Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
3.
Nucleic Acids Res ; 48(1): 231-248, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31722399

RESUMEN

Cockayne Syndrome (CS) is a severe neurodegenerative and premature aging autosomal-recessive disease, caused by inherited defects in the CSA and CSB genes, leading to defects in transcription-coupled nucleotide excision repair (TC-NER) and consequently hypersensitivity to ultraviolet (UV) irradiation. TC-NER is initiated by lesion-stalled RNA polymerase II, which stabilizes the interaction with the SNF2/SWI2 ATPase CSB to facilitate recruitment of the CSA E3 Cullin ubiquitin ligase complex. However, the precise biochemical connections between CSA and CSB are unknown. The small ubiquitin-like modifier SUMO is important in the DNA damage response. We found that CSB, among an extensive set of other target proteins, is the most dynamically SUMOylated substrate in response to UV irradiation. Inhibiting SUMOylation reduced the accumulation of CSB at local sites of UV irradiation and reduced recovery of RNA synthesis. Interestingly, CSA is required for the efficient clearance of SUMOylated CSB. However, subsequent proteomic analysis of CSA-dependent ubiquitinated substrates revealed that CSA does not ubiquitinate CSB in a UV-dependent manner. Surprisingly, we found that CSA is required for the ubiquitination of the largest subunit of RNA polymerase II, RPB1. Combined, our results indicate that the CSA, CSB, RNA polymerase II triad is coordinated by ubiquitin and SUMO in response to UV irradiation. Furthermore, our work provides a resource of SUMO targets regulated in response to UV or ionizing radiation.


Asunto(s)
ADN Helicasas/genética , Enzimas Reparadoras del ADN/genética , Reparación del ADN , Proteínas de Unión a Poli-ADP-Ribosa/genética , Procesamiento Proteico-Postraduccional , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Factores de Transcripción/genética , Transcripción Genética , Ubiquitina/genética , Línea Celular Transformada , Línea Celular Tumoral , ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/efectos de la radiación , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Osteoblastos/citología , Osteoblastos/metabolismo , Osteoblastos/efectos de la radiación , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sumoilación , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo , Ubiquitinación , Rayos Ultravioleta
4.
Nature ; 523(7558): 53-8, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26106861

RESUMEN

In response to DNA damage, tissue homoeostasis is ensured by protein networks promoting DNA repair, cell cycle arrest or apoptosis. DNA damage response signalling pathways coordinate these processes, partly by propagating gene-expression-modulating signals. DNA damage influences not only the abundance of messenger RNAs, but also their coding information through alternative splicing. Here we show that transcription-blocking DNA lesions promote chromatin displacement of late-stage spliceosomes and initiate a positive feedback loop centred on the signalling kinase ATM. We propose that initial spliceosome displacement and subsequent R-loop formation is triggered by pausing of RNA polymerase at DNA lesions. In turn, R-loops activate ATM, which signals to impede spliceosome organization further and augment ultraviolet-irradiation-triggered alternative splicing at the genome-wide level. Our findings define R-loop-dependent ATM activation by transcription-blocking lesions as an important event in the DNA damage response of non-replicating cells, and highlight a key role for spliceosome displacement in this process.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Daño del ADN/fisiología , Transducción de Señal , Empalmosomas/metabolismo , Empalme Alternativo/fisiología , Línea Celular , Cromatina/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Activación Enzimática , Humanos , Rayos Ultravioleta
5.
Arch Toxicol ; 94(5): 1655-1671, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32189037

RESUMEN

Damage to cellular macromolecules and organelles by chemical exposure evokes activation of various stress response pathways. To what extent different chemical stressors activate common and stressor-specific pathways is largely unknown. Here, we used quantitative phosphoproteomics to compare the signaling events induced by four stressors with different modes of action: the DNA damaging agent: cisplatin (CDDP), the topoisomerase II inhibitor: etoposide (ETO), the pro-oxidant: diethyl maleate (DEM) and the immunosuppressant: cyclosporine A (CsA) administered at an equitoxic dose to mouse embryonic stem cells. We observed major differences between the stressors in the number and identity of responsive phosphosites and the amplitude of phosphorylation. Kinase motif and pathway analyses indicated that the DNA damage response (DDR) activation by CDDP occurs predominantly through the replication-stress-related Atr kinase, whereas ETO triggers the DDR through Atr as well as the DNA double-strand-break-associated Atm kinase. CsA shares with ETO activation of CK2 kinase. Congruent with their known modes of action, CsA-mediated signaling is related to down-regulation of pathways that control hematopoietic differentiation and immunity, whereas oxidative stress is the most prominent initiator of DEM-modulated stress signaling. This study shows that even at equitoxic doses, different stressors induce distinctive and complex phosphorylation signaling cascades.


Asunto(s)
Proteoma/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Diferenciación Celular , Cisplatino/toxicidad , Roturas del ADN de Doble Cadena , Etopósido/toxicidad , Humanos , Ratones , Estrés Oxidativo , Fosforilación , Transducción de Señal , Inhibidores de Topoisomerasa II
6.
Photochem Photobiol Sci ; 17(12): 1842-1852, 2018 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-30065996

RESUMEN

Solar ultraviolet (UV) radiation generates bulky photodimers at di-pyrimidine sites that pose stress to cells and organisms by hindering DNA replication and transcription. In addition, solar UV also induces various types of oxidative DNA lesions and single strand DNA breaks. Relieving toxicity and maintenance of genomic integrity are of clinical importance in relation to erythema/edema and diseases such as cancer, neurodegeneration and premature ageing, respectively. Following solar UV radiation, a network of DNA damage response mechanisms triggers a signal transduction cascade to regulate various genome-protection pathways including DNA damage repair, cell cycle control, apoptosis, transcription and chromatin remodeling. The effects of UVC and UVB radiation on cellular DNA are predominantly accounted for by the formation of photodimers at di-pyrimidine sites. These photodimers are mutagenic: UVC, UVB and also UVA radiation induce a broadly similar pattern of transition mutations at di-pyrimidine sites. The mutagenic potency of solar UV is counteracted by efficient repair of photodimers involving global genome nucleotide excision repair (GG-NER) and transcription-coupled nucleotide excision repair (TC-NER); the latter is a specialized repair pathway to remove transcription-blocking photodimers and restore UV-inhibited transcription. On the molecular level these processes are facilitated and regulated by various post-translational modifications of NER factors and the chromatin substrate. Inherited defects in NER are manifested in different diseases including xeroderma pigmentosum (XP), Cockayne syndrome (CS), UV sensitive syndrome (UVsS) and the photosensitive form of trichothiodystrophy (TTD). XP patients are prone to sunlight-induced skin cancer. UVB irradiated XP and CS knockout mouse models unveiled that only TC-NER counteracts erythema/edema, whereas both GG-NER and TC-NER protect against UVB-induced cancer. Additionally, UVA radiation induces mutations characterized by oxidation-linked signature at non-di-pyrimidine sites. The biological relevance of oxidation damage is demonstrated by the cancer susceptibility of UVB-irradiated mice deficient in repair of oxidation damage, i.e., 8-oxoguanine.


Asunto(s)
Daño del ADN/efectos de la radiación , Rayos Ultravioleta , Animales , Reparación del ADN , Humanos , Dímeros de Pirimidina/química , Dímeros de Pirimidina/metabolismo , Transducción de Señal/efectos de la radiación , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/genética
7.
Mol Cell ; 38(5): 637-48, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20541997

RESUMEN

Transcription-coupled nucleotide excision repair (TC-NER) allows RNA polymerase II (RNAPII)-blocking lesions to be rapidly removed from the transcribed strand of active genes. Defective TCR in humans is associated with Cockayne syndrome (CS), typically caused by defects in either CSA or CSB. Here, we show that CSB contains a ubiquitin-binding domain (UBD). Cells expressing UBD-less CSB (CSB(del)) have phenotypes similar to those of cells lacking CSB, but these can be suppressed by appending a heterologous UBD, so ubiquitin binding is essential for CSB function. Surprisingly, CSB(del) remains capable of assembling nucleotide excision repair factors and repair synthesis proteins around damage-stalled RNAPII, but such repair complexes fail to excise the lesion. Together, our results indicate an essential role for protein ubiquitylation and CSB's UBD in triggering damage incision during TC-NER and allow us to integrate the function of CSA and CSB in a model for the process.


Asunto(s)
ADN Helicasas , Enzimas Reparadoras del ADN , Reparación del ADN , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Línea Celular/efectos de la radiación , Núcleo Celular/metabolismo , Síndrome de Cockayne/genética , Síndrome de Cockayne/metabolismo , Daño del ADN , ADN Helicasas/genética , ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación , Proteínas de Unión a Poli-ADP-Ribosa , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Tetrahidrofolato Deshidrogenasa/genética , Ubiquitina/genética , Rayos Ultravioleta
8.
Mol Cell ; 37(5): 714-27, 2010 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-20227374

RESUMEN

Nucleotide excision repair (NER) is the most versatile DNA repair system that deals with the major UV photoproducts in DNA, as well as many other DNA adducts. The early steps of NER are well understood, whereas the later steps of repair synthesis and ligation are not. In particular, which polymerases are definitely involved in repair synthesis and how they are recruited to the damaged sites has not yet been established. We report that, in human fibroblasts, approximately half of the repair synthesis requires both pol kappa and pol delta, and both polymerases can be recovered in the same repair complexes. Pol kappa is recruited to repair sites by ubiquitinated PCNA and XRCC1 and pol delta by the classical replication factor complex RFC1-RFC, together with a polymerase accessory factor, p66, and unmodified PCNA. The remaining repair synthesis is dependent on pol epsilon, recruitment of which is dependent on the alternative clamp loader CTF18-RFC.


Asunto(s)
Daño del ADN , Reparación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Fibroblastos/enzimología , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas Portadoras/metabolismo , Línea Celular , Senescencia Celular , ADN Polimerasa II/metabolismo , ADN Polimerasa III/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/genética , Fibroblastos/efectos de la radiación , Humanos , Proteínas Nucleares/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa , Antígeno Nuclear de Célula en Proliferación/metabolismo , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Interferencia de ARN , Proteínas Recombinantes de Fusión/metabolismo , Proteína de Replicación C/metabolismo , Factores de Tiempo , Transfección , Ubiquitina-Proteína Ligasas , Ubiquitinación , Rayos Ultravioleta , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
9.
Trends Biochem Sci ; 38(6): 321-30, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23562323

RESUMEN

Distinct types of DNA damage elicit signaling and repair pathways that counteract the adverse effect of DNA lesions to maintain genome stability. The negatively charged polymer poly(ADP-ribose), which is catalyzed by poly(ADP-ribose) polymerase (PARP) enzymes, is a post-translational modification that serves as a chromatin-based platform for the recruitment of a variety of repair factors and chromatin-remodeling enzymes. Recent work implicates PARP3 in the efficient joining of DNA double-strand breaks during non-homologous end-joining (NHEJ), whereas PARP1 modulates the repair of UV-induced DNA lesions. Here we discuss emerging roles of PARP enzymes in mechanistically distinct DNA repair pathways and highlight unresolved issues and questions for future research.


Asunto(s)
Daño del ADN , Reparación del ADN , Poli(ADP-Ribosa) Polimerasas/metabolismo , Rayos Ultravioleta
10.
Nucleic Acids Res ; 43(14): 6919-33, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26101254

RESUMEN

The faithful repair of DNA double-strand breaks (DSBs) is essential to safeguard genome stability. DSBs elicit a signaling cascade involving the E3 ubiquitin ligases RNF8/RNF168 and the ubiquitin-dependent assembly of the BRCA1-Abraxas-RAP80-MERIT40 complex. The association of BRCA1 with ubiquitin conjugates through RAP80 is known to be inhibitory to DSB repair by homologous recombination (HR). However, the precise regulation of this mechanism remains poorly understood. Through genetic screens we identified USP26 and USP37 as key de-ubiquitylating enzymes (DUBs) that limit the repressive impact of RNF8/RNF168 on HR. Both DUBs are recruited to DSBs where they actively remove RNF168-induced ubiquitin conjugates. Depletion of USP26 or USP37 disrupts the execution of HR and this effect is alleviated by the simultaneous depletion of RAP80. We demonstrate that USP26 and USP37 prevent excessive spreading of RAP80-BRCA1 from DSBs. On the other hand, we also found that USP26 and USP37 promote the efficient association of BRCA1 with PALB2. This suggests that these DUBs limit the ubiquitin-dependent sequestration of BRCA1 via the BRCA1-Abraxas-RAP80-MERIT40 complex, while promoting complex formation and cooperation of BRCA1 with PALB2-BRCA2-RAD51 during HR. These findings reveal a novel ubiquitin-dependent mechanism that regulates distinct BRCA1-containing complexes for efficient repair of DSBs by HR.


Asunto(s)
Proteínas Portadoras/antagonistas & inhibidores , Cisteína Endopeptidasas/metabolismo , Endopeptidasas/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Reparación del ADN por Recombinación , Proteína BRCA1/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN , Chaperonas de Histonas , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Recombinasa Rad51/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53 , Ubiquitina/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
11.
Nucleic Acids Res ; 42(7): 4406-13, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24464993

RESUMEN

Rev3, the catalytic subunit of DNA polymerase ζ, is essential for translesion synthesis of cytotoxic DNA photolesions, whereas the Rev1 protein plays a noncatalytic role in translesion synthesis. Here, we reveal that mammalian Rev3(-/-) and Rev1(-/-) cell lines additionally display a nucleotide excision repair (NER) defect, specifically during S phase. This defect is correlated with the normal recruitment but protracted persistence at DNA damage sites of factors involved in an early stage of NER, while repair synthesis is affected. Remarkably, the NER defect becomes apparent only at 2 h post-irradiation indicating that Rev3 affects repair synthesis only indirectly, rather than performing an enzymatic role in NER. We provide evidence that the NER defect is caused by scarceness of Replication protein A (Rpa) available to NER, resulting from its sequestration at stalled replication forks. Also the induction of replicative stress using hydroxyurea precludes the accumulation of Rpa at photolesion sites, both in Rev3(-/-) and in wild-type cells. These data support a model in which the limited Rpa pool coordinates replicative stress and NER, resulting in increased cytotoxicity of ultraviolet light when replicative stress exceeds a threshold.


Asunto(s)
Reparación del ADN , Replicación del ADN , Proteína de Replicación A/metabolismo , Animales , Línea Celular , Proliferación Celular , ADN Polimerasa Dirigida por ADN/genética , Ratones , Transcripción Genética , Rayos Ultravioleta/efectos adversos
12.
Exp Cell Res ; 329(1): 116-23, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25128816

RESUMEN

Nucleotide excision repair (NER) is a key component of the DNA damage response (DDR) and it is essential to safeguard genome integrity against genotoxic insults. The regulation of NER is primarily mediated by protein post-translational modifications (PTMs). The NER machinery removes a wide spectrum of DNA helix distorting lesions, including those induced by solar radiation, through two sub-pathways: global genome nucleotide excision repair (GG-NER) and transcription coupled nucleotide excision repair (TC-NER). Severe clinical consequences associated with inherited NER defects, including premature ageing, neurodegeneration and extreme cancer-susceptibility, underscore the biological relevance of NER. In the last two decades most of the core NER machinery has been elaborately described, shifting attention to molecular mechanisms that either facilitate NER in the context of chromatin or promote the timely and accurate interplay between NER factors and various post-translational modifications. In this review, we summarize and discuss the latest findings in NER. In particular, we focus on emerging factors and novel molecular mechanisms by which NER is regulated.


Asunto(s)
Reparación del ADN/genética , Procesamiento Proteico-Postraduccional , Transcripción Genética/genética , Animales , Humanos
13.
J Cell Sci ; 124(Pt 3): 435-46, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21224401

RESUMEN

Activation of signaling pathways by UV radiation is a key event in the DNA damage response and initiated by different cellular processes. Here we show that non-cycling cells proficient in nucleotide excision repair (NER) initiate a rapid but transient activation of the damage response proteins p53 and H2AX; by contrast, NER-deficient cells display delayed but persistent signaling and inhibition of cell cycle progression upon release from G0 phase. In the absence of repair, UV-induced checkpoint activation coincides with the formation of single-strand DNA breaks by the action of the endonuclease Ape1. Although temporally distinct, activation of checkpoint proteins in NER-proficient and NER-deficient cells depends on a common pathway involving the ATR kinase. These data reveal that damage signaling in non-dividing cells proceeds via NER-dependent and NER-independent processing of UV photolesions through generation of DNA strand breaks, ultimately preventing the transition from G1 to S phase.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Daño del ADN/fisiología , Reparación del ADN/fisiología , Histonas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteína p53 Supresora de Tumor/fisiología , Proteínas de la Ataxia Telangiectasia Mutada , Roturas del ADN de Cadena Simple , Daño del ADN/efectos de la radiación , ADN de Cadena Simple/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/fisiología , Humanos , Fase de Descanso del Ciclo Celular/fisiología , Transducción de Señal/fisiología , Rayos Ultravioleta
14.
BMC Cancer ; 13: 58, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23379751

RESUMEN

BACKGROUND: The risk of developing cutaneous squamous cell carcinoma (SCC) is markedly increased in organ transplant recipients (OTRs) compared to the normal population. Next to sun exposure, the immunosuppressive regimen is an important risk factor for the development of SCC in OTRs. Various gene mutations (e.g. TP53) and genetic alterations (e.g. loss of CDKN2A, amplification of RAS) have been found in SCCs. The aim of this genome-wide study was to identify pathways and genomic alterations that are consistently involved in the formation of SCCs and their precursor lesions, actinic keratoses (AKs). METHODS: To perform the analysis in an isogenic background, RNA and DNA were isolated from SCC, AK and normal (unexposed) epidermis (NS) from each of 13 OTRs. Samples were subjected to genome-wide expression analysis and genome SNP analysis using Illumina's HumanWG-6 BeadChips and Infinium II HumanHap550 Genotyping BeadChips, respectively. mRNA expression results were verified by quantitative PCR. RESULTS: Hierarchical cluster analysis of mRNA expression profiles showed SCC, AK and NS samples to separate into three distinct groups. Several thousand genes were differentially expressed between epidermis, AK and SCC; most upregulated in SCCs were hyperproliferation related genes and stress markers, such as keratin 6 (KRT6), KRT16 and KRT17. Matching to oncogenic pathways revealed activation of downstream targets of RAS and cMYC in SCCs and of NFκB and TNF already in AKs. In contrast to what has been reported previously, genome-wide SNP analysis showed very few copy number variations in AKs and SCCs, and these variations had no apparent relationship with observed changes in mRNA expression profiles. CONCLUSION: Vast differences in gene expression profiles exist between SCC, AK and NS from immunosuppressed OTRs. Moreover, several pathways activated in SCCs were already activated in AKs, confirming the assumption that AKs are the precursor lesions of SCCs. Since the drastic changes in gene expression appeared unlinked to specific genomic gains or losses, the causal events driving SCC development require further investigation. Other molecular mechanisms, such as DNA methylation or miRNA alterations, may affect gene expression in SCCs of OTRs. Further study is required to identify the mechanisms of early activation of NFκB and TNF, and to establish whether these pathways offer a feasible target for preventive intervention among OTRs.


Asunto(s)
Carcinoma de Células Escamosas/genética , Perfilación de la Expresión Génica , Queratosis Actínica/genética , Trasplante de Órganos/efectos adversos , Neoplasias Cutáneas/genética , Anciano , Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/inmunología , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Inmunosupresores/efectos adversos , Queratosis Actínica/etiología , Queratosis Actínica/inmunología , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , ARN Mensajero/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Reproducibilidad de los Resultados , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/inmunología
15.
Mutat Res ; 751-752: 8-14, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24144844

RESUMEN

Ultraviolet radiation is a highly mutagenic agent that damages the DNA by the formation of mutagenic photoproducts at dipyrimidine sites and by oxidative DNA damages via reactive oxygen species (ROS). ROS can also give rise to mutations via oxidation of dNTPs in the nucleotide pool, e.g. 8-oxo-dGTP and 2-OH-dATP and subsequent incorporation during DNA replication. Here we show that expression of human MutT homolog 1 (hMTH1) which sanitizes the nucleotide pool by dephosphorylating oxidized dNTPs, protects against mutagenesis induced by long wave UVA light and by UVB light but not by short wave UVC light. Mutational spectra analyses of UVA-induced mutations at the endogenous Thymidine kinase gene in human lymphoblastoid cells revealed that hMTH1 mainly protects cells from transitions at GC and AT base pairs.


Asunto(s)
Enzimas Reparadoras del ADN/genética , Mutación/efectos de la radiación , Monoéster Fosfórico Hidrolasas/genética , Rayos Ultravioleta , Emparejamiento Base/efectos de la radiación , Línea Celular , Enzimas Reparadoras del ADN/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Mutagénesis/efectos de la radiación , Tasa de Mutación , Nucleótidos/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Timidina Quinasa/genética
17.
Mutat Res ; 735(1-2): 32-8, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22564430

RESUMEN

Calcineurin is a Ca(2+)-dependent serine/threonine phosphatase and the target of the immunosuppressive drugs cyclosporin and tacrolimus, which are used in transplant recipients to prevent rejection. Unfortunately, the therapeutic use of this drugs is complicated by a high incidence of skin malignancy, which has set off a number of studies into the role of calcineurin signaling in skin, particularly with respect to cell cycle control and DNA repair. Both UVA1 radiation and arsenic species are known to promote skin cancer development via production of reactive oxygen species. In light of the well-documented sensitivity of calcineurin to oxidative stress, we examined and compared the effects of UVA1 and arsenite on calcineurin signaling. In this paper, we show that physiologically relevant doses of UVA1 radiation and low micromolar concentrations of arsenite strongly inhibit calcineurin phosphatase activity in Jurkat and skin cells and decrease NFAT nuclear translocation in Jurkat cells. The effects on calcineurin signaling could be partly prevented by inhibition of NADPH oxidase in Jurkat cells or increased dismutation of superoxide in Jurkat and skin cells. In addition, both UVA1 and arsenite decreased NF-κB activity, although at lower concentrations, arsenite enhanced NF-κB activity. These data indicate that UVA1 and arsenite affect a signal transduction route of growingly acknowledged importance in skin and that calcineurin may serve as a potential link between ROS exposure and impaired tumor suppression.


Asunto(s)
Arsenitos/farmacocinética , Calcineurina/metabolismo , Calcineurina/farmacología , Piel/efectos de los fármacos , Piel/efectos de la radiación , Rayos Ultravioleta , Células Cultivadas , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Humanos , Células Jurkat , Factores de Transcripción NFATC/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Piel/metabolismo , Superóxidos/metabolismo , Factor de Transcripción ReIA/metabolismo
18.
DNA Repair (Amst) ; 113: 103305, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35255311

RESUMEN

BACKGROUND: Although cancer risk is assumed to be linear with ionizing radiation (IR) dose, it is unclear to what extent low doses (LD) of IR from medical and occupational exposures pose a cancer risk for humans. Improved mechanistic understanding of the signaling responses to LD may help to clarify this uncertainty. Here, we performed quantitative mass spectrometry-based proteomics and phosphoproteomics experiments, using mouse embryonic stem cells, at 0.5 h and 4 h after exposure to LD (0.1 Gy) and high doses (HD; 1 Gy) of IR. RESULTS: The proteome remained relatively stable (29; 0.5% proteins responded), whereas the phosphoproteome changed dynamically (819; 7% phosphosites changed) upon irradiation. Dose-dependent alterations of 25 IR-responsive proteins were identified, with only four in common between LD and HD. Mitochondrial metabolic proteins and pathways responded to LD, whereas transporter proteins and mitochondrial uncoupling pathways responded to HD. Congruently, mitochondrial respiration increased after LD exposure but decreased after HD exposure. While the bulk of the phosphoproteome response to LD (76%) occurred already at 0.5 h, an equivalent proportion of the phosphosites responded to HD at both time points. Motif, kinome/phosphatome, kinase-substrate, and pathway analyses revealed a robust DNA damage response (DDR) activation after HD exposure but not after LD exposure. Instead, LD-irradiation induced (de)phosphorylation of kinases, kinase-substrates and phosphatases that predominantly respond to reactive oxygen species (ROS) production. CONCLUSION: Our analyses identify discrete global proteome and phosphoproteome responses after LD and HD, uncovering novel proteins and protein (de)phosphorylation events involved in the dose-dependent ionizing radiation responses.

19.
Cells ; 11(23)2022 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-36497055

RESUMEN

Cancer risk after ionizing radiation (IR) is assumed to be linear with the dose; however, for low doses, definite evidence is lacking. Here, using temporal multi-omic systems analyses after a low (LD; 0.1 Gy) or a high (HD; 1 Gy) dose of X-rays, we show that, although the DNA damage response (DDR) displayed dose proportionality, many other molecular and cellular responses did not. Phosphoproteomics uncovered a novel mode of phospho-signaling via S12-PPP1R7, and large-scale dephosphorylation events that regulate mitotic exit control in undamaged cells and the G2/M checkpoint upon IR in a dose-dependent manner. The phosphoproteomics of irradiated DNA double-strand breaks (DSBs) repair-deficient cells unveiled extended phospho-signaling duration in either a dose-dependent (DDR signaling) or independent (mTOR-ERK-MAPK signaling) manner without affecting signal magnitude. Nascent transcriptomics revealed the transcriptional activation of genes involved in NRF2-regulated antioxidant defense, redox-sensitive ERK-MAPK signaling, glycolysis and mitochondrial function after LD, suggesting a prominent role for reactive oxygen species (ROS) in molecular and cellular responses to LD exposure, whereas DDR genes were prominently activated after HD. However, how and to what extent the observed dose-dependent differences in molecular and cellular responses may impact cancer development remain unclear, as the induction of chromosomal damage was found to be dose-proportional (10-200 mGy).


Asunto(s)
Roturas del ADN de Doble Cadena , Radiación Ionizante , Puntos de Control de la Fase G2 del Ciclo Celular , Especies Reactivas de Oxígeno , Transducción de Señal
20.
Mutat Res ; 728(3): 107-17, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21763452

RESUMEN

The accumulation of DNA damage is a slow but hazardous phenomenon that may lead to cell death, accelerated aging features and cancer. One of the most versatile and important defense mechanisms against the accumulation of DNA damage is nucleotide excision repair (NER), in which the Xeroderma pigmentosum group C (XPC) protein plays a prominent role. NER can be divided into global genome repair (GG-NER) and transcription coupled repair (TC-NER). XPC is a key factor in GG-NER where it functions in DNA damage recognition and after which the repair machinery is recruited to eliminate the DNA damage. Defective XPC functioning has been shown to result in a cancer prone phenotype, in human as well as in mice. Mutation accumulation in XPC deficient mice is accelerated and increased, resulting in an increased tumor incidence. More recently XPC has also been linked to functions outside of NER since XPC deficient mice show a divergent tumor spectrum compared to other NER deficient mouse models. Multiple in vivo and in vitro experiments indicate that XPC appears to be involved in the initiation of several DNA damage-induced cellular responses. XPC seems to function in the removal of oxidative DNA damage, redox homeostasis and cell cycle control. We hypothesize that this combination of increased oxidative DNA damage sensitivity, disturbed redox homeostasis together with inefficient cell cycle control mechanisms are causes of the observed increased cancer susceptibility in oxygen exposed tissues. Such a phenotype is absent in other NER-deficient mice, including Xpa.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/metabolismo , Neoplasias/etiología , Envejecimiento , Animales , Puntos de Control del Ciclo Celular , Reparación del ADN , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Polimorfismo de Nucleótido Simple , Xerodermia Pigmentosa/genética , Xerodermia Pigmentosa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA