Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.109
Filtrar
1.
Nat Commun ; 15(1): 6223, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39043658

RESUMEN

Transcription coupled-nucleotide excision repair (TC-NER) removes DNA lesions that block RNA polymerase II (Pol II) transcription. A key step in TC-NER is the recruitment of the TFIIH complex, which initiates DNA unwinding and damage verification; however, the mechanism by which TFIIH is recruited during TC-NER, particularly in yeast, remains unclear. Here, we show that the C-terminal domain (CTD) of elongation factor-1 (Elf1) plays a critical role in TC-NER in yeast by binding TFIIH. Analysis of genome-wide repair of UV-induced cyclobutane pyrimidine dimers (CPDs) using CPD-seq indicates that the Elf1 CTD in yeast is required for efficient TC-NER. We show that the Elf1 CTD binds to the pleckstrin homology (PH) domain of the p62 subunit of TFIIH in vitro, and identify a putative TFIIH-interaction region (TIR) in the Elf1 CTD that is important for PH binding and TC-NER. The Elf1 TIR shows functional, structural, and sequence similarities to a conserved TIR in the mammalian UV sensitivity syndrome A (UVSSA) protein, which recruits TFIIH during TC-NER in mammalian cells. These findings suggest that the Elf1 CTD acts as a functional counterpart to mammalian UVSSA in TC-NER by recruiting TFIIH in response to Pol II stalling at DNA lesions.


Asunto(s)
Reparación del ADN , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Factor de Transcripción TFIIH , Factor de Transcripción TFIIH/metabolismo , Factor de Transcripción TFIIH/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Unión Proteica , Transcripción Genética , Rayos Ultravioleta , Dominios Proteicos , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Daño del ADN , Dímeros de Pirimidina/metabolismo , Reparación por Escisión
2.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-39000160

RESUMEN

222 nm far-ultraviolet (F-UV) light has a bactericidal effect similar to deep-ultraviolet (D-UV) light of about a 260 nm wavelength. The cytotoxic effect of 222 nm F-UV has not been fully investigated. DLD-1 cells were cultured in a monolayer and irradiated with 222 nm F-UV or 254 nm D-UV. The cytotoxicity of the two different wavelengths of UV light was compared. Changes in cell morphology after F-UV irradiation were observed by time-lapse imaging. Differences in the staining images of DNA-binding agents Syto9 and propidium iodide (PI) and the amount of cyclobutane pyrimidine dimer (CPD) were examined after UV irradiation. F-UV was cytotoxic to the monolayer culture of DLD-1 cells in a radiant energy-dependent manner. When radiant energy was set to 30 mJ/cm2, F-UV and D-UV showed comparable cytotoxicity. DLD-1 cells began to expand immediately after 222 nm F-UV light irradiation, and many cells incorporated PI; in contrast, PI uptake was at a low level after D-UV irradiation. The amount of CPD, an indicator of DNA damage, was higher in cells irradiated with D-UV than in cells irradiated with F-UV. This study proved that D-UV induced apoptosis from DNA damage, whereas F-UV affected membrane integrity in monolayer cells.


Asunto(s)
Apoptosis , Membrana Celular , Neoplasias del Colon , Daño del ADN , Rayos Ultravioleta , Humanos , Línea Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/efectos de la radiación , Neoplasias del Colon/patología , Neoplasias del Colon/metabolismo , Apoptosis/efectos de la radiación , Dímeros de Pirimidina/metabolismo
3.
BMC Plant Biol ; 24(1): 723, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39080534

RESUMEN

BACKGROUND: 6 - 4 photoproducts are the second most common UV-induced DNA lesions after cyclobutane pyrimidine dimers. In plants, they are mainly repaired by photolyases in a process called photoreactivation. While pyrimidine dimers can be deleterious, leading to mutagenesis or even cell death, 6 - 4 photoproducts can activate specific signaling pathways. Therefore, their removal is particularly important, especially for plants exposed to high UV intensities due to their sessile nature. Although photoreactivation in nuclear DNA is well-known, its role in plant organelles remains unclear. In this paper we analyzed the activity and localization of GFP-tagged AtUVR3, the 6 - 4 photoproduct specific photolyase. RESULTS: Using transgenic Arabidopsis with different expression levels of AtUVR3, we confirmed a positive trend between these levels and the rate of 6 - 4 photoproduct removal under blue light. Measurements of 6 - 4 photoproduct levels in chloroplast and nuclear DNA of wild type, photolyase mutants, and transgenic plants overexpressing AtUVR3 showed that the photoreactivation is the main repair pathway responsible for the removal of these lesions in both organelles. The GFP-tagged AtUVR3 was predominantly located in nuclei with a small fraction present in chloroplasts and mitochondria of transgenic Arabidopsis thaliana and Nicotiana tabacum lines. In chloroplasts, this photolyase co-localized with the nucleoid marked by plastid envelope DNA binding protein. CONCLUSIONS: Photolyases are mainly localized in plant nuclei, with only a small fraction present in chloroplasts and mitochondria. Despite this unbalanced distribution, photoreactivation is the primary mechanism responsible for the removal of 6 - 4 photoproducts from nuclear and chloroplast DNA in adult leaves. The amount of the AtUVR3 photolyase is the limiting factor influencing the photoreactivation rate of 6 - 4 photoproducts. The efficient photoreactivation of 6 - 4 photoproducts in 35S: AtUVR3-GFP Arabidopsis and Nicotiana tabacum is a promising starting point to evaluate whether transgenic crops overproducing this photolyase are more tolerant to high UV irradiation and how they respond to other abiotic and biotic stresses under field conditions.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Núcleo Celular , Reparación del ADN , Desoxirribodipirimidina Fotoliasa , Plantas Modificadas Genéticamente , Arabidopsis/genética , Arabidopsis/efectos de la radiación , Arabidopsis/metabolismo , Proteínas de Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Núcleo Celular/metabolismo , Núcleo Celular/efectos de la radiación , Desoxirribodipirimidina Fotoliasa/metabolismo , Desoxirribodipirimidina Fotoliasa/genética , Rayos Ultravioleta , ADN de Plantas/metabolismo , ADN de Plantas/genética , Dímeros de Pirimidina/metabolismo , Dímeros de Pirimidina/genética , ADN de Cloroplastos/genética , ADN de Cloroplastos/metabolismo , Cloroplastos/metabolismo , Daño del ADN
4.
Mol Biol (Mosk) ; 58(1): 3-21, 2024.
Artículo en Ruso | MEDLINE | ID: mdl-38943577

RESUMEN

Photochemical reactions in cell DNA are induced in various organisms by solar UV radiation and may lead to a series of biological responses to DNA damage, including apoptosis, mutagenesis, and carcinogenesis. The chemical nature and the amount of DNA lesions depend on the wavelength of UV radiation. UV type B (UVB, 290-320 nm) causes two main lesions, cyclobutane pyrimidine dimers (CPDs) and, with a lower yield, pyrimidine (6-4) pyrimidone photoproducts (6-4PPs). Their formation is a result of direct UVB photon absorption by DNA bases. UV type A (UVA, 320-400 nm) induces only cyclobutane dimers, which most likely arise via triplet-triplet energy transfer (TTET) from cell chromophores to DNA thymine bases. UVA is much more effective than UVB in inducing sensitized oxidative DNA lesions, such as single-strand breaks and oxidized bases. Of the latter, 8-oxo-dihydroguanine (8-oxodG) is the most frequent, being produced in several oxidation processes. Many recent studies reported novel, more detailed information about the molecular mechanisms of the photochemical reactions that underlie the formation of various DNA lesions. The information is mostly summarized and analyzed in the review. Special attention is paid to the oxidation reactions that are initiated by reactive oxygen species (ROS) and radicals generated by potential endogenous photosensitizers, such as pterins, riboflavin, protoporphyrin IX, NADH, and melanin. The review discusses the role that specific DNA photoproducts play in genotoxic processes induced in living systems by UV radiation of various wavelengths, including human skin carcinogenesis.


Asunto(s)
Daño del ADN , Dímeros de Pirimidina , Rayos Ultravioleta , Rayos Ultravioleta/efectos adversos , Humanos , Daño del ADN/efectos de la radiación , Dímeros de Pirimidina/metabolismo , Dímeros de Pirimidina/genética , Dímeros de Pirimidina/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , ADN/efectos de la radiación , ADN/metabolismo , ADN/genética , Animales , Apoptosis/efectos de la radiación , Oxidación-Reducción/efectos de la radiación , 8-Hidroxi-2'-Desoxicoguanosina/metabolismo
5.
Nucleic Acids Res ; 52(14): 8254-8270, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-38884271

RESUMEN

The histone methyltransferase ASH1L, first discovered for its role in transcription, has been shown to accelerate the removal of ultraviolet (UV) light-induced cyclobutane pyrimidine dimers (CPDs) by nucleotide excision repair. Previous reports demonstrated that CPD excision is most efficient at transcriptional regulatory elements, including enhancers, relative to other genomic sites. Therefore, we analyzed DNA damage maps in ASH1L-proficient and ASH1L-deficient cells to understand how ASH1L controls enhancer stability. This comparison showed that ASH1L protects enhancer sequences against the induction of CPDs besides stimulating repair activity. ASH1L reduces CPD formation at C-containing but not at TT dinucleotides, and no protection occurs against pyrimidine-(6,4)-pyrimidone photoproducts or cisplatin crosslinks. The diminished CPD induction extends to gene promoters but excludes retrotransposons. This guardian role against CPDs in regulatory elements is associated with the presence of H3K4me3 and H3K27ac histone marks, which are known to interact with the PHD and BRD motifs of ASH1L, respectively. Molecular dynamics simulations identified a DNA-binding AT hook of ASH1L that alters the distance and dihedral angle between neighboring C nucleotides to disfavor dimerization. The loss of this protection results in a higher frequency of C->T transitions at enhancers of skin cancers carrying ASH1L mutations compared to ASH1L-intact counterparts.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN , Elementos de Facilitación Genéticos , N-Metiltransferasa de Histona-Lisina , Dímeros de Pirimidina , Humanos , Ratones , ADN/metabolismo , ADN/química , ADN/genética , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Histonas/genética , Simulación de Dinámica Molecular , Regiones Promotoras Genéticas , Dímeros de Pirimidina/metabolismo , Dímeros de Pirimidina/genética , Dímeros de Pirimidina/química , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Rayos Ultravioleta
6.
Nucleic Acids Res ; 52(13): 7437-7446, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-38908029

RESUMEN

Formamidopyrimidine (Fapy•dG) is a major lesion arising from oxidation of dG that is produced from a common chemical precursor of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-OxodGuo). In human cells, replication of single-stranded shuttle vectors containing Fapy•dG is more mutagenic than 8-OxodGuo. Here, we present the first data regarding promoter dependent RNA polymerase II bypass of Fapy•dG. 8-OxodGuo bypass was examined side-by-side. Experiments were carried out using double-stranded shuttle vectors in HeLa cell nuclear lysates and in HEK 293T cells. The lesions do not significantly block transcriptional bypass efficiency. Less than 2% adenosine incorporation occurred in cells when the lesions were base paired with dC. Inhibiting base excision repair in HEK 293T cells significantly increased adenosine incorporation, particularly from Fapy•dG:dC bypass which yielded ∼25% adenosine incorporation. No effect was detected upon transcriptional bypass of either lesion in nucleotide excision repair deficient cells. Transcriptional mutagenesis was significantly higher when shuttle vectors containing dA opposite one of the lesions were employed. For Fapy•dG:dA bypass, adenosine incorporation was greater than 85%; whereas 8-OxodGuo:dA yielded >20% point mutations. The combination of more frequent replication mistakes and greater error-prone Pol II bypass suggest that Fapy•dG is more mutagenic than 8-OxodGuo.


Asunto(s)
8-Hidroxi-2'-Desoxicoguanosina , Daño del ADN , Desoxiguanosina , Regiones Promotoras Genéticas , ARN Polimerasa II , Humanos , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Células HEK293 , 8-Hidroxi-2'-Desoxicoguanosina/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Células HeLa , Reparación del ADN , Transcripción Genética , Pirimidinas , Dímeros de Pirimidina/metabolismo , Dímeros de Pirimidina/genética
7.
Genetics ; 227(3)2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38722894

RESUMEN

UV light is a potent mutagen that induces bulky DNA damage in the form of cyclobutane pyrimidine dimers (CPDs). Photodamage and other bulky lesions occurring in nuclear genomes can be repaired through nucleotide excision repair (NER), where incisions on both sides of a damaged site precede the removal of a single-stranded oligonucleotide containing the damage. Mitochondrial genomes (mtDNAs) are also susceptible to damage from UV light, but current evidence suggests that the only way to eliminate bulky mtDNA damage is through mtDNA degradation. Damage-containing oligonucleotides excised during NER can be captured with antidamage antibodies and sequenced (XR-seq) to produce high-resolution maps of active repair locations following UV exposure. We analyzed previously published datasets from Arabidopsis thaliana, Saccharomyces cerevisiae, and Drosophila melanogaster to identify reads originating from the mtDNA (and plastid genome in A. thaliana). In A. thaliana and S. cerevisiae, the mtDNA-mapping reads have unique length distributions compared to the nuclear-mapping reads. The dominant fragment size was 26 nt in S. cerevisiae and 28 nt in A. thaliana with distinct secondary peaks occurring in regular intervals. These reads also show a nonrandom distribution of di-pyrimidines (the substrate for CPD formation) with TT enrichment at positions 7-8 of the reads. Therefore, UV damage to mtDNA appears to result in production of DNA fragments of characteristic lengths and positions relative to the damaged location. The mechanisms producing these fragments are unclear, but we hypothesize that they result from a previously uncharacterized DNA degradation pathway or repair mechanism in mitochondria.


Asunto(s)
Arabidopsis , Daño del ADN , Reparación del ADN , ADN Mitocondrial , Drosophila melanogaster , Saccharomyces cerevisiae , Rayos Ultravioleta , ADN Mitocondrial/genética , Arabidopsis/genética , Arabidopsis/efectos de la radiación , Rayos Ultravioleta/efectos adversos , Animales , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/efectos de la radiación , Saccharomyces cerevisiae/metabolismo , Drosophila melanogaster/genética , Dímeros de Pirimidina/genética , Dímeros de Pirimidina/metabolismo , Genoma Mitocondrial
8.
Exp Eye Res ; 243: 109901, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38641197

RESUMEN

Xeroderma pigmentosum (XP) is a rare genetic disorder characterized by injury to the ocular surface due to exposure to ultraviolet (UV) radiation. UV-induced damage in the cells leads to the formation of cyclobutane pyrimidine dimers (CPDs) and 6-4 pyrimidine-pyrimidone photoproducts that are repaired by the NER (Nucleotide Excision Repair) pathway. Mutations in the genes coding for NER proteins, as reported in XP patients, would lead to sub-optimal damage repair resulting in clinical signs varying from photo-keratitis to cancerous lesions on the ocular surface. Here, we aimed to provide evidence for the accumulation of DNA damage and activation of DNA repair pathway proteins in the corneal cells of patients with XP. Corneal buttons of patients who underwent penetrating keratoplasty were stained to quantify DNA damage and the presence of activated DNA damage response proteins (DDR) using specific antibodies. Positive staining for pH2A.X and thymidine dimers confirmed the presence of DNA damage in the corneal cells. Positive cells were found in both control corneas and XP samples however, unlike normal tissues, positive cells were found in all cell layers of XP samples indicating that these cells were sensitive to very low levels of UV. pH2A.X-positive cells were significantly more in XP corneas (p < 0.05) indicating the presence of double strand breaks in these tissues. A positive expression of phosphorylated-forms of DDR proteins was noted in XP corneas (unlike controls) such as ataxia telangiectasia mutated/Rad-3 related proteins (ATM/ATR), breast cancer-1 and checkpoint kinases-1 and -2. Nuclear localization of XPA was noted in XP samples which co-localized (calculated using Pearson's correlation) with pATM (0.9 ± 0.007) and pATR (0.6 ± 0.053). The increased presence of these in the nucleus confirms that unresolved DNA damage was accumulating in these cells thereby leading to prolonged activation of the damage response proteins. An increase in pp53 and TUNEL positive cells in the XP corneas indicated cell death likely driven by the p53 pathway. For comparison, cultured normal corneal epithelial cells were exposed to UV-radiation and stained for DDR proteins at 3, 6 and 24 h after irradiation to quantify the time taken by cells with intact DDR pathway to repair damage. These cells, when exposed to UV showed nuclear translocation of DDR proteins at 3 and 6 h which reduced significantly by 24 h confirming that the damaged DNA was being actively repaired leading to cell survival. The persistent presence of the DDR proteins in XP corneas indicates that damage is being actively recognized and DNA replication is stalled, thereby causing accumulation of damaged DNA leading to cell death, which would explain the cancer incidence and cell loss reported in these patients.


Asunto(s)
Daño del ADN , Reparación del ADN , Dímeros de Pirimidina , Rayos Ultravioleta , Xerodermia Pigmentosa , Humanos , Rayos Ultravioleta/efectos adversos , Xerodermia Pigmentosa/metabolismo , Xerodermia Pigmentosa/genética , Xerodermia Pigmentosa/patología , Dímeros de Pirimidina/metabolismo , Queratoplastia Penetrante , Córnea/metabolismo , Córnea/patología , Córnea/efectos de la radiación , Femenino , Adulto , Histonas/metabolismo , Masculino , Persona de Mediana Edad , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/genética , Adolescente , Adulto Joven
9.
Biotechnol Lett ; 46(3): 459-467, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38523200

RESUMEN

Solar ultraviolet radiations induced DNA damages in human skin cells with cyclobutane pyrimidine dimers (CPD) and (6-4) photoproducts (6-4PPs) as the most frequent lesions. CPDs are repaired much slower than 6-4PPs by the nucleotide excision repair pathway, which are thus the major lesions that interfere with key cellular processes and give rise to gene mutations, possibly resulting in skin cancer. In prokaryotes and multicellular eukaryotes other than placental mammals, CPDs can be rapidly repaired by CPD photolyases in one simple enzymatic reaction using the energy of blue light. In this study, we aim to construct recombinant CPD photolyases that can autonomously enter human cell nuclei to fix UV-induced CPDs. A fly cell penetration peptide and a viral nucleus localization signal peptide were recombined with a fungal CPD photolyase to construct a recombinant protein. This engineered CPD photolyase autonomously crosses cytoplasm and nuclear membrane of human cell nuclei, which then efficiently photo-repairs UV-induced CPD lesions in the genomic DNA. This further protects the cells by increasing SOD activity, and decreasing cellular ROSs, malondialdehyde and apoptosis.


Asunto(s)
Núcleo Celular , Daño del ADN , Reparación del ADN , Desoxirribodipirimidina Fotoliasa , Dímeros de Pirimidina , Proteínas Recombinantes , Rayos Ultravioleta , Humanos , Desoxirribodipirimidina Fotoliasa/metabolismo , Desoxirribodipirimidina Fotoliasa/genética , Núcleo Celular/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/genética , Dímeros de Pirimidina/metabolismo , Dímeros de Pirimidina/genética , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética
10.
J Biochem ; 176(1): 35-42, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38426948

RESUMEN

The T7 gene 3 product, T7 endonuclease I, acts on various substrates with DNA structures, including Holliday junctions, heteroduplex DNAs and single-mismatch DNAs. Genetic analyses have suggested the occurrence of DNA recombination, replication and repair in Escherichia coli. In this study, T7 endonuclease I digested UV-irradiated covalently closed circular plasmid DNA into linear and nicked plasmid DNA, suggesting that the enzyme generates single- and double-strand breaks (SSB and DSB). To further investigate the biochemical functions of T7 endonuclease I, we have analysed endonuclease activity in UV-induced DNA substrates containing a single lesion, cyclobutane pyrimidine dimers (CPD) and 6-4 photoproducts (6-4PP). Interestingly, the leading cleavage site for CPD by T7 endonuclease I is at the second and fifth phosphodiester bonds that are 5' to the lesion of CPD on the lesion strand. However, in the case of 6-4PP, the cleavage pattern on the lesion strand resembled that of CPD, and T7 endonuclease I could also cleave the second phosphodiester bond that is 5' to the adenine-adenine residues opposite the lesion, indicating that the enzyme produces DSB in DNA containing 6-4PP. These findings suggest that T7endonuclease I accomplished successful UV damage repair by SSB in CPD and DSB in 6-4PP.


Asunto(s)
Daño del ADN , Desoxirribonucleasa I , Rayos Ultravioleta , Rayos Ultravioleta/efectos adversos , Desoxirribonucleasa I/metabolismo , Desoxirribonucleasa I/química , ADN/metabolismo , ADN/química , Escherichia coli/genética , Escherichia coli/metabolismo , Bacteriófago T7/enzimología , Bacteriófago T7/genética , Dímeros de Pirimidina/metabolismo , Dímeros de Pirimidina/química , Reparación del ADN
11.
Plant Physiol ; 195(1): 326-342, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38345835

RESUMEN

Photoreactivation enzyme that repairs cyclobutane pyrimidine dimer (CPD) induced by ultraviolet-B radiation, commonly called CPD photolyase (PHR) is essential for plants living under sunlight. Rice (Oryza sativa) PHR (OsPHR) is a unique triple-targeting protein. The signal sequences required for its translocation to the nucleus or mitochondria are located in the C-terminal region but have yet to be identified for chloroplasts. Here, we identified sequences located in the N-terminal region, including the serine-phosphorylation site at position 7 of OsPHR, and found that OsPHR is transported/localized to chloroplasts via a vesicle transport system under the control of serine-phosphorylation. However, the sequence identified in this study is only conserved in some Poaceae species, and in many other plants, PHR is not localized to the chloroplasts. Therefore, we reasoned that Poaceae species need the ability to repair CPD in the chloroplast genome to survive under sunlight and have uniquely acquired this mechanism for PHR chloroplast translocation.


Asunto(s)
Cloroplastos , Desoxirribodipirimidina Fotoliasa , Oryza , Rayos Ultravioleta , Cloroplastos/metabolismo , Desoxirribodipirimidina Fotoliasa/metabolismo , Desoxirribodipirimidina Fotoliasa/genética , Oryza/genética , Oryza/enzimología , Oryza/efectos de la radiación , Oryza/metabolismo , Proteínas de Plantas/metabolismo , Proteínas de Plantas/genética , Dímeros de Pirimidina/metabolismo , Poaceae/genética , Poaceae/enzimología , Poaceae/efectos de la radiación , Poaceae/metabolismo , Secuencia de Aminoácidos , Transporte de Proteínas
12.
Chem Biol Interact ; 388: 110837, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38104746

RESUMEN

Cyclobutane pyrimidine dimer (CPD) and (6-4)photoproduct (6-4 PP) are two major types of UV-induced DNA lesion and 6-4 PP is more mutagenic than CPD. Activated by lesion detection, nucleotide excision repair (NER) eliminates CPDs and 6-4 PPs. Thallium (Tl) is a toxic metal existing primarily as Tl+ in the aquatic environment. Ingestion of Tl+-contaminated foods and water is a major route of human poisoning. As Tl+ may inhibit enzyme activities via binding to sulfhydryl groups, this study explored if Tl+ could intensify UV mutagenicity by inactivating NER-linked damage recognition factors using zebrafish (Danio rerio) embryo as a model system. Incubation of Tl+ (as thallium nitrate) at 0.1-0.4 µg/mL with zebrafish extracts for 20 min caused a concentration-dependent inhibition of 6-4 PP binding activities as shown by a photolesion-specific band shift assay, while CPD binding activities were insensitive to Tl+. The ability of Tl+ to suppress 6-4 PP detection was stronger than that of Hg2+. Exposure of zebrafish embryos at 1 h post fertilization (hpf) to Tl+ at 0.4-1 µg/mL for 9 or 71 h also specifically inhibited 6-4 PP detection, indicating that Tl+ induced a prolonged inhibition of 6-4 PP sensing ability primarily via its direct interaction with damage recognition molecules. Tl+-mediated inhibition of 6-4 PP binding in embryos at distinct stages resulted in a suppression of NER capacity monitored by a transcription-based DNA repair assay. Our results revealed the potential of Tl+ to enhance UV mutagenicity by disturbing the removal of 6-4 PP through repressing the lesion detection step of NER.


Asunto(s)
Reparación por Escisión , Pez Cebra , Animales , Humanos , Pez Cebra/metabolismo , Talio/toxicidad , Talio/metabolismo , Reparación del ADN , Daño del ADN , Dímeros de Pirimidina/metabolismo , Rayos Ultravioleta
13.
Physiol Plant ; 175(5): e14049, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37882276

RESUMEN

The single-stranded DNA/RNA binding protein WHIRLY1 is a major chloroplast nucleoid-associated protein required for the compactness of nucleoids. Most nucleoids in chloroplasts of WHIRLY1-knockdown barley plants are less compact compared to nucleoids in wild-type plants. The reduced compaction leads to an enhanced optical cross-section, which may cause the plastid DNA to be a better target for damaging UV-B radiation. To investigate this hypothesis, primary foliage leaves, chloroplasts, and nuclei from wild-type and WHIRLY1-knockdown plants were exposed to experimental UV-B radiation. Thereafter, total, genomic and plastid DNA were isolated, respectively, and analyzed for the occurrence of cyclobutane pyrimidine dimers (CPDs), which is a parameter for genome stability. The results of this study revealed that WHIRLY1-deficient chloroplasts had strongly enhanced DNA damages, whereas isolated nuclei from the same plant line were not more sensitive than nuclei from the wild-type, indicating that WHIRLY1 has different functions in chloroplasts and nucleus. This supports the hypothesis that the compaction of nucleoids may provide protection against UV-B radiation.


Asunto(s)
Proteínas de Plantas , Dímeros de Pirimidina , Dímeros de Pirimidina/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Cloroplastos/metabolismo , Rayos Ultravioleta , ADN/metabolismo
14.
Mol Cell ; 83(20): 3669-3678.e7, 2023 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-37816354

RESUMEN

UV irradiation induces "bulky" DNA photodimers such as (6-4)-photoproducts and cyclobutane pyrimidine dimers that are removed by nucleotide excision repair, a complex process defective in the sunlight-sensitive and cancer-prone disease xeroderma pigmentosum. Some bacteria and lower eukaryotes can also repair photodimers by enzymatically simpler mechanisms, but such pathways have not been reported in normal human cells. Here, we have identified such a mechanism. We show that normal human cells can employ a DNA base excision repair process involving NTH1, APE1, PARP1, XRCC1, and FEN1 to rapidly remove a subset of photodimers at early times following UVC irradiation. Loss of these proteins slows the early rate of repair of photodimers in normal cells, ablates their residual repair in xeroderma pigmentosum cells, and increases UVC sensitivity ∼2-fold. These data reveal that human cells can excise photodimers using a long-patch base excision repair process that functions additively but independently of nucleotide excision repair.


Asunto(s)
Xerodermia Pigmentosa , Humanos , Xerodermia Pigmentosa/genética , Reparación del ADN/genética , Dímeros de Pirimidina/genética , Dímeros de Pirimidina/metabolismo , Daño del ADN/genética , ADN/genética , Rayos Ultravioleta , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/metabolismo
15.
J Photochem Photobiol B ; 245: 112757, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37481791

RESUMEN

The application of a far-ultraviolet C (UVC) light emitting diode (LED) of 233 nm showed significant bactericidal efficacy at an applied dose between 20 and 80 mJ cm-2 as reported recently. In addition, only minor epidermal DNA lesions were observed in ex vivo human skin and in vitro epidermal models <10% of the minimal erythema dose of UVB radiation. To broaden the potential range of applications of such systems, e.g. to include postoperative application on wounds for the purpose of decontamination, we assessed how a disruption of normal anatomic skin structure and function influences the skin damage induced by light from 233 nm far-UVC LEDs. Thus, we induced superficial skin wounds by mechanical detachment of the stratum corneum in ex vivo human skin. Barrier-disruption of the skin could be successfully determined by measuring an increase in the transepidermal water loss (TEWL) and the stratum corneum loss could be determined morphologically by 2-photon microscopy (2-PM). After far-UVC irradiation of the skin, we screened the tissue for the development of cyclobutane pyrimidine dimers (CPDs) and 6-4 photoproducts (6-4PPs). The abundance of DNA lesions was elevated in wound skin in comparison to intact skin after irradiation with far-UVC. However, no increase in DNA lesions was detected when artificial wound exudate consisting of cell culture medium and serum was applied to the disrupted skin surface prior to irradiation. This effect agrees with the results of ray tracing simulations of the absorption of far-UVC light incident on a superficial skin wound. Interestingly, no significant deviations in radical formation between intact skin and superficially wounded skin were detected after far-UVC irradiation as analyzed by electron paramagnetic resonance (EPR) spectroscopy. In conclusion, 233 nm LED light at a dose of 60 mJ/cm2 could be applied safely on superficial wounds for the purpose of skin antisepsis as long as the wounds are covered with wound fluid.


Asunto(s)
Dímeros de Pirimidina , Piel , Humanos , Piel/efectos de la radiación , Dímeros de Pirimidina/metabolismo , Epidermis , ADN/metabolismo , Rayos Ultravioleta , Daño del ADN
16.
Nat Commun ; 14(1): 2701, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37169761

RESUMEN

Decades ago, it was shown that proteins binding to DNA can quantitatively alter the formation of DNA damage by UV light. This established the principle of UV footprinting for non-intrusive study of protein-DNA contacts in living cells, albeit at limited scale and precision. Here, we perform deep base-resolution quantification of the principal UV damage lesion, the cyclobutane pyrimidine dimer (CPD), at select human promoter regions using targeted CPD sequencing. Several transcription factors exhibited distinctive and repeatable damage signatures indicative of site occupancy, involving strong (up to 17-fold) position-specific elevations and reductions in CPD formation frequency relative to naked DNA. Positive damage modulation at some ETS transcription factor binding sites coincided at base level with melanoma somatic mutation hotspots. Our work provides proof of concept for the study of protein-DNA interactions at individual loci using light and sequencing, and reveals widespread and potent modulation of UV damage in regulatory regions.


Asunto(s)
Proteínas de Unión al ADN , Rayos Ultravioleta , Humanos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Daño del ADN , Dímeros de Pirimidina/metabolismo , ADN/metabolismo
17.
J Photochem Photobiol B ; 243: 112713, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37086566

RESUMEN

Ultraviolet C (UVC) light has long been used as a sterilizing agent, primarily through devices that emit at 254 nm. Depending on the dose and duration of exposure, UV 254 nm can cause erythema and photokeratitis and potentially cause skin cancer since it directly modifies nitrogenated nucleic acid bases. Filtered KrCl excimer lamps (emitting mainly at 222 nm) have emerged as safer germicidal tools and have even been proposed as devices to sterilize surgical wounds. All the studies that showed the safety of 222 nm analyzed cell number and viability, erythema generation, epidermal thickening, the formation of genetic lesions such as cyclobutane pyrimidine dimers (CPDs) and pyrimidine-(6-4)-pyrimidone photoproducts (6-4PPs) and cancer-inducing potential. Although nucleic acids can absorb and be modified by both UV 254 nm and UV 222 nm equally, compared to UV 254 nm, UV 222 nm is more intensely absorbed by proteins (especially aromatic side chains), causing photooxidation and cross-linking. Here, in addition to analyzing DNA lesion formation, for the first time, we evaluated changes in the proteome and cellular pathways, reactive oxygen species formation, and metalloproteinase (MMP) levels and activity in full-thickness in vitro reconstructed human skin (RHS) exposed to UV 222 nm. We also performed the longest (40 days) in vivo study of UV 222 nm exposure in the HRS/J mouse model at the occupational threshold limit value (TLV) for indirect exposure (25 mJ/cm2) and evaluated overall skin morphology, cellular pathological alterations, CPD and 6-4PP formation and MMP-9 activity. Our study showed that processes related to reactive oxygen species and inflammatory responses were more altered by UV 254 nm than by UV 222 nm. Our chronic in vivo exposure assay using the TLV confirmed that UV 222 nm causes minor damage to the skin. However, alterations in pathways related to skin regeneration raise concerns about direct exposure to UV 222 nm.


Asunto(s)
Daño del ADN , Ácidos Nucleicos , Ratones , Animales , Humanos , Especies Reactivas de Oxígeno/metabolismo , Dímeros de Pirimidina/metabolismo , Piel/efectos de la radiación , Rayos Ultravioleta , Ácidos Nucleicos/metabolismo , Eritema
18.
PLoS One ; 18(4): e0283572, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37027425

RESUMEN

Absorption of ultraviolet radiation (UVR) by DNA leads to the predominant formation of cyclobutane pyrimidine dimers (CPD). Since those CPD are responsible for the driver mutations found in skin cancers, their efficient repair is critical. We previously showed that pre-stimulation of fibroblasts with chronic low doses of UVB (CLUV) increases CPD repair efficiency. Since skin cancers are not arising from dermal fibroblasts, this observation is not directly relevant to cutaneous carcinogenesis. We have now exposed HaCaT keratinocytes to a CLUV irradiation protocol to determine whether this pre-stimulation influences CPD removal rate. Similar to fibroblasts, CLUV treatment leads to the accumulation of residual CPD in keratinocytes, which are not repaired but rather tolerated and diluted through DNA replication. In contrast to fibroblasts, in keratinocytes we find that CLUV pre-treatment reduces CPD removal of newly generated damage without inducing a higher sensitivity to UVR-induced cell death. Using our experimental data, we derived a theoretical model to predict CPD induction, dilution and repair that occur in keratinocytes when chronically UVB-irradiated. Altogether, these results suggest that the accumulation of unrepaired CPD and the reduction in repair efficiency caused by chronic UVB exposure might lead to an increase in skin cancer driver mutations.


Asunto(s)
Neoplasias Cutáneas , Rayos Ultravioleta , Humanos , Rayos Ultravioleta/efectos adversos , Daño del ADN , Células HaCaT/metabolismo , Reparación del ADN/genética , Dímeros de Pirimidina/metabolismo , Queratinocitos/metabolismo , Neoplasias Cutáneas/genética
19.
J Biol Chem ; 299(5): 104679, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37028766

RESUMEN

The 3D organization of the eukaryotic genome is crucial for various cellular processes such as gene expression and epigenetic regulation, as well as for maintaining genome integrity. However, the interplay between UV-induced DNA damage and repair with the 3D structure of the genome is not well understood. Here, we used state-of-the-art Hi-C, Damage-seq, and XR-seq datasets and in silico simulations to investigate the synergistic effects of UV damage and 3D genome organization. Our findings demonstrate that the peripheral 3D organization of the genome shields the central regions of genomic DNA from UV-induced damage. Additionally, we observed that potential damage sites of pyrimidine-pyrimidone (6-4) photoproducts are more prevalent in the nucleus center, possibly indicating an evolutionary pressure against those sites at the periphery. Interestingly, we found no correlation between repair efficiency and 3D structure after 12 min of irradiation, suggesting that UV radiation alters the genome's 3D organization in a short period of time. Interestingly, however, 2 h after UV induction, we observed more efficient repair levels in the center of the nucleus relative to the periphery. These results have implications for understanding the etiology of cancer and other diseases, as the interplay between UV radiation and the 3D genome may play a role in the development of genetic mutations and genomic instability.


Asunto(s)
Daño del ADN , Reparación del ADN , Epigénesis Genética , Dímeros de Pirimidina/metabolismo , Rayos Ultravioleta/efectos adversos
20.
Int J Mol Sci ; 24(5)2023 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-36902353

RESUMEN

The calcium-sensing receptor (CaSR) is an important regulator of epidermal function. We previously reported that knockdown of the CaSR or treatment with its negative allosteric modulator, NPS-2143, significantly reduced UV-induced DNA damage, a key factor in skin cancer development. We subsequently wanted to test whether topical NPS-2143 could also reduce UV-DNA damage, immune suppression, or skin tumour development in mice. In this study, topical application of NPS-2143 (228 or 2280 pmol/cm2) to Skh:hr1 female mice reduced UV-induced cyclobutane pyrimidine dimers (CPD) (p < 0.05) and oxidative DNA damage (8-OHdG) (p < 0.05) to a similar extent as the known photoprotective agent 1,25(OH)2 vitamin D3 (calcitriol, 1,25D). Topical NPS-2143 failed to rescue UV-induced immunosuppression in a contact hypersensitivity study. In a chronic UV photocarcinogenesis protocol, topical NPS-2143 reduced squamous cell carcinomas for only up to 24 weeks (p < 0.02) but had no other effect on skin tumour development. In human keratinocytes, 1,25D, which protected mice from UV-induced skin tumours, significantly reduced UV-upregulated p-CREB expression (p < 0.01), a potential early anti-tumour marker, while NPS-2143 had no effect. This result, together with the failure to reduce UV-induced immunosuppression, may explain why the reduction in UV-DNA damage in mice with NPS-2143 was not sufficient to inhibit skin tumour formation.


Asunto(s)
Receptores Sensibles al Calcio , Neoplasias Cutáneas , Femenino , Animales , Ratones , Humanos , Ratones Pelados , Receptores Sensibles al Calcio/metabolismo , Rayos Ultravioleta , Daño del ADN , Neoplasias Cutáneas/metabolismo , Dímeros de Pirimidina/metabolismo , Piel/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA