Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 377
1.
Chem Commun (Camb) ; 59(91): 13603-13606, 2023 Nov 14.
Article En | MEDLINE | ID: mdl-37899697

Nucleic acids can be damaged by ultraviolet (UV) irradiation, forming structural photolesions such as cyclobutane-pyrimidine-dimers (CPD). In modern organisms, sophisticated enzymes repair CPD lesions in DNA, but to our knowledge, no RNA-specific enzymes exist for CPD repair. Here, we show for the first time that RNA can protect itself from photolesions by an intrinsic UV-induced self-repair mechanism. This mechanism, prior to this study, has exclusively been observed in DNA and is based on charge transfer from CPD-adjacent bases. In a comparative study, we determined the quantum yields of the self-repair of the CPD-containing RNA sequence, GAU = U to GAUU (0.23%), and DNA sequence, d(GAT = T) to d(GATT) (0.44%), upon 285 nm irradiation via UV/Vis spectroscopy and HPLC analysis. After several hours of irradiation, a maximum conversion yield of ∼16% for GAU = U and ∼33% for d(GAT = T) was reached. We examined the dynamics of the intermediate charge transfer (CT) state responsible for the self-repair with ultrafast UV pump - IR probe spectroscopy. In the dinucleotides GA and d(GA), we found comparable quantum yields of the CT state of ∼50% and lifetimes on the order of several hundred picoseconds. Charge transfer in RNA strands might lead to reactions currently not considered in RNA photochemistry and may help understanding RNA damage formation and repair in modern organisms and viruses. On the UV-rich surface of the early Earth, these self-stabilizing mechanisms likely affected the selection of the earliest nucleotide sequences from which the first organisms may have developed.


DNA Repair , Pyrimidine Dimers , Pyrimidine Dimers/chemistry , Pyrimidine Dimers/genetics , Pyrimidine Dimers/radiation effects , RNA , DNA/chemistry , Ultraviolet Rays , DNA Damage
2.
Biochemistry ; 62(15): 2269-2279, 2023 08 01.
Article En | MEDLINE | ID: mdl-37459251

G-quadruplexes are thought to play an important role in gene regulation and telomere maintenance, but developing probes for their presence and location is challenging due to their transitory and highly dynamic nature. The majority of probes for G-quadruplexes have relied on antibody or small-molecule binding agents, many of which can also alter the dynamics and relative populations of G-quadruplexes. Recently, it was discovered that ultraviolet B (UVB) irradiation of human telomeric DNA and various G-quadruplex forming sequences found in human promoters, as well as reverse Hoogsteen hairpins, produces a unique class of non-adjacent anti cyclobutane pyrimidine dimers (CPDs). Therefore, one can envision using a pulse of UVB light to irreversibly trap these non-B DNA structures via anti CPD formation without perturbing their dynamics, after which the anti CPDs can be identified and mapped. As a first step toward this goal, we report radioactive post- and pre-labeling assays for the detection of non-adjacent CPDs and illustrate their use in detecting trans,anti T=(T) CPD formation in a human telomeric DNA sequence. Both assays make use of snake venom phosphodiesterase (SVP) to degrade the trans,anti T=(T) CPD-containing DNA to the tetranucleotide pTT=(pTT) corresponding to CPD formation between the underlined T's of two separate dinucleotides while degrading the adjacent syn TT CPDs to the trinucleotide pGT=T. In the post-labeling assay, calf intestinal phosphodiesterase is used to dephosphorylate the tetranucleotides, which are then rephosphorylated with kinase and [32P]-ATP to produce radiolabeled mono- and diphosphorylated tetranucleotides. The tetranucleotides are confirmed to be non-adjacent CPDs by 254 nm photoreversion to the dinucleotide p*TT. In the pre-labeling assay, radiolabeled phosphates are introduced into non-adjacent CPD-forming sites by ligation prior to irradiation, thereby eliminating the dephosphorylation and rephosphorylation steps. The assays are also demonstrated to detect the stereoisomeric cis,anti T=(T) CPD.


G-Quadruplexes , Humans , DNA/chemistry , Pyrimidine Dimers/chemistry , Pyrimidine Dimers/radiation effects , Ultraviolet Rays , DNA Damage
3.
Nucleic Acids Res ; 51(11): 5341-5350, 2023 06 23.
Article En | MEDLINE | ID: mdl-37207339

Photochemical dimerization of adjacent pyrimidines is fundamental to the creation of mutagenic hotspots caused by ultraviolet light. Distribution of the resulting lesions (cyclobutane pyrimidine dimers, CPDs) is already known to be highly variable in cells, and in vitro models have implicated DNA conformation as a major basis for this observation. Past efforts have primarily focused on mechanisms that influence CPD formation and have rarely considered contributions of CPD reversion. However, reversion is competitive under the standard conditions of 254 nm irradiation as illustrated in this report based on the dynamic response of CPDs to changes in DNA conformation. A periodic profile of CPDs was recreated in DNA held in a bent conformation by λ repressor. After linearization of this DNA, the CPD profile relaxed to its characteristic uniform distribution over a similar time of irradiation to that required to generate the initial profile. Similarly, when a T tract was released from a bent conformation, its CPD profile converted under further irradiation to that consistent with a linear T tract. This interconversion of CPDs indicates that both its formation and reversion exert control on CPD populations long before photo-steady-state conditions are achieved and suggests that the dominant sites of CPDs will evolve as DNA conformation changes in response to natural cellular processes.


DNA Damage , Pyrimidine Dimers , Pyrimidine Dimers/radiation effects , DNA/genetics , DNA Repair , Ultraviolet Rays , Nucleic Acid Conformation
4.
PLoS One ; 18(1): e0281162, 2023.
Article En | MEDLINE | ID: mdl-36719882

Far-ultraviolet radiation C light (far-UVC; 222 nm wavelength) has received attention as a safer light for killing pathogenic bacteria and viruses, as no or little DNA damage is observed after irradiation in mammalian skin models. Far-UVC does not penetrate deeply into tissues; therefore, it cannot reach the underlying critical basal cells. However, it was unclear whether far-UVC (222-UVC) irradiation could cause more biological damage at shallower depths than the 254 nm UVC irradiation (254-UVC), which penetrates more deeply. This study investigated the biological effects of 222- and 254-UVC on the small and transparent model organism Caenorhabditis elegans. At the same energy level of irradiation, 222-UVC introduced slightly less cyclobutane pyrimidine dimer damage to naked DNA in solution than 254-UVC. The survival of eggs laid during 0-4 h after irradiation showed a marked decrease with 254-UVC but not 222-UVC. In addition, defect of chromosomal condensation was observed in a full-grown oocyte by 254-UVC irradiation. In contrast, 222-UVC had a significant effect on the loss of motility of C. elegans. The sensory nervous system, which includes dopamine CEP and PVD neurons on the body surface, was severely damaged by 222-UVC, but not by the same dose of 254-UVC. Interestingly, increasing 254-UVC irradiation by about 10-fold causes similar damage to CEP neurons. These results suggest that 222-UVC is less penetrating, so energy transfer occurs more effectively in tissues near the surface, causing more severe damage than 254-UVC.


Caenorhabditis elegans , Peripheral Nervous System Diseases , Animals , Caenorhabditis elegans/genetics , Ultraviolet Rays , DNA Damage , Pyrimidine Dimers/radiation effects , Skin/microbiology , Mammals
5.
Anticancer Res ; 42(10): 5069-5076, 2022 Oct.
Article En | MEDLINE | ID: mdl-36191964

BACKGROUND/AIM: Solar ultraviolet radiation (UVR) is a carcinogen and irradiation of the skin results in DNA damage. Cyclobutane pyrimidine dimers (CPDs), including thymidine dimers, are among the most frequent forms of DNA damage. When CPDs are formed, the nucleotide excision repair system is activated and CPDs are excreted in the urine. Here, we developed a mass spectrometry-based method to quantify thymidine dimers in the urine and tested the method on a small group of volunteers after whole-body UVR exposure. PATIENTS AND METHODS: Years of research resulted in a method based on the "dilute-and-shoot" principle and ultra-performance liquid chromatography (UPLC) coupled to mass spectrometry. The whole body of each of eight healthy volunteers was exposed to 1.5-2.0 standard erythema doses (SEDs) of UVR for 3 consecutive days. Morning urine was collected on Day 1 (before irradiation) and on the following 7-9 days. Prior to analysis, sample preparation consisted of a simple dilution. A tandem quadrupole mass spectrometer coupled to UPLC was used for quantitative analysis in the multiple reaction monitoring mode. RESULTS: After 3 consecutive days of 1.5-2 SEDs, the highest level of thymidine dimer excretion occurred on Day 6 (0.7 ng/ml urine). Compared with baseline, significantly more thymidine dimers were excreted every day until Day 8 (p<0.016). Our method quantifies thymidine dimers that are excreted as dimers (i.e., not degraded further) after nucleotide excision repair. CONCLUSION: This is the first published mass spectrometry-based method for quantifying thymidine dimers in the urine after whole-body UVR exposure.


Pyrimidine Dimers , Ultraviolet Rays , Carcinogens , Chromatography, High Pressure Liquid , Chromatography, Liquid , DNA Damage , DNA Repair , Humans , Pyrimidine Dimers/radiation effects , Pyrimidine Dimers/urine , Tandem Mass Spectrometry , Thymidine , Ultraviolet Rays/adverse effects , Volunteers
6.
J Invest Dermatol ; 142(11): 3062-3070.e3, 2022 11.
Article En | MEDLINE | ID: mdl-35691362

Solar radiation induces the formation of cyclobutane pyrimidine dimers (CPDs) and other UV photoproducts in the genomic DNA of epidermal keratinocytes. Although CPDs have been detected in urine from UV- and sun-exposed individuals, the pathway by which they arrive there and the mechanisms by which UV-induced DNA damage in the skin has systemic effects throughout the body are not clear. Consistent with previous reports that DNA associates with small extracellular vesicles that are released from a variety of cell types, we observed that a small fraction of CPDs formed in genomic DNA after UVB exposure can later be detected in the culture medium. These extracellular CPDs are found within large fragments of histone-associated DNA and are released in a time- and UVB dose‒dependent manner. Moreover, studies with both cultured cells and human skin explants revealed that CPD release into the extracellular environment is blocked by caspase inhibition, which indicates a role for apoptotic signaling in CPD release from UVB-irradiated keratinocytes. Finally, we show that this released CPD-containing DNA can be taken up by other keratinocytes. These results therefore provide possible mechanisms for the export of damaged DNA from UVB-irradiated cells and for systemic effects of UVB exposure throughout the body.


Caspases , Pyrimidine Dimers , Humans , Pyrimidine Dimers/radiation effects , Caspases/metabolism , Histones/metabolism , Ultraviolet Rays/adverse effects , Keratinocytes/metabolism , DNA Damage , DNA/metabolism
7.
Photochem Photobiol ; 98(6): 1365-1371, 2022 11.
Article En | MEDLINE | ID: mdl-35313036

For the prevention of surgical site infection (SSI), continuous disinfection could be helpful. Short wavelength ultraviolet radiation C (UVC) is highly bactericidal but shows cytotoxicity. Radiation of UVC with a wavelength of 222 nm to the skin is considered to be safe because it only reaches the stratum corneum. However, the safety of 222 nm irradiation to the surgical field not covered with skin is unknown. The purpose of this study was to examine the safety of 222 nm UVC irradiation on a surgical field in a rabbit model. Five types of tissue were surgically exposed and irradiated with 222 or 254 nm UVC. Immunohistological assessment against cyclobutane pyrimidine dimer (CPD), an index of DNA damage by UVC, was performed. The CPD-positive cell rate was significantly higher in the 254 nm group than in the other groups in all tissues. A 222 nm group showed significantly more CPD than control in fat tissue, but no significant difference in all other tissues. In fat tissue collected 24 h after irradiation, the 254 nm group showed higher CPD than the other groups, while the 222 nm group had reduced to the control level. These data suggest that 222 nm UVC irradiation could be a new method to safely prevent SSI.


Pyrimidine Dimers , Ultraviolet Rays , Animals , Rabbits , Pyrimidine Dimers/radiation effects , DNA Damage , Skin/radiation effects , Epidermis/radiation effects
8.
Photochem Photobiol ; 98(3): 609-616, 2022 05.
Article En | MEDLINE | ID: mdl-34706095

Some early reports demonstrate that levels of cyclobutane pyrimidine dimers (CPD) may increase after UVR exposure had ended, although these observations were treated as artifacts. More recently, it has been shown unequivocally that CPD formation does occur post-irradiation, with maximal levels occurring after about 2-3 h. These lesions have been termed "dark CPD" (dCPD). Subsequent studies have confirmed their presence in vitro, in mouse models and in human skin in vivo. Melanin carbonyls have a role in the formation of dCPD, but they have also been observed in amelanotic systems, indicating other, unknown process(es) exist. In both cases, the formation of dCPD can be prevented by the presence of certain antioxidants. We lack data on the spectral dependence of dCPD, but it is unlikely to be the same as for incident CPD (iCPD), which are formed only during irradiation. There is evidence that iCPD and dCPD may have different repair kinetics, although this remains to be elucidated. It is also unknown whether iCPD and dCPD have different biological properties. The formation of dCPD in human skin in vivo has implications for post solar exposure photoprotection, and skin carcinogenesis, with a need for this to be investigated further.


DNA Damage , Pyrimidine Dimers , Animals , DNA Repair , Melanins , Mice , Polymers , Pyrimidine Dimers/radiation effects , Skin/radiation effects , Ultraviolet Rays
9.
Curr Probl Dermatol ; 55: 53-61, 2021.
Article En | MEDLINE | ID: mdl-34698038

Sunlight comprises radiation of different wavelengths, of which UVA and UVB are most important with respect to human skin diseases. Next to erythema, edema, and sunburns, UV radiation causes skin cancer. UV radiation of any wavelength is now considered as a class I carcinogen to humans. The mutagenic effects of UV radiation depend on DNA damage following direct absorption by nuclear DNA, resulting in cyclobutane pyrimidine dimers and pyrimidine (6-4) pyrimidone photoproducts that, if not repaired by nucleotide excision repair pathway, result in characteristic UV signature mutations (C→T or CC→TT transition). In addition, increased formation of reactive oxygen species by UV exposure may cause formation of 8-oxo-7,8-dihydro-2'-deoxyguanosine leading to T→G transversion. In addition, UV radiation has been shown to induce a number of immune modulations that largely result in local and potentially also in systemic immunosuppression, which may not only impair control of dysplastic and neoplastic skin lesions but also affect immuno-pathological and infectious skin diseases. Recent find-ings have shown that ambient doses of high-energy visible light, beyond the UV range, may also cause damage to human skin.


Skin Diseases/etiology , Skin/radiation effects , Ultraviolet Rays/adverse effects , DNA Damage/radiation effects , Humans , Pyrimidine Dimers/radiation effects , Reactive Oxygen Species/metabolism , Skin/immunology , Skin/pathology , Skin Diseases/pathology
10.
EMBO J ; 40(20): e107795, 2021 10 18.
Article En | MEDLINE | ID: mdl-34487363

Somatic mutations in DNA-binding sites for CCCTC-binding factor (CTCF) are significantly elevated in many cancers. Prior analysis has suggested that elevated mutation rates at CTCF-binding sites in skin cancers are a consequence of the CTCF-cohesin complex inhibiting repair of UV damage. Here, we show that CTCF binding modulates the formation of UV damage to induce mutation hot spots. Analysis of genome-wide CPD-seq data in UV-irradiated human cells indicates that formation of UV-induced cyclobutane pyrimidine dimers (CPDs) is primarily suppressed by CTCF binding but elevated at specific locations within the CTCF motif. Locations of CPD hot spots in the CTCF-binding motif coincide with mutation hot spots in melanoma. A similar pattern of damage formation is observed at CTCF-binding sites in vitro, indicating that UV damage modulation is a direct consequence of CTCF binding. We show that CTCF interacts with binding sites containing UV damage and inhibits repair by a model repair enzyme in vitro. Structural analysis and molecular dynamic simulations reveal the molecular mechanism for how CTCF binding modulates CPD formation.


CCCTC-Binding Factor/chemistry , DNA Repair , Melanoma/genetics , Protein Serine-Threonine Kinases/chemistry , Pyrimidine Dimers/radiation effects , Skin Neoplasms/genetics , Binding Sites , Binding, Competitive , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Line, Tumor , DNA Damage , Gene Expression , Humans , Melanoma/metabolism , Melanoma/pathology , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyrimidine Dimers/biosynthesis , Pyrimidine Dimers/chemistry , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Ultraviolet Rays
11.
J Dermatol Sci ; 103(3): 151-155, 2021 Sep.
Article En | MEDLINE | ID: mdl-34391606

BACKGROUND: Maximum cyclobutane pyrimidine dimer (CPD) formation in the skin induced by ultraviolet B (UVB) irradiation is thought to occur within a few minutes and is immediately decreased by the DNA repair system. OBJECTIVE: We evaluated the time course and differential effects of narrowband (NB-UVB) and broadband (BB-UVB) UVB on CPD formation. METHODS: We investigated CPD formation at various time-points in vivo, from 3 min to 72 h, after UVB irradiation using 2 mouse strains, C57BL/6 J and BALB/c. The backs of the mice were shaved and irradiated with NB-UVB or BB-UVB. Skin specimens were obtained and stained with anti-CPD antibody. Positive signals in the epidermis were measured using ImageJ. DNA was extracted from the isolated epidermis and subjected to quantitative CPD analysis by enzyme-linked immunosorbent assay (ELISA). RESULTS: CPDs induced by UVB irradiation (1 minimum erythemal dose) in epidermal skin were detected in the nucleus. Although the CPD levels increased immediately after irradiation (3 min), the highest level was detected at 1 h and the increase lasted 24-48 h after irradiation. BB-UVB tended to induce greater CPD levels than NB-UVB in both mouse strains. The ELISA showed similar results. CONCLUSIONS: CPDs were induced immediately after UV irradiation, with the maximum level observed 1 h after irradiation. BB-UVB irradiation tended to induce greater levels of CPD formation. In addition to the direct effects of UVB, the presence of CPDs in hair follicles, which were not irradiated by UVB, suggests that reactive oxygen species are also involved in CPD formation in the skin.


DNA Damage/radiation effects , Epidermis/radiation effects , Pyrimidine Dimers/analysis , Ultraviolet Rays/adverse effects , Animals , DNA Repair , Epidermis/chemistry , Epidermis/metabolism , Hair Follicle/chemistry , Hair Follicle/metabolism , Mice , Models, Animal , Pyrimidine Dimers/radiation effects , Reactive Oxygen Species/metabolism , Time Factors
12.
Int J Mol Sci ; 22(4)2021 Feb 16.
Article En | MEDLINE | ID: mdl-33669452

Susceptibility to photoimmune suppression and photocarcinogenesis is greater in male than in female humans and mice and is exacerbated in female estrogen receptor-beta knockout (ER-ß-/-) mice. We previously reported that the active vitamin D hormone, 1,25-dihydroxyvitamin D3 (1,25(OH)2D), applied topically protects against the ultraviolet radiation (UV) induction of cutaneous cyclobutane pyrimidine dimers (CPDs) and the suppression of contact hypersensitivity (CHS) in female mice. Here, we compare these responses in female versus male Skh:hr1 mice, in ER-ß-/-/-- versus wild-type C57BL/6 mice, and in female ER-blockaded Skh:hr1 mice. The induction of CPDs was significantly greater in male than female Skh:hr1 mice and was more effectively reduced by 1,25(OH)2D in female Skh:hr1 and C57BL/6 mice than in male Skh:hr1 or ER-ß-/- mice, respectively. This correlated with the reduced sunburn inflammation due to 1,25(OH)2D in female but not male Skh:hr1 mice. Furthermore, although 1,25(OH)2D alone dose-dependently suppressed basal CHS responses in male Skh:hr1 and ER-ß-/- mice, UV-induced immunosuppression was universally observed. In female Skh:hr1 and C57BL/6 mice, the immunosuppression was decreased by 1,25(OH)2D dose-dependently, but not in male Skh:hr1, ER-ß-/-, or ER-blockaded mice. These results reveal a sex bias in genetic, inflammatory, and immune photoprotection by 1,25(OH)2D favoring female mice that is dependent on the presence of ER-ß.


Calcitriol/administration & dosage , Estrogen Receptor beta/metabolism , Signal Transduction/radiation effects , Sunburn/drug therapy , Sunburn/metabolism , Sunscreening Agents/administration & dosage , Ultraviolet Rays , Administration, Cutaneous , Animals , Dermatitis, Contact/drug therapy , Disease Models, Animal , Estrogen Receptor beta/genetics , Female , Immune Tolerance/drug effects , Immune Tolerance/radiation effects , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyrimidine Dimers/metabolism , Pyrimidine Dimers/radiation effects , Sex Factors , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin/radiation effects , Skin Neoplasms/prevention & control , Sunburn/prevention & control
13.
J Invest Dermatol ; 141(7): 1656-1663, 2021 07.
Article En | MEDLINE | ID: mdl-33524369

Vitamin D receptor (VDR) is important for normal DNA repair, although the mechanism by which it acts is unclear. After focal UV irradiation to create subnuclear spots of DNA damage, epidermal keratinocytes from VDR-null mice as well as human epidermal keratinocytes depleted of VDR with small interfering RNA removed pyrimidine-pyrimidone (6-4) photoproducts more slowly than control cells. Costaining with antibodies to XPC, the DNA damage recognition sensor that initiates nucleotide excision repair, showed that XPC rapidly accumulated at spots of damage and gradually faded in control human keratinocytes. In VDR-depleted keratinocytes, XPC associated with DNA damage with comparable efficiency; however, XPC's dissociation dynamics were altered so that significantly more XPC was bound and retained over time than in control cells. The XPF endonuclease, which acts subsequently in nucleotide excision repair, bound and dissociated with comparable kinetics in control and VDR-depleted cells, but the extent of binding was reduced in the latter. These results as well as kinetic modeling of the data suggest that VDR's importance in the repair of UV-induced DNA damage is mediated in part by its ability to facilitate the dissociation of XPC from damaged DNA for the normal recruitment and assembly of other repair proteins to proceed.


DNA Repair , DNA-Binding Proteins/metabolism , Receptors, Calcitriol/metabolism , Animals , Cells, Cultured , DNA/metabolism , DNA/radiation effects , DNA Damage/radiation effects , Humans , Keratinocytes/metabolism , Keratinocytes/radiation effects , Male , Mice, Knockout , Primary Cell Culture , Pyrimidine Dimers/metabolism , Pyrimidine Dimers/radiation effects , RNA Interference , Receptors, Calcitriol/genetics , Ultraviolet Rays/adverse effects
14.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article En | MEDLINE | ID: mdl-33526704

In this study, absorption, fluorescence, synchronous fluorescence, and Raman spectra of nonirradiated and ultraviolet (UV)-irradiated thymine solutions were recorded in order to detect thymine dimer formation. The thymine dimer formation, as a function of irradiation dose, was determined by Raman spectroscopy. In addition, the formation of a mutagenic (6-4) photoproduct was identified by its synchronous fluorescence spectrum. Our spectroscopic data suggest that the rate of conversion of thymine to thymine dimer decreases after 20 min of UV irradiation, owing to the formation of an equilibrium between the thymine dimers and monomers. However, the formation of the (6-4) photoproduct continued to increase with UV irradiation. In addition, the Raman spectra of nonirradiated and irradiated calf thymus DNA were recorded, and the formation of thymine dimers was detected. The spectroscopic data presented make it possible to determine the mechanism of thymine dimer formation, which is known to be responsible for the inhibition of DNA replication that causes bacteria inactivation.


DNA Damage/radiation effects , DNA/genetics , Pyrimidine Dimers/genetics , Thymine/chemistry , Animals , Cattle , DNA/chemistry , DNA/radiation effects , DNA Damage/genetics , Pyrimidine Dimers/chemistry , Pyrimidine Dimers/radiation effects , Spectrometry, Fluorescence , Spectrum Analysis, Raman , Thymine/radiation effects , Ultraviolet Rays/adverse effects
15.
Life Sci Alliance ; 4(4)2021 04.
Article En | MEDLINE | ID: mdl-33514655

By extending synthesis opposite from a diverse array of DNA lesions, DNA polymerase (Pol) ζ performs a crucial role in translesion synthesis (TLS). In yeast and cancer cells, Rev1 functions as an indispensable scaffolding component of Polζ and it imposes highly error-prone TLS upon Polζ. However, for TLS that occurs during replication in normal human cells, Rev1 functions instead as a scaffolding component of Pols η, ι, and κ and Rev1-dependent TLS by these Pols operates in a predominantly error-free manner. The lack of Rev1 requirement for Polζ function in TLS in normal cells suggested that some other protein substitutes for this Rev1 role. Here, we identify a novel role of Polλ as an indispensable scaffolding component of Polζ. TLS studies opposite a number of DNA lesions support the conclusion that as an integral component, Polλ adapts Polζ-dependent TLS to operate in a predominantly error-free manner in human cells, essential for genome integrity and cellular homeostasis.


DNA Polymerase beta/metabolism , DNA Replication , Catalysis , Cells, Cultured , DNA Adducts , DNA Damage , DNA Replication/radiation effects , DNA-Directed DNA Polymerase/metabolism , Gene Knockdown Techniques , Humans , Mutation , Pyrimidine Dimers/radiation effects , Recombinant Fusion Proteins , Ultraviolet Rays
16.
Pigment Cell Melanoma Res ; 34(3): 575-584, 2021 05.
Article En | MEDLINE | ID: mdl-33369222

INTRODUCTION: Unlike "light" cylobutane pyrimidine dimers (CPD) formed during ultraviolet radiation (UVR) exposure, dark CPD (dCPD) are formed afterwards. Studies have attributed this to delayed melanin sensitization. There are no data on the role of melanin in dCPD formation in human skin. METHODS AND RESULTS: Volunteers of Fitzpatrick skin types (FST I/II vs. VI) were exposed to erythemally equivalent doses of solar simulated radiation. CPD were assessed by semi-quantitative immunostaining in whole epidermis and in three epidermal zones, and quantitative HPLC-MS/MS (whole epidermis) at different times post-exposure up to 24 hr. A CPD peak that appeared at 1-2 hr post-exposure in whole epidermis measurements, in all skin types, demonstrated dCPD. However, both dCPD and light CPD were absent in the basal layer of FST VI with the greatest melanin concentration. Modelling the whole epidermis data showed no differences between the repair kinetics of FST I/II and VI. DISCUSSION: Melanin may be a sensitizer or "sunscreen" for dCPD depending on its location and concentration. Previous CPD repair studies in human skin have assumed peak CPD immediately after UVR exposure and so have overestimated total repair.


Epidermis/pathology , Melanins/metabolism , Pyrimidine Dimers/chemistry , Skin Aging , Skin Pigmentation , Skin/pathology , Ultraviolet Rays/adverse effects , Adult , DNA Damage , Epidermis/metabolism , Epidermis/radiation effects , Female , Humans , Male , Pyrimidine Dimers/radiation effects , Skin/metabolism , Skin/radiation effects , Young Adult
17.
Nucleic Acids Res ; 48(22): 12845-12857, 2020 12 16.
Article En | MEDLINE | ID: mdl-33270891

Photolyases are ubiquitously occurring flavoproteins for catalyzing photo repair of UV-induced DNA damages. All photolyases described so far have a bilobal architecture with a C-terminal domain comprising flavin adenine dinucleotide (FAD) as catalytic cofactor and an N-terminal domain capable of harboring an additional antenna chromophore. Using sequence-similarity network analysis we discovered a novel subgroup of the photolyase/cryptochrome superfamily (PCSf), the NewPHLs. NewPHL occur in bacteria and have an inverted topology with an N-terminal catalytic domain and a C-terminal domain for sealing the FAD binding site from solvent access. By characterizing two NewPHL we show a photochemistry characteristic of other PCSf members as well as light-dependent repair of CPD lesions. Given their common specificity towards single-stranded DNA many bacterial species use NewPHL as a substitute for DASH-type photolyases. Given their simplified architecture and function we suggest that NewPHL are close to the evolutionary origin of the PCSf.


Cryptochromes/genetics , DNA, Single-Stranded/genetics , Deoxyribodipyrimidine Photo-Lyase/genetics , Amino Acid Sequence/genetics , Catalytic Domain/genetics , Catalytic Domain/radiation effects , DNA Damage/radiation effects , DNA Repair/radiation effects , DNA, Single-Stranded/radiation effects , Deoxyribodipyrimidine Photo-Lyase/radiation effects , Methylobacterium/genetics , Pyrimidine Dimers/genetics , Pyrimidine Dimers/radiation effects , Rhodobacteraceae/genetics , Ultraviolet Rays
18.
Genes (Basel) ; 11(11)2020 11 04.
Article En | MEDLINE | ID: mdl-33158066

Although solar light is indispensable for the functioning of plants, this environmental factor may also cause damage to living cells. Apart from the visible range, including wavelengths used in photosynthesis, the ultraviolet (UV) light present in solar irradiation reaches the Earth's surface. The high energy of UV causes damage to many cellular components, with DNA as one of the targets. Putting together the puzzle-like elements responsible for the repair of UV-induced DNA damage is of special importance in understanding how plants ensure the stability of their genomes between generations. In this review, we have presented the information on DNA damage produced under UV with a special focus on the pyrimidine dimers formed between the neighboring pyrimidines in a DNA strand. These dimers are highly mutagenic and cytotoxic, thus their repair is essential for the maintenance of suitable genetic information. In prokaryotic and eukaryotic cells, with the exception of placental mammals, this is achieved by means of highly efficient photorepair, dependent on blue/UVA light, which is performed by specialized enzymes known as photolyases. Photolyase properties, as well as their structure, specificity and action mechanism, have been briefly discussed in this paper. Additionally, the main gaps in our knowledge on the functioning of light repair in plant organelles, its regulation and its interaction between different DNA repair systems in plants have been highlighted.


DNA Repair/physiology , Deoxyribodipyrimidine Photo-Lyase/metabolism , Pyrimidine Dimers/genetics , Animals , DNA/genetics , DNA/metabolism , DNA Damage/genetics , DNA Repair/genetics , Deoxyribodipyrimidine Photo-Lyase/chemistry , Deoxyribodipyrimidine Photo-Lyase/genetics , Humans , Mutagenesis/genetics , Pyrimidine Dimers/radiation effects , Ultraviolet Rays/adverse effects
19.
PLoS One ; 15(8): e0235948, 2020.
Article En | MEDLINE | ID: mdl-32785216

INTRODUCTION: Surgical site infection is one of the most severe complications of surgical treatments. However, the optimal procedure to prevent such infections remains uninvestigated. Ultraviolet radiation C (UVC) with a short wavelength has a high bactericidal effect; however, it is cytotoxic. Nonetheless, given that UVC with a wavelength of 222 nm reaches only the stratum corneum, it does not affect the skin cells. This study aimed to investigate the safety of 222-nm UVC irradiation and to examine its skin sterilization effect in healthy volunteers. METHODS: This trial was conducted on 20 healthy volunteers. The back of the subject was irradiated with 222-nm UVC at 50-500 mJ/cm2, and the induced erythema (redness of skin) was evaluated. Subsequently, the back was irradiated with a maximum amount of UVC not causing erythema, and the skin swabs before and after the irradiation were cultured. The number of colonies formed after 24 hours was measured. In addition, cyclobutene pyrimidine dimer (CPD) as an indicator of DNA damage was measured using skin tissues of the nonirradiated and irradiated regions. RESULTS: All subjects experienced no erythema at all doses. The back of the subject was irradiated at 500 mJ/cm2, and the number of bacterial colonies in the skin swab culture was significantly decreased by 222-nm UVC irradiation. The CPD amount produced in the irradiated region was slightly but significantly higher than that of the non-irradiated region. CONCLUSION: A 222-nm UVC at 500 mJ/cm2 was a safe irradiation dose and possessed bactericidal effects. In the future, 222-nm UVC irradiation is expected to contribute to the prevention of perioperative infection.


DNA Damage/radiation effects , Microbiota/radiation effects , Skin/radiation effects , Sterilization/methods , Ultraviolet Rays/adverse effects , Adult , Back , Biopsy , Colony Count, Microbial , Erythema/diagnosis , Erythema/etiology , Healthy Volunteers , Humans , Male , Pyrimidine Dimers/analysis , Pyrimidine Dimers/radiation effects , Skin/microbiology , Surgical Wound Infection/microbiology , Surgical Wound Infection/prevention & control , Treatment Outcome
20.
Mol Cell Biol ; 40(20)2020 09 28.
Article En | MEDLINE | ID: mdl-32778572

Ultraviolet (UV) radiation is a major environmental mutagen. Exposure to UV leads to a sharp peak of γH2AX, the phosphorylated form of the histone variant H2AX, in the S phase within an asynchronous population of cells. γH2AX is often considered a definitive marker of DNA damage inside a cell. In this report, we show that γH2AX in the S-phase cells after UV irradiation reports neither on the extent of primary DNA damage in the form of cyclobutane pyrimidine dimers nor on the extent of its secondary manifestations in the form of DNA double-strand breaks or in the inhibition of global transcription. Instead, γH2AX in the S phase corresponds to the sites of active replication at the time of UV irradiation. This accumulation of γH2AX at replication sites slows down the replication. However, the cells do complete the replication of their genomes and arrest within the G2 phase. Our study suggests that it is not DNA damage, but the response elicited, which peaks in the S phase upon UV irradiation.


DNA Breaks, Double-Stranded/radiation effects , DNA Replication/genetics , Histones/genetics , Pyrimidine Dimers/radiation effects , S Phase/radiation effects , A549 Cells , Cell Line, Tumor , DNA/biosynthesis , DNA Repair/genetics , DNA Replication/radiation effects , Humans , Phosphorylation/radiation effects , S Phase/genetics , Ultraviolet Rays
...