Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell ; 82(7): 1343-1358.e8, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35271816

ABSTRACT

Nucleotide excision repair (NER) counteracts the onset of cancer and aging by removing helix-distorting DNA lesions via a "cut-and-patch"-type reaction. The regulatory mechanisms that drive NER through its successive damage recognition, verification, incision, and gap restoration reaction steps remain elusive. Here, we show that the RAD5-related translocase HLTF facilitates repair through active eviction of incised damaged DNA together with associated repair proteins. Our data show a dual-incision-dependent recruitment of HLTF to the NER incision complex, which is mediated by HLTF's HIRAN domain that binds 3'-OH single-stranded DNA ends. HLTF's translocase motor subsequently promotes the dissociation of the stably damage-bound incision complex together with the incised oligonucleotide, allowing for an efficient PCNA loading and initiation of repair synthesis. Our findings uncover HLTF as an important NER factor that actively evicts DNA damage, thereby providing additional quality control by coordinating the transition between the excision and DNA synthesis steps to safeguard genome integrity.


Subject(s)
DNA Repair , DNA-Binding Proteins , DNA/genetics , DNA/metabolism , DNA Damage , DNA Replication , DNA-Binding Proteins/genetics
2.
Nucleic Acids Res ; 47(8): 4011-4025, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30715484

ABSTRACT

Transcription-coupled nucleotide excision repair (TC-NER) is a dedicated DNA repair pathway that removes transcription-blocking DNA lesions (TBLs). TC-NER is initiated by the recognition of lesion-stalled RNA Polymerase II by the joint action of the TC-NER factors Cockayne Syndrome protein A (CSA), Cockayne Syndrome protein B (CSB) and UV-Stimulated Scaffold Protein A (UVSSA). However, the exact recruitment mechanism of these factors toward TBLs remains elusive. Here, we study the recruitment mechanism of UVSSA using live-cell imaging and show that UVSSA accumulates at TBLs independent of CSA and CSB. Furthermore, using UVSSA deletion mutants, we could separate the CSA interaction function of UVSSA from its DNA damage recruitment activity, which is mediated by the UVSSA VHS and DUF2043 domains, respectively. Quantitative interaction proteomics showed that the Spt16 subunit of the histone chaperone FACT interacts with UVSSA, which is mediated by the DUF2043 domain. Spt16 is recruited to TBLs, independently of UVSSA, to stimulate UVSSA recruitment and TC-NER-mediated repair. Spt16 specifically affects UVSSA, as Spt16 depletion did not affect CSB recruitment, highlighting that different chromatin-modulating factors regulate different reaction steps of the highly orchestrated TC-NER pathway.


Subject(s)
Carrier Proteins/genetics , Cell Cycle Proteins/genetics , DNA Repair , DNA-Binding Proteins/genetics , DNA/genetics , High Mobility Group Proteins/genetics , RNA Polymerase II/genetics , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Cell Line, Tumor , Chromatin/metabolism , Chromatin/ultrastructure , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , High Mobility Group Proteins/metabolism , Humans , Optical Imaging , Osteoblasts/metabolism , Osteoblasts/ultrastructure , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Binding , Protein Domains , Protein Transport , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism
3.
Nucleic Acids Res ; 47(7): 3536-3549, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30698791

ABSTRACT

UV light induces cyclobutane pyrimidine dimers (CPDs) and pyrimidine-pyrimidone (6-4) photoproducts (6-4PPs), which can result in carcinogenesis and aging, if not properly repaired by nucleotide excision repair (NER). Assays to determine DNA damage load and repair rates are invaluable tools for fundamental and clinical NER research. However, most current assays to quantify DNA damage and repair cannot be performed in real time. To overcome this limitation, we made use of the damage recognition characteristics of CPD and 6-4PP photolyases (PLs). Fluorescently-tagged PLs efficiently recognize UV-induced DNA damage without blocking NER activity, and therefore can be used as sensitive live-cell damage sensors. Importantly, FRAP-based assays showed that PLs bind to damaged DNA in a highly sensitive and dose-dependent manner, and can be used to quantify DNA damage load and to determine repair kinetics in real time. Additionally, PLs can instantly reverse DNA damage by 405 nm laser-assisted photo-reactivation during live-cell imaging, opening new possibilities to study lesion-specific NER dynamics and cellular responses to damage removal. Our results show that fluorescently-tagged PLs can be used as a versatile tool to sense, quantify and repair DNA damage, and to study NER kinetics and UV-induced DNA damage response in living cells.


Subject(s)
DNA Damage/genetics , DNA/genetics , Pyrimidine Dimers/genetics , Carcinogenesis/genetics , Carcinogenesis/radiation effects , DNA/radiation effects , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Deoxyribodipyrimidine Photo-Lyase/genetics , Deoxyribodipyrimidine Photo-Lyase/radiation effects , Humans , Pyrimidine Dimers/radiation effects , Ultraviolet Rays/adverse effects
4.
Nat Commun ; 6: 7499, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26151477

ABSTRACT

XPC recognizes UV-induced DNA lesions and initiates their removal by nucleotide excision repair (NER). Damage recognition in NER is tightly controlled by ubiquitin and SUMO modifications. Recent studies have shown that the SUMO-targeted ubiquitin ligase RNF111 promotes K63-linked ubiquitylation of SUMOylated XPC after DNA damage. However, the exact regulatory function of these modifications in vivo remains elusive. Here we show that RNF111 is required for efficient repair of ultraviolet-induced DNA lesions. RNF111-mediated ubiquitylation promotes the release of XPC from damaged DNA after NER initiation, and is needed for stable incorporation of the NER endonucleases XPG and ERCC1/XPF. Our data suggest that RNF111, together with the CRL4(DDB2) ubiquitin ligase complex, is responsible for sequential XPC ubiquitylation, which regulates the recruitment and release of XPC and is crucial for efficient progression of the NER reaction, thereby providing an extra layer of quality control of NER.


Subject(s)
DNA Repair/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Nuclear Proteins/metabolism , SUMO-1 Protein/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , DNA Damage , DNA-Binding Proteins/genetics , Endonucleases/genetics , Endonucleases/metabolism , Humans , Nuclear Proteins/genetics , RNA, Small Interfering , SUMO-1 Protein/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...