Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.606
Filter
Add more filters

Publication year range
1.
Mol Cell ; 83(20): 3669-3678.e7, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37816354

ABSTRACT

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.


Subject(s)
Xeroderma Pigmentosum , Humans , Xeroderma Pigmentosum/genetics , DNA Repair/genetics , Pyrimidine Dimers/genetics , Pyrimidine Dimers/metabolism , DNA Damage/genetics , DNA/genetics , Ultraviolet Rays , X-ray Repair Cross Complementing Protein 1/metabolism
2.
Cell ; 163(7): 1692-701, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26687357

ABSTRACT

Vesicular nucleo-cytoplasmic transport is becoming recognized as a general cellular mechanism for translocation of large cargoes across the nuclear envelope. Cargo is recruited, enveloped at the inner nuclear membrane (INM), and delivered by membrane fusion at the outer nuclear membrane. To understand the structural underpinning for this trafficking, we investigated nuclear egress of progeny herpesvirus capsids where capsid envelopment is mediated by two viral proteins, forming the nuclear egress complex (NEC). Using a multi-modal imaging approach, we visualized the NEC in situ forming coated vesicles of defined size. Cellular electron cryo-tomography revealed a protein layer showing two distinct hexagonal lattices at its membrane-proximal and membrane-distant faces, respectively. NEC coat architecture was determined by combining this information with integrative modeling using small-angle X-ray scattering data. The molecular arrangement of the NEC establishes the basic mechanism for budding and scission of tailored vesicles at the INM.


Subject(s)
Active Transport, Cell Nucleus , Capsid/metabolism , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Transport Vesicles/ultrastructure , Animals , Capsid/ultrastructure , Chlorocebus aethiops , Cryoelectron Microscopy , Electron Microscope Tomography , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Suid/metabolism , Nuclear Envelope/chemistry , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Pyrimidine Dimers , Scattering, Small Angle , Transport Vesicles/metabolism , Vero Cells , Viral Proteins/chemistry , Viral Proteins/metabolism
3.
Proc Natl Acad Sci U S A ; 121(4): e2310854121, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38241433

ABSTRACT

Noncoding mutation hotspots have been identified in melanoma and many of them occur at the binding sites of E26 transformation-specific (ETS) proteins; however, their formation mechanism and functional impacts are not fully understood. Here, we used UV (Ultraviolet) damage sequencing data and analyzed cyclobutane pyrimidine dimer (CPD) formation, DNA repair, and CPD deamination in human cells at single-nucleotide resolution. Our data show prominent CPD hotspots immediately after UV irradiation at ETS binding sites, particularly at sites with a conserved TTCCGG motif, which correlate with mutation hotspots identified in cutaneous melanoma. Additionally, CPDs are repaired slower at ETS binding sites than in flanking DNA. Cytosine deamination in CPDs to uracil is suggested as an important step for UV mutagenesis. However, we found that CPD deamination is significantly suppressed at ETS binding sites, particularly for the CPD hotspot on the 5' side of the ETS motif, arguing against a role for CPD deamination in promoting ETS-associated UV mutations. Finally, we analyzed a subset of frequently mutated promoters, including the ribosomal protein genes RPL13A and RPS20, and found that mutations in the ETS motif can significantly reduce the promoter activity. Thus, our data identify high UV damage and low repair, but not CPD deamination, as the main mechanism for ETS-associated mutations in melanoma and uncover important roles of often-overlooked mutation hotspots in perturbing gene transcription.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Melanoma/genetics , Cytosine , Deamination , Skin Neoplasms/genetics , Mutation , Pyrimidine Dimers , Binding Sites , Ultraviolet Rays , DNA Damage , DNA Repair/genetics
4.
Nucleic Acids Res ; 52(14): 8254-8270, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-38884271

ABSTRACT

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.


Subject(s)
DNA Repair , DNA-Binding Proteins , Enhancer Elements, Genetic , Histone-Lysine N-Methyltransferase , Pyrimidine Dimers , Humans , Mice , DNA/metabolism , DNA/chemistry , DNA/genetics , DNA Damage , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Histones/genetics , Molecular Dynamics Simulation , Promoter Regions, Genetic , Pyrimidine Dimers/metabolism , Pyrimidine Dimers/genetics , Pyrimidine Dimers/chemistry , Transcription Factors/metabolism , Transcription Factors/genetics , Ultraviolet Rays
5.
Nucleic Acids Res ; 52(13): 7437-7446, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38908029

ABSTRACT

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.


Subject(s)
8-Hydroxy-2'-Deoxyguanosine , DNA Damage , Deoxyguanosine , Promoter Regions, Genetic , RNA Polymerase II , Humans , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , HEK293 Cells , 8-Hydroxy-2'-Deoxyguanosine/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , HeLa Cells , DNA Repair , Transcription, Genetic , Pyrimidines , Pyrimidine Dimers/metabolism , Pyrimidine Dimers/genetics
6.
Proc Natl Acad Sci U S A ; 120(25): e2220132120, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37307476

ABSTRACT

Understanding and predicting the outcome of the interaction of light with DNA has a significant impact on the study of DNA repair and radiotherapy. We report on a combination of femtosecond pulsed laser microirradiation at different wavelengths, quantitative imaging, and numerical modeling that yields a comprehensive picture of photon-mediated and free-electron-mediated DNA damage pathways in live cells. Laser irradiation was performed under highly standardized conditions at four wavelengths between 515 nm and 1,030 nm, enabling to study two-photon photochemical and free-electron-mediated DNA damage in situ. We quantitatively assessed cyclobutane pyrimidine dimer (CPD) and γH2AX-specific immunofluorescence signals to calibrate the damage threshold dose at these wavelengths and performed a comparative analysis of the recruitment of DNA repair factors xeroderma pigmentosum complementation group C (XPC) and Nijmegen breakage syndrome 1 (Nbs1). Our results show that two-photon-induced photochemical CPD generation dominates at 515 nm, while electron-mediated damage dominates at wavelengths ≥620 nm. The recruitment analysis revealed a cross talk between nucleotide excision and homologous recombination DNA repair pathways at 515 nm. Numerical simulations predicted electron densities and electron energy spectra, which govern the yield functions of a variety of direct electron-mediated DNA damage pathways and of indirect damage by •OH radicals resulting from laser and electron interactions with water. Combining these data with information on free electron-DNA interactions gained in artificial systems, we provide a conceptual framework for the interpretation of the wavelength dependence of laser-induced DNA damage that may guide the selection of irradiation parameters in studies and applications that require the selective induction of DNA lesions.


Subject(s)
DNA Damage , Electrons , Pyrimidine Dimers , DNA Repair , Lasers
7.
Proc Natl Acad Sci U S A ; 120(10): e2216907120, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36853943

ABSTRACT

Ultraviolet (UV) light induces different classes of mutagenic photoproducts in DNA, namely cyclobutane pyrimidine dimers (CPDs), 6-4 photoproducts (6-4PPs), and atypical thymine-adenine photoproducts (TA-PPs). CPD formation is modulated by nucleosomes and transcription factors (TFs), which has important ramifications for Ultraviolet (UV) mutagenesis. How chromatin affects the formation of 6-4PPs and TA-PPs is unclear. Here, we use UV damage endonuclease-sequencing (UVDE-seq) to map these UV photoproducts across the yeast genome. Our results indicate that nucleosomes, the fundamental building block of chromatin, have opposing effects on photoproduct formation. Nucleosomes induce CPDs and 6-4PPs at outward rotational settings in nucleosomal DNA but suppress TA-PPs at these settings. Our data also indicate that DNA binding by different classes of yeast TFs causes lesion-specific hotspots of 6-4PPs or TA-PPs. For example, DNA binding by the TF Rap1 generally suppresses CPD and 6-4PP formation but induces a TA-PP hotspot. Finally, we show that 6-4PP formation is strongly induced at the binding sites of TATA-binding protein (TBP), which is correlated with higher mutation rates in UV-exposed yeast. These results indicate that the formation of 6-4PPs and TA-PPs is modulated by chromatin differently than CPDs and that this may have important implications for UV mutagenesis.


Subject(s)
Chromatin , Saccharomyces cerevisiae , Chromatin/genetics , Saccharomyces cerevisiae/genetics , Nucleosomes/genetics , Mutagenesis , Mutagens , Adenine , Pyrimidine Dimers/genetics
8.
EMBO J ; 40(20): e107795, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34487363

ABSTRACT

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.


Subject(s)
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
9.
Plant Physiol ; 195(1): 326-342, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38345835

ABSTRACT

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.


Subject(s)
Chloroplasts , Deoxyribodipyrimidine Photo-Lyase , Oryza , Ultraviolet Rays , Chloroplasts/metabolism , Deoxyribodipyrimidine Photo-Lyase/metabolism , Deoxyribodipyrimidine Photo-Lyase/genetics , Oryza/genetics , Oryza/enzymology , Oryza/radiation effects , Oryza/metabolism , Plant Proteins/metabolism , Plant Proteins/genetics , Pyrimidine Dimers/metabolism , Poaceae/genetics , Poaceae/enzymology , Poaceae/radiation effects , Poaceae/metabolism , Amino Acid Sequence , Protein Transport
10.
Nature ; 571(7763): 79-84, 2019 07.
Article in English | MEDLINE | ID: mdl-31142837

ABSTRACT

Access to DNA packaged in nucleosomes is critical for gene regulation, DNA replication and DNA repair. In humans, the UV-damaged DNA-binding protein (UV-DDB) complex detects UV-light-induced pyrimidine dimers throughout the genome; however, it remains unknown how these lesions are recognized in chromatin, in which nucleosomes restrict access to DNA. Here we report cryo-electron microscopy structures of UV-DDB bound to nucleosomes bearing a 6-4 pyrimidine-pyrimidone dimer or a DNA-damage mimic in various positions. We find that UV-DDB binds UV-damaged nucleosomes at lesions located in the solvent-facing minor groove without affecting the overall nucleosome architecture. In the case of buried lesions that face the histone core, UV-DDB changes the predominant translational register of the nucleosome and selectively binds the lesion in an accessible, exposed position. Our findings explain how UV-DDB detects occluded lesions in strongly positioned nucleosomes, and identify slide-assisted site exposure as a mechanism by which high-affinity DNA-binding proteins can access otherwise occluded sites in nucleosomal DNA.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , DNA/metabolism , DNA/ultrastructure , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Pyrimidine Dimers/analysis , Cryoelectron Microscopy , DNA/chemistry , DNA/radiation effects , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/ultrastructure , Histones/chemistry , Histones/metabolism , Histones/ultrastructure , Humans , Models, Molecular , Nucleosomes/genetics , Nucleosomes/radiation effects , Pyrimidine Dimers/chemistry , Pyrimidine Dimers/genetics , Thermodynamics , Ultraviolet Rays/adverse effects
11.
Nucleic Acids Res ; 51(11): 5341-5350, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37207339

ABSTRACT

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.


Subject(s)
DNA Damage , Pyrimidine Dimers , Pyrimidine Dimers/radiation effects , DNA/genetics , DNA Repair , Ultraviolet Rays , Nucleic Acid Conformation
12.
Nucleic Acids Res ; 51(10): 4982-4994, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37026475

ABSTRACT

Accumulation of DNA damage resulting from reactive oxygen species was proposed to cause neurological and degenerative disease in patients, deficient in nucleotide excision repair (NER) or its transcription-coupled subpathway (TC-NER). Here, we assessed the requirement of TC-NER for the repair of specific types of oxidatively generated DNA modifications. We incorporated synthetic 5',8-cyclo-2'-deoxypurine nucleotides (cyclo-dA, cyclo-dG) and thymine glycol (Tg) into an EGFP reporter gene to measure transcription-blocking potentials of these modifications in human cells. Using null mutants, we further identified the relevant DNA repair components by a host cell reactivation approach. The results indicated that NTHL1-initiated base excision repair is by far the most efficient pathway for Tg. Moreover, Tg was efficiently bypassed during transcription, which effectively rules out TC-NER as an alternative repair mechanism. In a sharp contrast, both cyclopurine lesions robustly blocked transcription and were repaired by NER, wherein the specific TC-NER components CSB/ERCC6 and CSA/ERCC8 were as essential as XPA. Instead, repair of classical NER substrates, cyclobutane pyrimidine dimer and N-(deoxyguanosin-8-yl)-2-acetylaminofluorene, occurred even when TC-NER was disrupted. The strict requirement of TC-NER highlights cyclo-dA and cyclo-dG as candidate damage types, accountable for cytotoxic and degenerative responses in individuals affected by genetic defects in this pathway.


Subject(s)
DNA Repair , Transcription, Genetic , Humans , DNA Damage , DNA Repair Enzymes/genetics , Pyrimidine Dimers , Transcription Factors/genetics
13.
Nucleic Acids Res ; 51(22): 12261-12274, 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-37933861

ABSTRACT

Ultraviolet light generates cyclobutane pyrimidine dimer (CPD) and pyrimidine 6-4 pyrimidone (6-4PP) photoproducts that cause skin malignancies if not repaired by nucleotide excision repair (NER). While the faster repair of the more distorting 6-4PPs is attributed mainly to more efficient recognition by XPC, the XPD lesion verification helicase may play a role, as it directly scans the damaged DNA strand. With extensive molecular dynamics simulations of XPD-bound single-strand DNA containing each lesion outside the entry pore of XPD, we elucidate strikingly different verification processes for these two lesions that have very different topologies. The open book-like CPD thymines are sterically blocked from pore entry and preferably entrapped by sensors that are outside the pore; however, the near-perpendicular 6-4PP thymines can enter, accompanied by a displacement of the Arch domain toward the lesion, which is thereby tightly accommodated within the pore. This trapped 6-4PP may inhibit XPD helicase activity to foster lesion verification by locking the Arch to other domains. Furthermore, the movement of the Arch domain, only in the case of 6-4PP, may trigger signaling to the XPG nuclease for subsequent lesion incision by fostering direct contact between the Arch domain and XPG, and thereby facilitating repair of 6-4PP.


Subject(s)
DNA Repair , Pyrimidine Dimers , Humans , DNA , DNA Damage , DNA Helicases/genetics , Ultraviolet Rays
14.
Nucleic Acids Res ; 51(19): 10132-10146, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37757853

ABSTRACT

UV radiation-induced DNA damages have adverse effects on genome integrity and cellular function. The most prevalent UV-induced DNA lesion is the cyclobutane pyrimidine dimer (CPD), which can cause skin disorders and cancers in humans. Rad4/XPC is a damage sensing protein that recognizes and repairs CPD lesions with high fidelity. However, the molecular mechanism of how Rad4/XPC interrogates CPD lesions remains elusive. Emerging viewpoints indicate that the association of Rad4/XPC with DNA, the insertion of a lesion-sensing ß-hairpin of Rad4/XPC into the lesion site and the flipping of CPD's partner bases (5'-dA and 3'-dA) are essential for damage recognition. Characterizing these slow events is challenging due to their infrequent occurrence on molecular time scales. Herein, we have used enhanced sampling and molecular dynamics simulations to investigate the mechanism and energetics of lesion recognition by Rad4/XPC, considering multiple plausible pathways between the crystal structure of the Rad4-DNA complex and nine intermediate states. Our results shed light on the most likely sequence of events, their potential coupling and energetics. Upon association, Rad4 and DNA form an encounter complex in which CPD and its partner bases remain in the duplex and the BHD3 ß-hairpin is yet to be inserted into the lesion site. Subsequently, sequential base flipping occurs, with the flipping of the 5'-dA base preceding that of the 3'-dA base, followed by the insertion of the BHD3 ß-hairpin into the lesion site. The results presented here have significant implications for understanding the molecular basis of UV-related skin disorders and cancers and for paving the way for novel therapeutic strategies.


Subject(s)
Neoplasms , Saccharomyces cerevisiae Proteins , Humans , Pyrimidine Dimers/chemistry , DNA Damage , DNA Repair , Saccharomyces cerevisiae Proteins/genetics , Protein Binding , DNA-Binding Proteins/metabolism , DNA/chemistry , Ultraviolet Rays
15.
PLoS Genet ; 18(9): e1010426, 2022 09.
Article in English | MEDLINE | ID: mdl-36155646

ABSTRACT

Nucleotide excision repair is the primary repair mechanism that removes UV-induced DNA lesions in placentals. Unrepaired UV-induced lesions could result in mutations during DNA replication. Although the mutagenesis of pyrimidine dimers is reasonably well understood, the direct effects of replication fork progression on nucleotide excision repair are yet to be clarified. Here, we applied Damage-seq and XR-seq techniques and generated replication maps in synchronized UV-treated HeLa cells. The results suggest that ongoing replication stimulates local repair in both early and late replication domains. Additionally, it was revealed that lesions on lagging strand templates are repaired slower in late replication domains, which is probably due to the imbalanced sequence context. Asymmetric relative repair is in line with the strand bias of melanoma mutations, suggesting a role of exogenous damage, repair, and replication in mutational strand asymmetry.


Subject(s)
Pyrimidine Dimers , Ultraviolet Rays , DNA/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA Replication/genetics , HeLa Cells , Humans , Pyrimidine Dimers/genetics , Ultraviolet Rays/adverse effects
16.
PLoS Genet ; 18(4): e1010167, 2022 04.
Article in English | MEDLINE | ID: mdl-35486666

ABSTRACT

Ultraviolet light causes DNA lesions that are removed by nucleotide excision repair (NER). The efficiency of NER is conditional to transcription and chromatin structure. UV induced photoproducts are repaired faster in the gene transcribed strands than in the non-transcribed strands or in transcriptionally inactive regions of the genome. This specificity of NER is known as transcription-coupled repair (TCR). The discovery of pervasive non-coding RNA transcription (ncRNA) advocates for ubiquitous contribution of TCR to the repair of UV photoproducts, beyond the repair of active gene-transcribed strands. Chromatin rules transcription, and telomeres form a complex structure of proteins that silences nearby engineered ectopic genes. The essential protective function of telomeres also includes preventing unwanted repair of double-strand breaks. Thus, telomeres were thought to be transcriptionally inert, but more recently, ncRNA transcription was found to initiate in subtelomeric regions. On the other hand, induced DNA lesions like the UV photoproducts must be recognized and repaired also at the ends of chromosomes. In this study, repair of UV induced DNA lesions was analyzed in the subtelomeric regions of budding yeast. The T4-endonuclease V nicking-activity at cyclobutene pyrimidine dimer (CPD) sites was exploited to monitor CPD formation and repair. The presence of two photoproducts, CPDs and pyrimidine (6,4)-pyrimidones (6-4PPs), was verified by the effective and precise blockage of Taq DNA polymerase at these sites. The results indicate that UV photoproducts in silenced heterochromatin are slowly repaired, but that ncRNA transcription enhances NER throughout one subtelomeric element, called Y', and in distinct short segments of the second, more conserved element, called X. Therefore, ncRNA-transcription dependent TCR assists global genome repair to remove CPDs and 6-4PPs from subtelomeric DNA.


Subject(s)
Saccharomyces cerevisiae , Ultraviolet Rays , Chromatin , DNA , DNA Damage/genetics , DNA Repair/genetics , Heterochromatin , Pyrimidine Dimers/genetics , RNA, Untranslated/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomere/genetics , Telomere/metabolism , Transcription, Genetic
17.
J Biol Chem ; 299(5): 104679, 2023 05.
Article in English | MEDLINE | ID: mdl-37028766

ABSTRACT

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.


Subject(s)
DNA Damage , DNA Repair , Epigenesis, Genetic , Pyrimidine Dimers/metabolism , Ultraviolet Rays/adverse effects
18.
J Am Chem Soc ; 146(30): 20742-20749, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39037865

ABSTRACT

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


Subject(s)
DNA Damage , DNA , Epigenesis, Genetic , Ultraviolet Rays , DNA/chemistry , DNA/radiation effects , Epigenesis, Genetic/radiation effects , DNA Damage/radiation effects , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Pyrimidine Dimers/chemistry , Deoxyuridine/analogs & derivatives , Deoxyuridine/chemistry
19.
BMC Plant Biol ; 24(1): 723, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080534

ABSTRACT

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.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Cell Nucleus , DNA Repair , Deoxyribodipyrimidine Photo-Lyase , Plants, Genetically Modified , Arabidopsis/genetics , Arabidopsis/radiation effects , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Deoxyribodipyrimidine Photo-Lyase/metabolism , Deoxyribodipyrimidine Photo-Lyase/genetics , Ultraviolet Rays , DNA, Plant/metabolism , DNA, Plant/genetics , Pyrimidine Dimers/metabolism , Pyrimidine Dimers/genetics , DNA, Chloroplast/genetics , DNA, Chloroplast/metabolism , Chloroplasts/metabolism , DNA Damage
20.
Exp Eye Res ; 243: 109901, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641197

ABSTRACT

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.


Subject(s)
DNA Damage , DNA Repair , Pyrimidine Dimers , Ultraviolet Rays , Xeroderma Pigmentosum , Humans , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/metabolism , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/pathology , Pyrimidine Dimers/metabolism , Keratoplasty, Penetrating , Cornea/metabolism , Cornea/pathology , Cornea/radiation effects , Female , Adult , Histones/metabolism , Male , Middle Aged , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Adolescent , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL