Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 51(10): 4959-4981, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37021581

ABSTRACT

Apurinic/apyrimidinic (AP) sites are DNA lesions created under normal growth conditions that result in cytotoxicity, replication-blocks, and mutations. AP sites are susceptible to ß-elimination and are liable to be converted to DNA strand breaks. HMCES (5-hydroxymethylcytosine binding, ES cell specific) protein interacts with AP sites in single stranded (ss) DNA exposed at DNA replication forks to generate a stable thiazolidine protein-DNA crosslink and protect cells against AP site toxicity. The crosslinked HMCES is resolved by proteasome-mediated degradation; however, it is unclear how HMCES-crosslinked ssDNA and the resulting proteasome-degraded HMCES adducts are processed and repaired. Here, we describe methods for the preparation of thiazolidine adduct-containing oligonucleotides and determination of their structure. We demonstrate that the HMCES-crosslink is a strong replication blocking adduct and that protease-digested HMCES adducts block DNA replication to a similar extent as AP sites. Moreover, we show that the human AP endonuclease APE1 incises DNA 5' to the protease-digested HMCES adduct. Interestingly, while HMCES-ssDNA crosslinks are stable, the crosslink is reversed upon the formation of dsDNA, possibly due to a catalytic reverse reaction. Our results shed new light on damage tolerance and repair pathways for HMCES-DNA crosslinks in human cells.


Subject(s)
DNA Adducts , DNA Repair , Humans , Thiazolidines , Proteasome Endopeptidase Complex/metabolism , DNA/chemistry , DNA Damage , DNA, Single-Stranded/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism
2.
Life Sci Alliance ; 5(12)2022 07 29.
Article in English | MEDLINE | ID: mdl-35905994

ABSTRACT

DNA damage tolerance pathways are regulated by proliferating cell nuclear antigen (PCNA) modifications at lysine 164. Translesion DNA synthesis by DNA polymerase η (Polη) is well studied, but less is known about Polη-independent mechanisms. Illudin S and its derivatives induce alkyl DNA adducts, which are repaired by transcription-coupled nucleotide excision repair (TC-NER). We demonstrate that in addition to TC-NER, PCNA modification at K164 plays an essential role in cellular resistance to these compounds by overcoming replication blockages, with no requirement for Polη. Polκ and RING finger and WD repeat domain 3 (RFWD3) contribute to tolerance, and are both dependent on PCNA modifications. Although RFWD3 is a FANC protein, we demonstrate that it plays a role in DNA damage tolerance independent of the FANC pathway. Finally, we demonstrate that RFWD3-mediated cellular survival after UV irradiation is dependent on PCNA modifications but is independent of Polη. Thus, RFWD3 contributes to PCNA modification-dependent DNA damage tolerance in addition to translesion DNA polymerases.


Subject(s)
DNA Damage , DNA-Directed DNA Polymerase , DNA Repair , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism
3.
J Biol Chem ; 294(11): 4177-4187, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30647135

ABSTRACT

Ubiquitin-specific protease 7 (USP7) regulates various cellular pathways through its deubiquitination activity. Despite the identification of a growing number of substrates of USP7, the molecular mechanism by which USP7 removes ubiquitin chains from polyubiquitinated substrates remains unexplored. The present study investigated the mechanism underlying the deubiquitination of Lys63-linked polyubiquitinated proliferating cell nuclear antigen (PCNA). Biochemical analyses demonstrated that USP7 efficiently removes polyubiquitin chains from polyubiquitinated PCNA by preferential cleavage of the PCNA-ubiquitin linkage. This property was largely attributed to the poor activity toward Lys63-linked ubiquitin chains. The preferential cleavage of substrate-ubiquitin linkages was also observed for Lys48-linked polyubiquitinated p53 because of the inefficient cleavage of the Lys48-linked ubiquitin chains. The present findings suggest a mechanism underlying the removal of polyubiquitin signals by USP7.


Subject(s)
Proliferating Cell Nuclear Antigen/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitins/metabolism , Humans , Substrate Specificity
4.
Nucleic Acids Res ; 46(21): 11340-11356, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30335157

ABSTRACT

DNA-damage tolerance protects cells via at least two sub-pathways regulated by proliferating cell nuclear antigen (PCNA) ubiquitination in eukaryotes: translesion DNA synthesis (TLS) and template switching (TS), which are stimulated by mono- and polyubiquitination, respectively. However, how cells choose between the two pathways remains unclear. The regulation of ubiquitin ligases catalyzing polyubiquitination, such as helicase-like transcription factor (HLTF), could play a role in the choice of pathway. Here, we demonstrate that the ligase activity of HLTF is stimulated by double-stranded DNA via HIRAN domain-dependent recruitment to stalled primer ends. Replication factor C (RFC) and PCNA located at primer ends, however, suppress en bloc polyubiquitination in the complex, redirecting toward sequential chain elongation. When PCNA in the complex is monoubiquitinated by RAD6-RAD18, the resulting ubiquitin moiety is immediately polyubiquitinated by coexisting HLTF, indicating a coupling reaction between mono- and polyubiquitination. By contrast, when PCNA was monoubiquitinated in the absence of HLTF, it was not polyubiquitinated by subsequently recruited HLTF unless all three-subunits of PCNA were monoubiquitinated, indicating that the uncoupling reaction specifically occurs on three-subunit-monoubiquitinated PCNA. We discuss the physiological relevance of the different modes of the polyubiquitination to the choice of cells between TLS and TS under different conditions.


Subject(s)
DNA Repair , DNA-Binding Proteins/genetics , DNA/genetics , Proliferating Cell Nuclear Antigen/genetics , Protein Processing, Post-Translational , Replication Protein C/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , DNA/metabolism , DNA Damage , DNA Primers/chemistry , DNA Primers/metabolism , DNA-Binding Proteins/metabolism , Humans , Polyubiquitin/genetics , Polyubiquitin/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Replication Protein C/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Transcription Factors/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
5.
Mutat Res ; 803-805: 82-88, 2017 10.
Article in English | MEDLINE | ID: mdl-28666590

ABSTRACT

DNA damage tolerance pathways, which include translesion DNA synthesis (TLS) and template switching, are crucial for prevention of DNA replication arrest and maintenance of genomic stability. However, these pathways utilize error-prone DNA polymerases or template exchange between sister DNA strands, and consequently have the potential to induce mutations or chromosomal rearrangements. Post-translational modifications of proliferating cell nuclear antigen (PCNA) play important roles in controlling these pathways. For example, TLS is mediated by mono-ubiquitination of PCNA at lysine 164, for which RAD6-RAD18 is the primary E2-E3 complex. Elaborate protein-protein interactions between mono-ubiquitinated PCNA and Y-family DNA polymerases constitute the core of the TLS regulatory system, and enhancers of PCNA mono-ubiquitination and de-ubiquitinating enzymes finely regulate TLS and suppress TLS-mediated mutagenesis. The template switching pathway is promoted by K63-linked poly-ubiquitination of PCNA at lysine 164. Poly-ubiquitination is achieved by a coupled reaction mediated by two sets of E2-E3 complexes, RAD6-RAD18 and MMS2-UBC13-HTLF/SHPRH. In addition to these mono- and poly-ubiquitinations, simultaneous mono-ubiquitinations on multiple units of the PCNA homotrimeric ring promote an unidentified damage tolerance mechanism that remains to be fully characterized. Furthermore, SUMOylation of PCNA in mammalian cells can negatively regulate recombination. Other modifications, including ISGylation, acetylation, methylation, or phosphorylation, may also play roles in DNA damage tolerance and control of genomic stability.


Subject(s)
DNA Damage , Proliferating Cell Nuclear Antigen/genetics , Protein Processing, Post-Translational , Animals , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Genomic Instability , Humans , Ligases/genetics , Ligases/metabolism , Mutagenesis , Proliferating Cell Nuclear Antigen/metabolism , Sumoylation , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
6.
Cell Rep ; 13(10): 2072-80, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26673319

ABSTRACT

Mono-ubiquitinated PCNA activates error-prone DNA polymerases; therefore, strict regulation of PCNA mono-ubiquitination is crucial in avoiding undesired mutagenesis. In this study, we used an in vitro assay system to identify USP7 as a deubiquitinating enzyme of mono-ubiquitinated PCNA. Suppression of USP1, a previously identified PCNA deubiquitinase, or USP7 increased UV- and H2O2-induced PCNA mono-ubiquitination in a distinct and additive manner, suggesting that USP1 and USP7 make different contributions to PCNA deubiquitination in human cells. Cell-cycle-synchronization analyses revealed that USP7 suppression increased H2O2-induced PCNA ubiquitination throughout interphase, whereas USP1 suppression specifically increased ubiquitination in S-phase cells. UV-induced mutagenesis was elevated in USP1-suppressed cells, whereas H2O2-induced mutagenesis was elevated in USP7-suppressed cells. These results suggest that USP1 suppresses UV-induced mutations produced in a manner involving DNA replication, whereas USP7 suppresses H2O2-induced mutagenesis involving cell-cycle-independent processes such as DNA repair.


Subject(s)
Mutagenesis/physiology , Oxidative Stress/physiology , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin Thiolesterase/metabolism , Cell Line , DNA Damage/physiology , Flow Cytometry , Humans , Hydrogen Peroxide/toxicity , Mutagenesis, Site-Directed , Polymerase Chain Reaction , RNA, Small Interfering , Transfection , Ubiquitin-Specific Peptidase 7 , Ubiquitination
7.
Nucleic Acids Res ; 43(16): 7898-910, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26170230

ABSTRACT

Translesion DNA synthesis (TLS) by the Y-family DNA polymerases Polη, Polι and Polκ, mediated via interaction with proliferating cell nuclear antigen (PCNA), is a crucial pathway that protects human cells against DNA damage. We report that Polη has three PCNA-interacting protein (PIP) boxes (PIP1, 2, 3) that contribute differentially to two distinct functions, stimulation of DNA synthesis and promotion of PCNA ubiquitination. The latter function is strongly associated with formation of nuclear Polη foci, which co-localize with PCNA. We also show that Polκ has two functionally distinct PIP boxes, like Polη, whereas Polι has a single PIP box involved in stimulation of DNA synthesis. All three polymerases were additionally stimulated by mono-ubiquitinated PCNA in vitro. The three PIP boxes and a ubiquitin-binding zinc-finger of Polη exert redundant and additive effects in vivo via distinct molecular mechanisms. These findings provide an integrated picture of the orchestration of TLS polymerases.


Subject(s)
DNA-Directed DNA Polymerase/chemistry , DNA/biosynthesis , Proliferating Cell Nuclear Antigen/metabolism , Amino Acid Motifs , Cell Line , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Humans , Mutation , Protein Interaction Domains and Motifs , Ubiquitination , DNA Polymerase iota
8.
DNA Repair (Amst) ; 29: 139-46, 2015 May.
Article in English | MEDLINE | ID: mdl-25733082

ABSTRACT

Xeroderma pigmentosum variant (XP-V) is a human rare inherited recessive disease, predisposed to sunlight-induced skin cancer, which is caused by deficiency in DNA polymerase η (Polη). Polη catalyzes accurate translesion synthesis (TLS) past pyrimidine dimers, the most prominent UV-induced lesions. DNA polymerase ι (Polι) is a paralog of Polη that has been suggested to participate in TLS past UV-induced lesions, but its function in vivo remains uncertain. We have previously reported that Polη-deficient and Polη/Polι double-deficient mice showed increased susceptibility to UV-induced carcinogenesis. Here, we investigated UV-induced mutation frequencies and spectra in the epidermal cells of Polη- and/or Polι-deficient mice. While Polη-deficient mice showed significantly higher UV-induced mutation frequencies than wild-type mice, Polι deficiency did not influence the frequencies in the presence of Polη. Interestingly, the frequencies in Polη/Polι double-deficient mice were statistically lower than those in Polη-deficient mice, although they were still higher than those of wild-type mice. Sequence analysis revealed that most of the UV-induced mutations in Polη-deficient and Polη/Polι double-deficient mice were base substitutions at dipyrimidine sites. An increase in UV-induced mutations at both G:C and A:T pairs associated with Polη deficiency suggests that Polη contributes to accurate TLS past both thymine- and cytosine-containing dimers in vivo. A significant decrease in G:C to A:T transition in Polη/Polι double-deficient mice when compared with Polη-deficient mice suggests that Polι is involved in error-prone TLS past cytosine-containing dimers when Polη is inactivated.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , Epidermis/metabolism , Pyrimidine Dimers/metabolism , Animals , DNA/metabolism , DNA/radiation effects , DNA Damage , DNA Repair , DNA Replication , DNA-Directed DNA Polymerase/genetics , Epidermal Cells , Epidermis/radiation effects , Mice , Mice, Knockout , Mutation , Ultraviolet Rays , DNA Polymerase iota
9.
PLoS One ; 10(2): e0118775, 2015.
Article in English | MEDLINE | ID: mdl-25692884

ABSTRACT

DNA damage tolerance (DDT) pathways, including translesion synthesis (TLS) and additional unknown mechanisms, enable recovery from replication arrest at DNA lesions. DDT pathways are regulated by post-translational modifications of proliferating cell nuclear antigen (PCNA) at its K164 residue. In particular, mono-ubiquitination by the ubiquitin ligase RAD18 is crucial for Polη-mediated TLS. Although the importance of modifications of PCNA to DDT pathways is well known, the relevance of its homo-trimer form, in which three K164 residues are present in a single ring, remains to be elucidated. Here, we show that multiple units of a PCNA homo-trimer are simultaneously mono-ubiquitinated in vitro and in vivo. RAD18 catalyzed sequential mono-ubiquitinations of multiple units of a PCNA homo-trimer in a reconstituted system. Exogenous PCNA formed hetero-trimers with endogenous PCNA in WI38VA13 cell transformants. When K164R-mutated PCNA was expressed in these cells at levels that depleted endogenous PCNA homo-trimers, multiple modifications of PCNA complexes were reduced and the cells showed defects in DDT after UV irradiation. Notably, ectopic expression of mutant PCNA increased the UV sensitivities of Polη-proficient, Polη-deficient, and REV1-depleted cells, suggesting the disruption of a DDT pathway distinct from the Polη- and REV1-mediated pathways. These results suggest that simultaneous modifications of multiple units of a PCNA homo-trimer are required for a certain DDT pathway in human cells.


Subject(s)
DNA-Binding Proteins/metabolism , Lysine/genetics , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/genetics , Cell Line , DNA Damage , DNA Repair/radiation effects , DNA-Directed DNA Polymerase/metabolism , Humans , In Vitro Techniques , Male , Mutation , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin-Protein Ligases , Ubiquitination
10.
Sci Rep ; 4: 5220, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24910358

ABSTRACT

The genetic information encoded in genomes must be faithfully replicated and transmitted to daughter cells. The recent discovery of consecutive DNA conversions by TET family proteins of 5-methylcytosine into 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine (5caC) suggests these modified cytosines act as DNA lesions, which could threaten genome integrity. Here, we have shown that although 5caC pairs with guanine during DNA replication in vitro, G·5caC pairs stimulated DNA polymerase exonuclease activity and were recognized by the mismatch repair (MMR) proteins. Knockdown of thymine DNA glycosylase increased 5caC in genome, affected cell proliferation via MMR, indicating MMR is a novel reader for 5caC. These results suggest the epigenetic modification products of 5caC behave as DNA lesions.


Subject(s)
Base Pairing/genetics , Cytosine/analogs & derivatives , DNA Mismatch Repair/genetics , DNA Replication/genetics , Guanine/metabolism , 5-Methylcytosine/metabolism , Cell Proliferation/genetics , Cytosine/metabolism , DNA/genetics , Epigenesis, Genetic/genetics , Humans , Thymine DNA Glycosylase/metabolism
11.
Biochem Biophys Res Commun ; 389(1): 40-5, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19703417

ABSTRACT

Human DNA polymerase eta (Poleta) is the gene product underlying xeroderma pigmentosum variant, and plays principal roles in translesion DNA synthesis. Here, we identified human MLH1, an essential component of mismatch repair (MMR), as a Poleta-interacting protein. The middle area residues, which include the little finger domain, of Poleta are important for the interaction with MLH1. Poleta also interacts with the MLH1/PMS2 heterodimer (MutLalpha). Co-immunoprecipitation analyses revealed that MutLalpha, and also MSH2 and MSH6, components of the MutSalpha heterodimer, form complexes with Poleta in human cells. Although MutSalpha had been reported to interact with C-terminal residues of Poleta, MutLalpha and MutSalpha co-precipitated with C-terminally truncated Poleta, suggesting that MutSalpha can interact with Poleta through MutLalpha. MMR proteins were more abundant in the Poleta complex on the chromatin of S phase-synchronized cells than of asynchronous cells, suggesting that the interaction between Poleta and MLH1 is involved in DNA replication.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphatases/metabolism , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenosine Triphosphatases/genetics , Cell Line , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Humans , Mismatch Repair Endonuclease PMS2 , MutL Protein Homolog 1 , MutL Proteins , Nuclear Proteins/genetics , Protein Structure, Tertiary , Two-Hybrid System Techniques
12.
Mol Cell Biol ; 26(20): 7696-706, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17015482

ABSTRACT

DNA polymerase eta (Pol eta) is the product of the Polh gene, which is responsible for the group variant of xeroderma pigmentosum, a rare inherited recessive disease which is characterized by susceptibility to sunlight-induced skin cancer. We recently reported in a study of Polh mutant mice that Pol eta is involved in the somatic hypermutation of immunoglobulin genes, but the cancer predisposition of Polh-/- mice has not been examined until very recently. Another translesion synthesis polymerase, Pol iota, a Pol eta paralog encoded by the Poli gene, is naturally deficient in the 129 mouse strain, and the function of Pol iota is enigmatic. Here, we generated Polh Poli double-deficient mice and compared the tumor susceptibility of them with Polh- or Poli-deficient animals under the same genetic background. While Pol iota deficiency does not influence the UV sensitivity of mouse fibroblasts irrespective of Polh genotype, Polh Poli double-deficient mice show slightly earlier onset of skin tumor formation. Intriguingly, histological diagnosis after chronic treatment with UV light reveals that Pol iota deficiency leads to the formation of mesenchymal tumors, such as sarcomas, that are not observed in Polh(-/-) mice. These results suggest the involvement of the Pol eta and Pol iota proteins in UV-induced skin carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/radiation effects , DNA-Directed DNA Polymerase/deficiency , Epithelial Cells/enzymology , Epithelial Cells/radiation effects , Mesoderm/pathology , Skin Neoplasms/enzymology , Alleles , Animals , Cell Transformation, Neoplastic/pathology , Cells, Cultured , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Epithelial Cells/pathology , Fibroblasts , Mesoderm/enzymology , Mice , Mice, Knockout , Mutation/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , DNA Polymerase iota
SELECTION OF CITATIONS
SEARCH DETAIL
...