615 was poorly expressed in CRC cells. Overexpression of miR-615 suppressed the growth of CRC cells. 125I seed-irradiated CRC cells showed increased miR-615 expression, reduced growth rate and enhanced apoptosis. The methylation level of miR-615 promoter region in CRC cells was decreased after 125I seed treatment. In vivo experiments confirmed that 125I seed-irradiated xenograft tumors showed reduced methylation of the miR-615 promoter and increased miR-615 expression, as well as decreased Ki67 expression and enhanced apoptosis. The target genes of miR-615 and its regulatory downstream pathway were further predicted by bioinformatics analysis. CONCLUSIONS: 125I seed repressed the growth and facilitated the apoptosis of CRC cells by suppressing the methylation of the miR-615 promoter and thus activating miR-615 expression. The possible mechanism was that miR-615-5p targeted MAPK13, thus affecting the MAPK pathway and the progression of CRC.


Asunto(s)
Apoptosis , Neoplasias Colorrectales , Metilación de ADN , Radioisótopos de Yodo/farmacología , MicroARNs/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Braquiterapia , Metilación de ADN/efectos de los fármacos , Metilación de ADN/efectos de la radiación , Humanos , Masculino , Ratones , Ratones Desnudos , MicroARNs/metabolismo , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/efectos de la radiación
3.
Mol Cell Biochem ; 477(1): 267-281, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34708334

RESUMEN

DNA methylation is an epigenetic mechanism, which plays an important role in gene regulation. The present study evaluated DNA methylation profile of LINE1 repeats and promoter methylation of DNA damage response (DDR) and DNA repair (DR) genes (PARP1, ATM, BRCA1, MLH1, XPC, RAD23B, APC, TNFα, DNMT3A, MRE11A, MGMT, CDKN2A, MTHFR) in human peripheral blood mononuclear cells (PBMCs) of healthy donors in response to γ-radiation. Methylation level was correlated with gene expression profile of selected DDR and DR genes (APC, MLH1, PARP1, MRE11A, TNFα, MGMT) to understand their role in gene regulation. Blood samples were collected from 15 random healthy donors, PBMCs were isolated, exposed to 0.1 Gy (low) and 2.0 Gy (high) doses of γ-radiation and proliferated for 48 h and 72 h. Genomic DNA and total RNA were isolated from irradiated PBMCs along with un-irradiated control. Methylation profile was determined from bisulphite converted DNA and amplified by methylation sensitive high resolution melting (MS-HRM) method. Total RNA was converted to cDNA and relative expression was analysed using real time quantitative-PCR. Our results revealed that at 0.1 Gy, MRE11A and TNFα showed significant (P < 0.05) increase in methylation at 72 h. At 2.0 Gy, significant increase (P < 0.05) in methylation profile was observed at LINE1, MRE11A, PARP1, BRCA1, DNMT3A and RAD23B at 48 h and 72 h. PARP1 showed significant positive correlation of methylation status with gene expression. In conclusion, low and high doses of γ-radiation have significant influence on DNA methylation status of LINE1, DDR and DR genes suggesting their potential role as epigenetic signatures in human PBMCs, which can be further explored in human populations.


Asunto(s)
Daño del ADN , Metilación de ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Rayos gamma/efectos adversos , Leucocitos Mononucleares/metabolismo , Elementos de Nucleótido Esparcido Largo , Adulto , Femenino , Humanos , Masculino
4.
PLoS Genet ; 15(11): e1008476, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31738755

RESUMEN

Plants are exposed to the damaging effect of sunlight that induces DNA photolesions. In order to maintain genome integrity, specific DNA repair pathways are mobilized. Upon removal of UV-induced DNA lesions, the accurate re-establishment of epigenome landscape is expected to be a prominent step of these DNA repair pathways. However, it remains poorly documented whether DNA methylation is accurately maintained at photodamaged sites and how photodamage repair pathways contribute to the maintenance of genome/methylome integrities. Using genome wide approaches, we report that UV-C irradiation leads to CHH DNA methylation changes. We identified that the specific DNA repair pathways involved in the repair of UV-induced DNA lesions, Direct Repair (DR), Global Genome Repair (GGR) and small RNA-mediated GGR prevent the excessive alterations of DNA methylation landscape. Moreover, we identified that UV-C irradiation induced chromocenter reorganization and that photodamage repair factors control this dynamics. The methylome changes rely on misregulation of maintenance, de novo and active DNA demethylation pathways highlighting that molecular processes related to genome and methylome integrities are closely interconnected. Importantly, we identified that photolesions are sources of DNA methylation changes in repressive chromatin. This study unveils that DNA repair factors, together with small RNA, act to accurately maintain both genome and methylome integrities at photodamaged silent genomic regions, strengthening the idea that plants have evolved sophisticated interplays between DNA methylation dynamics and DNA repair.


Asunto(s)
Daño del ADN/genética , Metilación de ADN/genética , Reparación del ADN/genética , Epigenoma/genética , Arabidopsis/genética , Arabidopsis/efectos de la radiación , Cromatina/genética , Cromatina/efectos de la radiación , Daño del ADN/efectos de la radiación , Metilación de ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Epigenoma/efectos de la radiación , Genoma de Planta/genética , Genoma de Planta/efectos de la radiación , Rayos Ultravioleta
5.
Plant Physiol ; 182(2): 949-961, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31792150

RESUMEN

Senescence occurs in a programmed manner to dismantle the vegetative tissues and redirect nutrients towards metabolic pathways supporting reproductive success. External factors can trigger the senescence program as an adaptive strategy, indicating that this terminal program is controlled at different levels. It has been proposed that epigenetic factors accompany the reprogramming of the senescent genome; however, the mechanism and extent of this reprogramming remain unknown. Using bisulphite conversion followed by sequencing, we assessed changes in the methylome of senescent Arabidopsis (Arabidopsis thaliana) leaves induced by darkness and monitored their effect on gene and transposable element (TE) expression with transcriptome sequencing. Upon dark-induced senescence, genes controlling chromatin silencing were collectively down-regulated. As a consequence, the silencing of TEs was impaired, causing in particular young TEs to become preferentially reactivated. In parallel, heterochromatin at chromocenters was decondensed. Despite the disruption of the chromatin maintenance network, the global DNA methylation landscape remained highly stable, with localized changes mainly restricted to CHH methylation. Together, our data show that the terminal stage of plant life is accompanied by global changes in chromatin structure but only localized changes in DNA methylation, adding another example of the dynamics of DNA methylation during plant development.


Asunto(s)
Arabidopsis/genética , Senescencia Celular/genética , Metilación de ADN , Elementos Transponibles de ADN , Heterocromatina/metabolismo , Hojas de la Planta/metabolismo , Arabidopsis/crecimiento & desarrollo , Arabidopsis/fisiología , Senescencia Celular/efectos de la radiación , Cromatina/genética , Cromatina/metabolismo , Metilación de ADN/efectos de la radiación , Oscuridad , Regulación hacia Abajo , Sequías , Epigénesis Genética , Epigenoma , Perfilación de la Expresión Génica , Regulación de la Expresión Génica de las Plantas/genética , Silenciador del Gen , Genoma de Planta , Histonas/metabolismo , Hojas de la Planta/genética , Hojas de la Planta/efectos de la radiación , Transcriptoma/genética
6.
Int J Mol Sci ; 22(22)2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34830365

RESUMEN

BACKGROUND: Cerebral ischemia, a common cerebrovascular disease, is one of the great threats to human health and new targets for stroke therapy are needed. The transcriptional activity in the cell is regulated by epigenetic processes such as DNA methylation/demethylation, acetylation/deacetylation, histone methylation, etc. Changes in DNA methylation after ischemia can have both neuroprotective and neurotoxic effects depending on the degree of ischemia damage, the time elapsed after injury, and the site of methylation. METHODS: In this study, we investigated the changes in the expression and intracellular localization of DNA methyltransferase DNMT1, histone methyltransferases SUV39H1, and G9a in penumbra neurons and astrocytes at 4 and 24 h after stroke in the rat cerebral cortex using photothrombotic stroke (PTS) model. Methods of immunofluorescence microscopy analysis, apoptosis analysis, and immunoblotting were used. Additionally, we have studied the effect of DNMT1 and G9a inhibitors on the volume of PTS-induced infarction and apoptosis of penumbra cells in the cortex of mice after PTS. RESULTS: This study has shown that the level of DNMT1 increased in the nuclear and cytoplasmic fractions of the penumbra tissue at 24 h after PTS. Inhibition of DNMT1 by 5-aza-2'-deoxycytidine protected cells of PTS-induced penumbra from apoptosis. An increase in the level of SUV39H1 in the penumbra was found at 24 h after PTS and G9a was overexpressed at 4 and 24 h after PTS. G9a inhibitors A-366 and BIX01294 protected penumbra cells from apoptosis and reduced the volume of PTS-induced cerebral infarction. CONCLUSION: Thus, the data obtained show that DNA methyltransferase DNMT1 and histone methyltransferase G9a can be potential protein targets in ischemic penumbra cells, and their inhibitors are potential neuroprotective agents capable of protecting penumbra cells from postischemic damage to the cerebral cortex.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasa 1/genética , N-Metiltransferasa de Histona-Lisina/genética , Metiltransferasas/genética , Neuronas/metabolismo , Proteínas Represoras/genética , Accidente Cerebrovascular/genética , Animales , Astrocitos/metabolismo , Astrocitos/efectos de la radiación , Corteza Cerebral/metabolismo , Corteza Cerebral/efectos de la radiación , Metilación de ADN/efectos de la radiación , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica/efectos de la radiación , Humanos , Luz , Ratones , Neuronas/patología , Neuronas/efectos de la radiación , Ratas , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/terapia
7.
Angew Chem Int Ed Engl ; 60(24): 13507-13512, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33826797

RESUMEN

5-Methylcytosine (5mC), the central epigenetic mark of mammalian DNA, plays fundamental roles in chromatin regulation. 5mC is written onto genomes by DNA methyltransferases (DNMT), and perturbation of this process is an early event in carcinogenesis. However, studying 5mC functions is limited by the inability to control individual DNMTs with spatiotemporal resolution in vivo. We report light-control of DNMT catalysis by genetically encoding a photocaged cysteine as a catalytic residue. This enables translation of inactive DNMTs, their rapid activation by light-decaging, and subsequent monitoring of de novo DNA methylation. We provide insights into how cancer-related DNMT mutations alter de novo methylation in vivo, and demonstrate local and tuneable cytosine methylation by light-controlled DNMTs fused to a programmable transcription activator-like effector domain targeting pericentromeric satellite-3 DNA. We further study early events of transcriptome alterations upon DNMT-catalyzed cytosine methylation. Our study sets a basis to dissect the order and kinetics of diverse chromatin-associated events triggered by normal and aberrant DNA methylation.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de la radiación , Luz , 5-Metilcitosina/metabolismo , Biocatálisis , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasas/química , ADN (Citosina-5-)-Metiltransferasas/genética , Células HEK293 , Humanos , Mutación , Transcriptoma/efectos de la radiación
8.
Cancer Sci ; 111(4): 1407-1416, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32012407

RESUMEN

Irradiation, or chemoradiotherapy, is a curative treatment for oropharyngeal squamous cell carcinoma (OPSCC). Its invasiveness, however, can often negate its efficacy. Therefore, developing methods to predict which patients would benefit from irradiation is urgent. Promoter DNA hypermethylation was recently reported to correlate with favorable OPSCC prognosis. It is still unclear, however, whether there is an association between promoter DNA methylation and response to irradiation. In this study, we analyzed DNA methylation in the specimens from 40 OPSCC patients who had undergone irradiation, using the Infinium assay. Our results showed significant correlation between high levels of promoter DNA methylation and better response to treatment (P < 0.01). We used the 10 most differentially-methylated genes between responders and non-responders to develop a panel of predictive markers for efficacy. Our panel had high sensitivity, specificity and accuracy (92%, 93% and 93%, respectively). We conducted pyrosequencing to quantitatively validate the methylation levels of 8 of the 10 marker genes (ROBO1, ULK4P3, MYOD1, LBX1, CACNA1A, IRX4, DPYSL3 and ELAVL2) obtained by Infinium. The validation by pyrosequencing showed that these 8 genes had a high prediction performance for the training set of 40 specimens and for a validation set of 35 OPSCC specimens, showing 96% sensitivity, 89% specificity and 94% accuracy. Methylation of these markers correlated significantly with better progression-free and overall survival rates, regardless of human papillomavirus status. These results indicate that increased DNA methylation is associated with better responses to irradiation therapy and that DNA methylation can help establish efficacy prediction markers in OPSCC.


Asunto(s)
Biomarcadores de Tumor/genética , Metilación de ADN/efectos de la radiación , Neoplasias Orofaríngeas/radioterapia , Infecciones por Papillomavirus/radioterapia , Anciano , Metilación de ADN/genética , Epigenómica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Orofaríngeas/genética , Neoplasias Orofaríngeas/patología , Neoplasias Orofaríngeas/virología , Papillomaviridae/patogenicidad , Papillomaviridae/efectos de la radiación , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Regiones Promotoras Genéticas/efectos de la radiación
9.
Lasers Med Sci ; 35(2): 299-306, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31494789

RESUMEN

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.


Asunto(s)
Diferenciación Celular/efectos de la radiación , Epigénesis Genética/efectos de la radiación , Terapia por Luz de Baja Intensidad , Células Madre/citología , Células Madre/efectos de la radiación , Animales , Metilación de ADN/genética , Metilación de ADN/efectos de la radiación , Desmetilación/efectos de la radiación , Humanos , Células Madre/metabolismo
10.
Int J Mol Sci ; 21(11)2020 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-32512748

RESUMEN

Embryonic development is particularly vulnerable to stress and DNA damage, as mutations can accumulate through cell proliferation in a wide number of cells and organs. However, the biological effects of chronic exposure to ionising radiation (IR) at low and moderate dose rates (< 6 mGy/h) remain largely controversial, raising concerns for environmental protection. The present study focuses on the molecular effects of IR (0.005 to 50 mGy/h) on zebrafish embryos at the gastrula stage (6 hpf), at both the transcriptomics and epigenetics levels. Our results show that exposure to IR modifies the expression of genes involved in mitochondrial activity from 0.5 to 50 mGy/h. In addition, important developmental pathways, namely, the Notch, retinoic acid, BMP and Wnt signalling pathways, were altered at 5 and 50 mGy/h. Transcriptional changes of genes involved in the morphogenesis of the ectoderm and mesoderm were detected at all dose rates, but were prominent from 0.5 to 50 mGy/h. At the epigenetic level, exposure to IR induced a hypomethylation of DNA in the promoter of genes that colocalised with both H3K27me3 and H3Kme4 histone marks and correlated with changes in transcriptional activity. Finally, pathway enrichment analysis demonstrated that the DNA methylation changes occurred in the promoter of important developmental genes, including morphogenesis of the ectoderm and mesoderm. Together, these results show that the transcriptional program regulating morphogenesis in gastrulating embryos was modified at dose rates greater than or equal to 0.5 mGy/h, which might predict potential neurogenesis and somitogenesis defects observed at similar dose rates later in development.


Asunto(s)
Metilación de ADN/efectos de la radiación , Regulación del Desarrollo de la Expresión Génica/efectos de la radiación , Morfogénesis/genética , Organogénesis/genética , Regiones Promotoras Genéticas , Radiación Ionizante , Activación Transcripcional/efectos de la radiación , Pez Cebra/genética , Animales , Biología Computacional/métodos , Ectodermo/embriología , Ectodermo/metabolismo , Ectodermo/efectos de la radiación , Perfilación de la Expresión Génica , Mesodermo/embriología , Mesodermo/metabolismo , Mesodermo/efectos de la radiación , Transcriptoma , Pez Cebra/embriología
11.
Int J Cancer ; 143(2): 343-354, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29435983

RESUMEN

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.


Asunto(s)
Metilación de ADN/efectos de la radiación , Perfilación de la Expresión Génica/métodos , Neoplasias Mamarias Experimentales/genética , Neoplasias Inducidas por Radiación/genética , Análisis de Secuencia de ADN/métodos , Animales , Proteína Potenciadora del Homólogo Zeste 2/genética , Epigénesis Genética/efectos de la radiación , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Complejo Represivo Polycomb 2/genética , Ratas , Factores de Transcripción/genética
12.
Plant Cell Environ ; 41(7): 1657-1672, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29707792

RESUMEN

The capacity for plant stress priming and memory and the notion of this being underpinned by DNA methylation-mediated memory is an appealing hypothesis for which there is mixed evidence. We previously established a lack of drought-induced methylome variation in Arabidopsis thaliana (Arabidopsis); however, this was tied to only minor observations of physiological memory. There are numerous independent observations demonstrating that photoprotective mechanisms, induced by excess-light stress, can lead to robust programmable changes in newly developing leaf tissues. Although key signalling molecules and transcription factors are known to promote this priming signal, an untested question is the potential involvement of chromatin marks towards the maintenance of light stress acclimation, or memory. Thus, we systematically tested our previous hypothesis of a stress-resistant methylome using a recurring excess-light stress, then analysing new, emerging, and existing tissues. The DNA methylome showed negligible stress-associated variation, with the vast majority attributable to stochastic differences. Yet, photoacclimation was evident through enhanced photosystem II performance in exposed tissues, and nonphotochemical quenching and fluorescence decline ratio showed evidence of mitotic transmission. Thus, we have observed physiological acclimation in new and emerging tissues in the absence of substantive DNA methylome changes.


Asunto(s)
Metilación de ADN/efectos de la radiación , Arabidopsis/genética , Arabidopsis/metabolismo , Arabidopsis/fisiología , Arabidopsis/efectos de la radiación , Clorofila/metabolismo , Genoma de Planta/genética , Luz , Fotosíntesis/efectos de la radiación , Complejo de Proteína del Fotosistema II/efectos de la radiación , Análisis de Secuencia de ADN , Estrés Fisiológico , Xantófilas/metabolismo
13.
Acta Neuropathol ; 135(4): 601-615, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29368212

RESUMEN

The optimal treatment for patients with low-grade glioma (LGG) WHO grade II remains controversial. Overall survival ranges from 2 to over 15 years depending on molecular and clinical factors. Hence, risk-adjusted treatments are required for optimizing outcome and quality of life. We aim at identifying mechanisms and associated molecular markers predictive for benefit from radiotherapy (RT) or temozolomide (TMZ) in LGG patients treated in the randomized phase III trial EORTC 22033. As candidate biomarkers for these genotoxic treatments, we considered the DNA methylome of 410 DNA damage response (DDR) genes. We first identified 62 functionally relevant CpG sites located in the promoters of 24 DDR genes, using the LGG data from The Cancer Genome Atlas. Then we tested their association with outcome [progression-free survival (PFS)] depending on treatment in 120 LGG patients of EORTC 22033, whose tumors were mutant for isocitrate dehydrogenase 1 or 2 (IDHmt), the molecular hallmark of LGG. The results suggested that seven CpGs of four DDR genes may be predictive for longer PFS in one of the treatment arms that comprised MGMT, MLH3, RAD21, and SMC4. Most interestingly, the two CpGs identified for MGMT are the same, previously selected for the MGMT-STP27 score that is used to determine the methylation status of the MGMT gene. This score was higher in the LGG with 1p/19q codeletion, in this and other independent LGG datasets. It was predictive for PFS in the TMZ, but not in the RT arm of EORTC 22033. The results support the hypothesis that a high score predicts benefit from TMZ treatment for patients with IDHmt LGG, regardless of the 1p/19q status. This MGMT methylation score may identify patients who benefit from first-line treatment with TMZ, to defer RT for long-term preservation of cognitive function and quality of life.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Metilación de ADN , Receptores con Dominio Discoidina/genética , Glioma/genética , Glioma/terapia , Adulto , Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/patología , Islas de CpG , ADN , Metilación de ADN/efectos de los fármacos , Metilación de ADN/efectos de la radiación , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Epigénesis Genética , Femenino , Glioma/patología , Humanos , Isocitrato Deshidrogenasa/genética , Masculino , Clasificación del Tumor , Supervivencia sin Progresión , Regiones Promotoras Genéticas , Temozolomida/uso terapéutico , Resultado del Tratamiento , Proteínas Supresoras de Tumor/genética
14.
Cancer Control ; 25(1): 1073274818812908, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30477310

RESUMEN

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.


Asunto(s)
Epigénesis Genética/efectos de la radiación , Iluminación/efectos adversos , Neoplasias Pulmonares/epidemiología , Neoplasias Mamarias Experimentales/etiología , Melatonina/metabolismo , Animales , Línea Celular Tumoral/trasplante , Corticosterona/orina , Metilación de ADN/efectos de la radiación , Femenino , Humanos , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/orina , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/orina , Melatonina/administración & dosificación , Melatonina/análogos & derivados , Melatonina/orina , Ratones , Ratones Endogámicos BALB C , Fotoperiodo
15.
Indian J Med Res ; 147(2): 151-157, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29806603

RESUMEN

BACKGROUND & OBJECTIVES: Invasive cervical cancer patients are primarily treated with chemoradiation therapy. The overall and disease-free survival in these patients is variable and depends on the tumoral response apart from the tumour stage. This study was undertaken to assess whether in vivo changes in gene promoter methylation and transcript expression in invasive cervical cancer were induced by chemoradiation. Hence, paired pre- and post-treatment biopsy samples were evaluated for in vivo changes in promoter methylation and transcript expression of 10 genes (ESR1, BRCA1, RASSF1A, MYOD1, MLH1, hTERT, MGMT, DAPK1, BAX and BCL2L1) in response to chemoradiation therapy. METHODS: In patients with locally advanced invasive cervical cancer, paired pre- and post-treatment biopsies after 10 Gy chemoradiation were obtained. DNA/RNA was extracted and gene promoter methylation status was evaluated by custom-synthesized methylation PCR arrays, and the corresponding gene transcript expression was determined by absolute quantification method using quantitative reverse transcription PCR. RESULTS: Changes in the gene promoter methylation as well as gene expression following chemoradiation therapy were observed. BAX promoter methylation showed a significant increase (P< 0.01) following treatment. There was a significant increase in the gene transcript expression of BRCA1 (P< 0.01), DAPK1 and ESR1 (P< 0.05), whereas MYOD1 and MLH1 gene transcript expression was significantly decreased (P< 0.05) following treatment. INTERPRETATION & CONCLUSIONS: The findings of our study show that chemoradiation therapy can induce epigenetic alterations as well as affect gene expression in tissues of invasive cervical cancer which may have implications in determining radiation response.


Asunto(s)
Metilación de ADN/genética , Proteínas de Neoplasias/genética , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/radioterapia , Adulto , Anciano , Metilación de ADN/efectos de los fármacos , Metilación de ADN/efectos de la radiación , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Persona de Mediana Edad , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de la radiación , Proteínas Supresoras de Tumor/genética , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
16.
Annu Rev Med ; 66: 129-43, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25423595

RESUMEN

Cellular responses to DNA damage are important determinants of both cancer development and cancer outcome following radiation therapy and chemotherapy. Identification of molecular pathways governing DNA damage signaling and DNA repair in response to different types of DNA lesions allows for a better understanding of the effects of radiation and chemotherapy on normal and tumor cells. Although dysregulation of the DNA damage response (DDR) is associated with predisposition to cancer development, it can also result in hypersensitivity or resistance of tumors to therapy and can be exploited for improvement of cancer treatment. We highlight the DDR pathways that are activated after treatment with radiation and different classes of chemotherapeutic drugs and describe mechanisms determining tumor sensitivity and resistance to these agents. Further, we discuss approaches to enhance tumor sensitivity to radiation and chemotherapy by modulating the DDR with a goal of enhancing the effectiveness of cancer therapies.


Asunto(s)
Antineoplásicos/uso terapéutico , Daño del ADN/genética , Reparación del ADN/genética , Neoplasias/genética , Radioterapia , Antineoplásicos/farmacología , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Metilación de ADN/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Humanos , Neoplasias/terapia
17.
Am J Hum Biol ; 29(5)2017 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-28432711

RESUMEN

OBJECTIVES: The vitamin D receptor (VDR) is a member of the nuclear receptor family of transcription factors. We examined whether degree of VDR gene methylation acts as a molecular adaptation to light exposure. We explored this in the context of photoperiod at conception, recent UV irradiance at 305 nm, and gene-latitude effects. METHODS: Eighty subjects were examined for VDR gene-CpG island methylation density. VDR gene variants were also examined by PCR-RFLP. RESULTS: Photoperiod at conception was significantly positively related to VDR methylation density, explaining 17% of the variance in methylation (r2 = 0.17; P = .001). Within this model, photoperiod at conception and plasma 25(OH)D independently predicted methylation density at the VDR-CpG island. Recent UV exposure at 305 nm led to a fivefold increase in mean methylation density (P = .02). Again, UV exposure and plasma 25(OH)D independently predicted methylation density at the VDR-CpG island. In the presence of the BsmI mutant allele, methylation density was increased (P = .01), and in the presence of the TaqI or FokI mutant allele, methylation density was decreased (P = .007 and .04 respectively). Multivariate modelling suggests plasma 25(OH)D, photoperiod at conception, recent solar irradiance, and VDR genotype combine as independent predictors of methylation at the VDR-CpG island, explaining 34% of the variance in methylation (R2 = 0.34, P < .0001). CONCLUSIONS: Duration of early-life light exposure and strength of recent irradiance, along with latitudinal genetic factors, influence degree of VDR gene methylation consistent with this epigenetic phenomenon being a molecular adaptation to variation in ambient light exposure. Findings contribute to our understanding of human biology.


Asunto(s)
Metilación de ADN/efectos de la radiación , Polimorfismo Genético , Receptores de Calcitriol/genética , Rayos Ultravioleta/efectos adversos , Anciano , Anciano de 80 o más Años , Estudios Transversales , Femenino , Humanos , Masculino , Nueva Gales del Sur
18.
Int J Mol Sci ; 18(7)2017 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-28677663

RESUMEN

Long Interspersed Nuclear Element 1 (LINE-1) retrotransposons are the major repetitive elements in mammalian genomes. LINE-1s are well-accepted as driving forces of evolution and critical regulators of the expression of genetic information. Alterations in LINE-1 DNA methylation may lead to its aberrant activity and are reported in virtually all human cancers and in experimental carcinogenesis. In this study, we investigated the endogenous DNA methylation status of the 5' untranslated region (UTR) of LINE-1 elements in the bone marrow hematopoietic stem cells (HSCs), hematopoietic progenitor cells (HPCs), and mononuclear cells (MNCs) in radioresistant C57BL/6J and radiosensitive CBA/J mice and in response to ionizing radiation (IR). We demonstrated that basal levels of DNA methylation within the 5'-UTRs of LINE-1 elements did not differ significantly between the two mouse strains and were negatively correlated with the evolutionary age of LINE-1 elements. Meanwhile, the expression of LINE-1 elements was higher in CBA/J mice. At two months after irradiation to 0.1 or 1 Gy of 137Cs (dose rate 1.21 Gy/min), significant decreases in LINE-1 DNA methylation in HSCs were observed in prone to radiation-induced carcinogenesis CBA/J, but not C57BL/6J mice. At the same time, no residual DNA damage, increased ROS, or changes in the cell cycle were detected in HSCs of CBA/J mice. These results suggest that epigenetic alterations may potentially serve as driving forces of radiation-induced carcinogenesis; however, future studies are needed to demonstrate the direct link between the LINE-1 DNA hypomethylation and radiation carcinogenesis.


Asunto(s)
Metilación de ADN/efectos de la radiación , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de la radiación , Elementos de Nucleótido Esparcido Largo , Radiación Ionizante , Animales , Daño del ADN , Relación Dosis-Respuesta en la Radiación , Regulación de la Expresión Génica/efectos de la radiación , Hematopoyesis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Retroelementos , Especificidad de la Especie
19.
Inflammopharmacology ; 25(2): 203-210, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28190128

RESUMEN

OBJECTIVE: In the present study, DNA methylation level of CD4+ T cells exposed to ultraviolet B (UVB) was investigated and its potential mechanisms were also explored. METHODS: CD4+ T cells from 12 cases of healthy subjects and 33 cases of SLE patients were isolated and exposed to different dosages (0, 50, 100 mJ/cm2) of UVB. Further, SLE patients were divided into two groups: active SLE group (22 cases, SLEDAI scores >4) and inactive SLE group (11 cases, SLEDAI scores ≤4). DNA methylation was evaluated by the Methylamp™ Global DNA Methylation Quantification Ultra Kit. The mRNA and protein expression levels of DNA methyltransferases (DNMT1 and DNMT3A) were detected by real-time PCR and western blot, respectively. RESULTS: The levels of DNA methylation and DNMT3A mRNA in SLE patients were significantly decreased compared with those in healthy subjects at baseline. After different dosages of ultraviolet irradiation (0, 50 and 100 mJ/cm2), DNA methylation levels of CD4+ T cells were all reduced in a dose-dependent manner in three subgroups. Additionally, 100 mJ/cm2 ultraviolet irradiation in active SLE group contributed to a significant decrease of both DNA methylation and DNMT3A mRNA levels in CD4+ T cells. UVB exposure had no significant effects on expression levels of DNMT1 mRNA and protein and DNMT3A protein. CONCLUSION: UVB decreases DNA methylation level of CD4+ T cells in SLE patients probably via inhibiting DNMT3A mRNA expression level, which needs to be further explored.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de la radiación , ADN (Citosina-5-)-Metiltransferasas/efectos de la radiación , Metilación de ADN/efectos de la radiación , Lupus Eritematoso Sistémico , Rayos Ultravioleta , Adulto , Linfocitos T CD4-Positivos/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/fisiología , ADN Metiltransferasa 3A , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/patología , Masculino , Persona de Mediana Edad , Rayos Ultravioleta/efectos adversos
20.
J Biol Chem ; 290(44): 26597-609, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26354431

RESUMEN

Sunlight-induced C to T mutation hot spots in skin cancers occur primarily at methylated CpG sites that coincide with sites of UV-induced cyclobutane pyrimidine dimer (CPD) formation. The C and 5-methyl-C in CPDs are not stable and deaminate to U and T, respectively, which leads to the insertion of A by the DNA damage bypass polymerase η, thereby defining a probable mechanism for the origin of UV-induced C to T mutations. Deamination rates for T(m)CG CPDs have been found to vary 12-fold with rotational position in a nucleosome in vitro. To determine the influence of nucleosome structure on deamination rates in vivo, we determined the deamination rates of CPDs at TCG sites in a stably positioned nucleosome within the FOS promoter in HeLa cells. A procedure for in vivo hydroxyl radical footprinting with Fe-EDTA was developed, and, together with results from a cytosine methylation protection assay, we determined the translational and rotational positions of the TCG sites. Consistent with the in vitro observations, deamination was slower for one CPD located at an intermediate rotational position compared with two other sites located at outside positions, and all were much faster than for CPDs at non-TCG sites. Photoproduct formation was also highly suppressed at one site, possibly due to its interaction with a histone tail. Thus, it was shown that CPDs of TCG sites deaminate the fastest in vivo and that nucleosomes can modulate both their formation and deamination, which could contribute to the UV mutation hot spots and cold spots.


Asunto(s)
ADN Polimerasa Dirigida por ADN/química , Histonas/química , Radical Hidroxilo/química , Nucleosomas/metabolismo , Dímeros de Pirimidina/química , Proteínas Recombinantes de Fusión/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Ensamble y Desensamble de Cromatina/efectos de la radiación , Metilación de ADN/efectos de la radiación , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Desaminación , Desoxirribodipirimidina Fotoliasa/química , Desoxirribodipirimidina Fotoliasa/genética , Desoxirribodipirimidina Fotoliasa/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Células HeLa , Histonas/genética , Histonas/metabolismo , Humanos , Radical Hidroxilo/metabolismo , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Nucleosomas/química , Nucleosomas/efectos de la radiación , Regiones Promotoras Genéticas , Dímeros de Pirimidina/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA