Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Bioconjug Chem ; 34(11): 2022-2033, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37861691

ABSTRACT

Modified antibodies have essential roles in analytic, diagnostic, and therapeutic uses, and thus, these antibodies are required to have optimal physical and biological properties. Consequently, the development of methods for site-selective antibody modification is crucial. Herein, we used epitope-based affinity labeling to introduce a Fab region-selective antibody modification method. Although labeling that exploits the high affinity between an antibody and its epitope may appear straightforward, it remains challenging probably because of the loss of target affinity caused by modification around the epitope-binding site. By thoroughly screening the modifying agent structure, reaction conditions, and purification methods, we developed an efficient method for the selective modification of the Fab region of the antibody while maintaining the high affinity for the epitope.


Subject(s)
Antibodies, Monoclonal , Immunoglobulin Fab Fragments , Epitopes/chemistry , Antibodies, Monoclonal/chemistry , Antibody Affinity
2.
Article in English | MEDLINE | ID: mdl-37003653

ABSTRACT

Evidence is accumulating that ultraviolet A (UVA) plays an important role in photo-carcinogenesis. However, the types of DNA damage involved in the resulting mutations remain unclear. Previously, using Drosophila, we found that UVA from light-emitting diode (LED-UVA) induces double-strand breaks in DNA through oxidative damage in an oxidative damage-sensitive (urate-null) strain. Recently, it was proposed that cyclobutane pyrimidine dimers (CPDs), which also are induced by UVA irradiation, might play a significant role in the induction of mutations. In the present study, we investigated whether reactive oxygen species (ROS) and CPDs are produced in larval bodies following LED-UVA irradiation. In addition, we assessed the somatic cell mutation rate in urate-null Drosophila induced by monochromatic UVA irradiation. The production of ROS through LED-UVA irradiation was markedly higher in the urate-null strain than in the wild-type Drosophila. CPDs were detected in the DNA of both of UVA- and UVB-irradiated larvae. The level of CPDs was unexpectedly higher in the wild-type strain than in urate-null flies following UVA irradiation, whereas this parameter was expectedly similar between the urate-null and wild-type Drosophila following UVB irradiation. The somatic cell mutation rate induced by UVA irradiation was higher in the urate-null strain than in the wild-type strain. These results suggest that mutations induced by UVA-specific pathways occur through ROS production, rather than via CPD formation.


Subject(s)
Drosophila , Mutagens , Animals , Reactive Oxygen Species , Drosophila/genetics , Drosophila/metabolism , Larva/genetics , Uric Acid , DNA Damage , Pyrimidine Dimers , Ultraviolet Rays/adverse effects , DNA
3.
Genes Cells ; 27(5): 356-367, 2022 May.
Article in English | MEDLINE | ID: mdl-35238109

ABSTRACT

The structure-specific endonuclease XPF-ERCC1 is a multi-functional heterodimer that participates in a variety of DNA repair mechanisms for maintaining genome integrity. Both subunits contain C-terminal tandem helix-hairpin-helix (HhH2 ) domains, which are necessary for not only their dimerization but also enzymatic activity as well as protein stability. However, the interdependency of both subunits in their nuclear localization remains poorly understood. In this study, we have analyzed the region(s) that affects the subcellular localization of XPF and ERCC1 using various deletion mutants. We first identified the nuclear localization signal (NLS) in XPF, which was essential for its nuclear localization under the ERCC1-free condition, but dispensable in the presence of ERCC1 (probably as XPF-ERCC1 heterodimer). Interestingly, in the NLS-independent and ERCC1-dependent XPF nuclear localization, the physical interaction between XPF and ERCC1 via C-terminal HhH2 domains was not needed. Instead, the amino acid regions 311-469 of XPF and 216-260 of ERCC1 are required for the nuclear localization. Furthermore, we found that the 311-469 region of XPF interacts with ERCC1 in a co-immunoprecipitation assay. These results suggest that the nuclear localization of XPF-ERCC1 heterodimer is regulated at multiple levels in an interdependent manner.


Subject(s)
DNA Repair , Endonucleases , Endonucleases/chemistry , Endonucleases/genetics , Endonucleases/metabolism
4.
DNA Repair (Amst) ; 113: 103318, 2022 May.
Article in English | MEDLINE | ID: mdl-35325630

ABSTRACT

DNA-damaging anti-cancer drugs are used clinically to induce cell death by causing DNA strand breaks or DNA replication stress. Camptothecin (CPT) and cisplatin are commonly used anti-cancer drugs, and their combined use enhances the anti-tumour effects. However, the mechanism underlying this enhanced effect has not been well studied. In this study, we analysed the combined effect of CPT and cisplatin or ultraviolet (UV) and found that CPT suppresses transcription recovery after UV damage and induces the disappearance of the Cockayne syndrome group B (CSB) protein, a transcription-coupled nucleotide excision repair (TC-NER) factor. This CPT-induced disappearance of CSB expression was suppressed by proteasome and transcription inhibitors. Moreover, CSB ubiquitination was detected after CPT treatment in a transcription-dependent manner, suggesting that the transcription stress caused by CPT induces CSB ubiquitination, resulting in CSB undetectability. However, Cockayne syndrome group A (CSA) and CUL4A were not involved in the CPT-induced CSB undetectability, suggesting that CSB ubiquitination caused by CPT is regulated differently from the UV response. However, cisplatin or UV sensitivity was enhanced by CPT even in CSB- or CSA-knockout cells. Furthermore, the excessive CSB expression, which suppressed CSB ubiquitination, did not cancel the combined effect of CPT. These results suggest that CPT blocks the repair of cisplatin or UV-induced DNA damage regardless of TC-NER status. CPT possibly compromised the alternative repair pathways other than TC-NER, leading to the suppression of transcription recovery and enhancement of cell killing.

5.
Proc Natl Acad Sci U S A ; 117(25): 14412-14420, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32513688

ABSTRACT

Nucleotide excision repair (NER) removes helix-destabilizing adducts including ultraviolet (UV) lesions, cyclobutane pyrimidine dimers (CPDs), and pyrimidine (6-4) pyrimidone photoproducts (6-4PPs). In comparison with CPDs, 6-4PPs have greater cytotoxicity and more strongly destabilizing properties of the DNA helix. It is generally believed that NER is the only DNA repair pathway that removes the UV lesions as evidenced by the previous data since no repair of UV lesions was detected in NER-deficient skin fibroblasts. Topoisomerase I (TOP1) constantly creates transient single-strand breaks (SSBs) releasing the torsional stress in genomic duplex DNA. Stalled TOP1-SSB complexes can form near DNA lesions including abasic sites and ribonucleotides embedded in chromosomal DNA. Here we show that base excision repair (BER) increases cellular tolerance to UV independently of NER in cancer cells. UV lesions irreversibly trap stable TOP1-SSB complexes near the UV damage in NER-deficient cells, and the resulting SSBs activate BER. Biochemical experiments show that 6-4PPs efficiently induce stable TOP1-SSB complexes, and the long-patch repair synthesis of BER removes 6-4PPs downstream of the SSB. Furthermore, NER-deficient cancer cell lines remove 6-4PPs within 24 h, but not CPDs, and the removal correlates with TOP1 expression. NER-deficient skin fibroblasts weakly express TOP1 and show no detectable repair of 6-4PPs. Remarkably, the ectopic expression of TOP1 in these fibroblasts led them to completely repair 6-4PPs within 24 h. In conclusion, we reveal a DNA repair pathway initiated by TOP1, which significantly contributes to cellular tolerance to UV-induced lesions particularly in malignant cancer cells overexpressing TOP1.


Subject(s)
DNA Breaks, Single-Stranded/radiation effects , DNA Repair , DNA Topoisomerases, Type I/metabolism , Ultraviolet Rays/adverse effects , CRISPR-Cas Systems/genetics , DNA Polymerase beta/genetics , DNA Polymerase beta/metabolism , Fibroblasts , Gene Knockout Techniques , Humans , MCF-7 Cells , Primary Cell Culture , Skin/cytology , Skin/pathology , Skin/radiation effects , X-ray Repair Cross Complementing Protein 1/genetics , X-ray Repair Cross Complementing Protein 1/metabolism , Xeroderma Pigmentosum/etiology , Xeroderma Pigmentosum/pathology , Xeroderma Pigmentosum Group A Protein/genetics , Xeroderma Pigmentosum Group A Protein/metabolism
6.
Biochem Biophys Res Commun ; 519(1): 204-210, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31493872

ABSTRACT

The ERCC1-XPF heterodimer is a structure-specific endonuclease and plays multiple roles in various DNA repair pathways including nucleotide excision repair and also telomere maintenance. The dimer formation, which is mediated by their C-terminal helix-hairpin-helix regions, is essential for their endonuclease activity as well as the stability of each protein. However, the detailed mechanism of how a cellular level of ERCC1-XPF is regulated still remains elusive. Here, we report the identification of DDB1- and CUL4-associated factor 7 (DCAF7, also known as WDR68/HAN11) as a novel interacting protein of ERCC1-XPF by mass spectrometry after tandem purification. Immunoprecipitation experiments confirmed their interaction and suggested dominant association of DCAF7 with XPF but not ERCC1. Interestingly, siRNA-mediated knockdown of DCAF7, but not DDB1, attenuated the cellular level of ERCC1-XPF, which is partly dependent on proteasome. The depletion of TCP1α, one of components of the molecular chaperon TRiC/CCT known to interact with DCAF7 and promote its folding, also reduced ERCC1-XPF level. Finally, we show that the depletion of DCAF7 causes inefficient repair of UV-induced (6-4) photoproducts, which can be rescued by ectopic overexpression of XPF or ERCC1-XPF. Altogether, our results strongly suggest that DCAF7 is a novel regulator of ERCC1-XPF protein level and cellular nucleotide excision repair activity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Cell Line , Down-Regulation , Humans , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Multimerization
7.
PLoS One ; 14(3): e0213383, 2019.
Article in English | MEDLINE | ID: mdl-30840704

ABSTRACT

Replicative DNA polymerases are frequently stalled at damaged template strands. Stalled replication forks are restored by the DNA damage tolerance (DDT) pathways, error-prone translesion DNA synthesis (TLS) to cope with excessive DNA damage, and error-free template switching (TS) by homologous DNA recombination. PDIP38 (Pol-delta interacting protein of 38 kDa), also called Pol δ-interacting protein 2 (PolDIP2), physically associates with TLS DNA polymerases, polymerase η (Polη), Polλ, and PrimPol, and activates them in vitro. It remains unclear whether PDIP38 promotes TLS in vivo, since no method allows for measuring individual TLS events in mammalian cells. We disrupted the PDIP38 gene, generating PDIP38-/- cells from the chicken DT40 and human TK6 B cell lines. These PDIP38-/- cells did not show a significant sensitivity to either UV or H2O2, a phenotype not seen in any TLS-polymerase-deficient DT40 or TK6 mutants. DT40 provides a unique opportunity of examining individual TLS and TS events by the nucleotide sequence analysis of the immunoglobulin variable (Ig V) gene as the cells continuously diversify Ig V by TLS (non-templated Ig V hypermutation) and TS (Ig gene conversion) during in vitro culture. PDIP38-/- cells showed a shift in Ig V diversification from TLS to TS. We measured the relative usage of TLS and TS in TK6 cells at a chemically synthesized UV damage (CPD) integrated into genomic DNA. The loss of PDIP38 also caused an increase in the relative usage of TS. The number of UV-induced sister chromatid exchanges, TS events associated with crossover, was increased a few times in PDIP38-/- human and chicken cells. Collectively, the loss of PDIP38 consistently causes a shift in DDT from TLS to TS without enhancing cellular sensitivity to DNA damage. We propose that PDIP38 controls the relative usage of TLS and TS increasing usage of TLS without changing the overall capability of DDT.


Subject(s)
DNA Damage , Nuclear Proteins/metabolism , Animals , Avian Proteins/deficiency , Avian Proteins/genetics , Avian Proteins/metabolism , Cell Line , Chickens , DNA/biosynthesis , DNA/genetics , DNA Polymerase beta/deficiency , DNA Polymerase beta/genetics , DNA Polymerase beta/metabolism , DNA Primase/deficiency , DNA Primase/genetics , DNA Primase/metabolism , DNA Repair , DNA Replication , DNA-Directed DNA Polymerase/deficiency , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Gene Knockout Techniques , Genes, Immunoglobulin , Humans , Multifunctional Enzymes/deficiency , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Templates, Genetic
8.
Genes Cells ; 24(4): 284-296, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30762924

ABSTRACT

The multisubunit complex transcription factor IIH (TFIIH) has dual functions in transcriptional initiation and nucleotide excision repair (NER). TFIIH is comprised of two subcomplexes, the core subcomplex (seven subunits) including XPB and XPD helicases and the cyclin-dependent kinase (CDK)-activating kinase (CAK) subcomplex (three subunits) containing CDK7 kinase. Recently, it has been reported that spironolactone, an anti-aldosterone drug, inhibits cellular NER by inducing proteasomal degradation of XPB and potentiates the cytotoxicity of platinum-based drugs in cancer cells, suggesting possible drug repositioning. In this study, we have tried to uncover the mechanism underlying the chemical-induced XPB destabilization. Based on siRNA library screening and subsequent analyses, we identified SCFFBXL18 E3 ligase consisting of Skp1, Cul1, F-box protein FBXL18 and Rbx1 responsible for spironolactone-induced XPB polyubiquitination and degradation. In addition, we showed that CDK7 kinase activity is required for this process. Finally, we found that the Ser90 residue of XPB is essential for the chemical-induced destabilization. These results led us to propose a model that spironolactone may trigger the phosphorylation of XPB at Ser90 by CDK7, which promotes the recognition and polyubiquitination of XPB by SCFFBXL18 for proteasomal degradation.


Subject(s)
Cyclin-Dependent Kinases/metabolism , F-Box Proteins/metabolism , Spironolactone/pharmacology , Transcription Factor TFIIH/metabolism , HEK293 Cells , HeLa Cells , Humans , Proteolysis/drug effects , Cyclin-Dependent Kinase-Activating Kinase
9.
Sci Rep ; 7(1): 13808, 2017 10 23.
Article in English | MEDLINE | ID: mdl-29061988

ABSTRACT

Accumulating evidence indicates that transcription is closely related to DNA damage formation and that the loss of RNA biogenesis factors causes genome instability. However, whether such factors are involved in DNA damage responses remains unclear. We focus here on the RNA helicase Aquarius (AQR), a known R-loop processing factor, and show that its depletion in human cells results in the accumulation of DNA damage during S phase, mediated by R-loop formation. We investigated the involvement of Aquarius in DNA damage responses and found that AQR knockdown decreased DNA damage-induced foci formation of Rad51 and replication protein A, suggesting that Aquarius contributes to homologous recombination (HR)-mediated repair of DNA double-strand breaks (DSBs). Interestingly, the protein level of CtIP, a DSB processing factor, was decreased in AQR-knockdown cells. Exogenous expression of Aquarius partially restored CtIP protein level; however, CtIP overproduction did not rescue defective HR in AQR-knockdown cells. In accordance with these data, Aquarius depletion sensitized cells to genotoxic agents. We propose that Aquarius contributes to the maintenance of genomic stability via regulation of HR by CtIP-dependent and -independent pathways.


Subject(s)
Carrier Proteins/metabolism , DNA Breaks, Double-Stranded , Genomic Instability , Neoplasms/genetics , Nuclear Proteins/metabolism , RNA Helicases/metabolism , Recombinational DNA Repair , Carrier Proteins/genetics , Endodeoxyribonucleases , Humans , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/genetics , RNA Helicases/antagonists & inhibitors , RNA Helicases/genetics , Tumor Cells, Cultured
10.
J Biol Chem ; 289(41): 28730-7, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25164823

ABSTRACT

Histone H2A variant H2AX is phosphorylated at Ser(139) in response to DNA double-strand break (DSB) and single-stranded DNA (ssDNA) formation. UV light dominantly induces pyrimidine photodimers, which are removed from the mammalian genome by nucleotide excision repair (NER). We previously reported that in quiescent G0 phase cells, UV induces ATR-mediated H2AX phosphorylation plausibly caused by persistent ssDNA gap intermediates during NER. In this study, we have found that DSB is also generated following UV irradiation in an NER-dependent manner and contributes to an earlier fraction of UV-induced H2AX phosphorylation. The NER-dependent DSB formation activates ATM kinase and triggers the accumulation of its downstream factors, MRE11, NBS1, and MDC1, at UV-damaged sites. Importantly, ATM-deficient cells exhibited enhanced UV sensitivity under quiescent conditions compared with asynchronously growing conditions. Finally, we show that the NER-dependent H2AX phosphorylation is also observed in murine peripheral T lymphocytes, typical nonproliferating quiescent cells in vivo. These results suggest that in vivo quiescent cells may suffer from NER-mediated secondary DNA damage including ssDNA and DSB.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , Resting Phase, Cell Cycle/radiation effects , Signal Transduction/radiation effects , Adaptor Proteins, Signal Transducing , Animals , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Transformed , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Humans , MRE11 Homologue Protein , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , Primary Cell Culture , Resting Phase, Cell Cycle/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects , Trans-Activators/genetics , Trans-Activators/metabolism , Ultraviolet Rays
11.
Genes Cells ; 19(4): 350-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24520900

ABSTRACT

The ultraviolet B (UVB) component of sunlight can cause severe damage to skin cells and even induce skin cancer. Growing evidence indicates that the UVB-induced signaling network is complex and involves diverse cellular processes. In this study, we investigated the role of c-Jun NH2 -terminal kinase-associated leucine zipper protein (JLP), a scaffold protein for mitogen-activated protein kinase (MAPK) signaling cascades, in UVB-induced apoptosis. We found that UVB-induced skin epidermal apoptosis was prevented in Jlp knockout (KO) as well as in keratinocyte-specific Jlp KO mice. Analysis of the repair of UVB-induced DNA damage over time showed no evidence for the involvement of JLP in this process. In contrast, UVB-stimulated p38 MAPK activation in the skin was impaired in both Jlp KO and keratinocyte-specific Jlp KO mice. Moreover, topical treatment of UVB-irradiated mouse skin with a p38 inhibitor significantly suppressed the epidermal apoptosis in wild-type mice, but not in Jlp KO mice. Our findings suggest that JLP in skin basal keratinocytes plays an important role in UVB-induced apoptosis by modulating p38 MAPK signaling pathways. This is the first study to show a critical role for JLP in an in vivo response to environmental stimulation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/radiation effects , Ultraviolet Rays/adverse effects , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis/drug effects , DNA Fragmentation , Epidermis/drug effects , Epidermis/metabolism , Epidermis/pathology , Epidermis/radiation effects , Imidazoles/pharmacology , Keratinocytes/drug effects , Keratinocytes/pathology , Keratinocytes/radiation effects , MAP Kinase Signaling System , Mice, Inbred C57BL , Mice, Knockout , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Photochem Photobiol ; 88(2): 356-62, 2012.
Article in English | MEDLINE | ID: mdl-22220555

ABSTRACT

DNA photolesions induced by UV, cyclobutane pyrimidine dimer (CPD) and (6-4) photoproduct (6-4PP), are repaired by nucleotide excision repair (NER) in human cells. Various immunoassays using monoclonal antibodies specific for the photolesions have been developed and widely used for the analysis of cellular NER activity. In this study, we have newly developed a microplate-formatted cell-based immunoassay, based on indirect immunofluorescence staining with lesion-specific antibodies combined with an infrared imaging system. Using this assay, we show the repair kinetics of CPD and 6-4PP in various fibroblasts from newborn and adult donors with no age-related difference. Furthermore, epidermal keratinocytes and melanocytes exhibit comparable NER activity, and calcium ion-induced differentiation of keratinocytes has no significant impacts on their NER activity. We also evaluated the effects of a proteasome inhibitor, MG132, and a histone deacetylase inhibitor, sodium butyrate, on NER efficiency using this assay. All these results suggest that the new assay is highly useful for the rapid and quantitative analysis of NER activity in various primary cells with limited growth activity and is applicable to a screening system for drugs affecting NER efficiency.


Subject(s)
DNA Repair , Epidermis/metabolism , Fibroblasts/metabolism , Immunoassay/methods , Keratinocytes/metabolism , Melanocytes/metabolism , Adult , Butyrates/pharmacology , Calcium/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , DNA Damage , DNA Repair/drug effects , DNA Repair/radiation effects , Epidermis/drug effects , Epidermis/radiation effects , Fibroblasts/drug effects , Fibroblasts/radiation effects , Humans , Infant, Newborn , Keratinocytes/drug effects , Keratinocytes/radiation effects , Leupeptins/pharmacology , Melanocytes/drug effects , Melanocytes/radiation effects , Primary Cell Culture , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Pyrimidine Dimers/pharmacology , Spectrophotometry, Infrared , Ultraviolet Rays
13.
Cell ; 147(5): 1024-39, 2011 Nov 23.
Article in English | MEDLINE | ID: mdl-22118460

ABSTRACT

The DDB1-CUL4-RBX1 (CRL4) ubiquitin ligase family regulates a diverse set of cellular pathways through dedicated substrate receptors (DCAFs). The DCAF DDB2 detects UV-induced pyrimidine dimers in the genome and facilitates nucleotide excision repair. We provide the molecular basis for DDB2 receptor-mediated cyclobutane pyrimidine dimer recognition in chromatin. The structures of the fully assembled DDB1-DDB2-CUL4A/B-RBX1 (CRL4(DDB2)) ligases reveal that the mobility of the ligase arm creates a defined ubiquitination zone around the damage, which precludes direct ligase activation by DNA lesions. Instead, the COP9 signalosome (CSN) mediates the CRL4(DDB2) inhibition in a CSN5 independent, nonenzymatic, fashion. In turn, CSN inhibition is relieved upon DNA damage binding to the DDB2 module within CSN-CRL4(DDB2). The Cockayne syndrome A DCAF complex crystal structure shows that CRL4(DCAF(WD40)) ligases share common architectural features. Our data support a general mechanism of ligase activation, which is induced by CSN displacement from CRL4(DCAF) on substrate binding to the DCAF.


Subject(s)
Ubiquitin-Protein Ligases/chemistry , Animals , Crystallography, X-Ray , Cullin Proteins/chemistry , DNA Damage , DNA-Binding Proteins/chemistry , Enzyme Activation , Humans , Models, Molecular , Ubiquitin-Protein Ligases/metabolism , Xeroderma Pigmentosum Group A Protein/chemistry
14.
J Mol Biol ; 413(2): 337-46, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21875596

ABSTRACT

Nucleotide excision repair (NER) is a very important defense system against various types of DNA damage, and it is necessary for maintaining genomic stability. The molecular mechanism of NER has been studied in considerable detail, and it has been shown that proper protein-protein interactions among NER factors are critical for efficient repair. A structure-specific endonuclease, XPF-ERCC1, which makes the 5' incision in NER, was shown to interact with a single-stranded DNA binding protein, RPA. However, the biological significance of this interaction was not studied in detail. We used the yeast two-hybrid assay to determine that XPF interacts with the p70 subunit of RPA. To further examine the role of this XPF-p70 interaction, we isolated a p70-interaction-deficient mutant form of XPF that contains a single amino acid substitution in the N-terminus of XPF by the reverse yeast two-hybrid assay using randomly mutagenized XPF. The biochemical properties of this RPA-interaction-deficient mutant XPF-ERCC1 are very similar to those of wild-type XPF-ERCC1 in vitro. Interestingly, expression of this mutated form of XPF in the XPF-deficient Chinese hamster ovary cell line, UV41, only partially restores NER activity and UV resistance in vivo compared to wild-type XPF. We discovered that the RPA-interaction-deficient XPF is not localized in nuclei and the mislocalization of XPF-ERCC1 prevents the complex from functioning in NER.


Subject(s)
DNA Damage/genetics , DNA Repair , DNA-Binding Proteins/metabolism , Replication Protein A/metabolism , Animals , Cell Fractionation , Cells, Cultured , Cricetinae , Cricetulus , DNA-Binding Proteins/genetics , Humans , Mutagenesis , Replication Protein A/genetics , Two-Hybrid System Techniques
15.
Nucleic Acids Res ; 37(2): 516-25, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19056823

ABSTRACT

Damaged DNA-binding protein (DDB), consisting of DDB1 and DDB2 subunits recognizes a wide spectrum of DNA lesions. DDB is dispensable for in vitro nucleotide excision repair (NER) reaction, but stimulates this reaction especially for cyclobutane pyrimidine dimer (CPD). Here we show that DDB directly interacts with XPA, one of core NER factors, mainly through DDB2 subunit and the amino-acid residues between 185 and 226 in XPA are important for the interaction. Interestingly, the point mutation causing the substitution from Arg-207 to Gly, which was previously identified in a XP-A revertant cell-line XP129, diminished the interaction with DDB in vitro and in vivo. In a defined system containing R207G mutant XPA and other core NER factors, DDB failed to stimulate the excision of CPD, although the mutant XPA was competent for the basal NER reaction. Moreover, in vivo experiments revealed that the mutant XPA is recruited to damaged DNA sites with much less efficiency compared with wild-type XPA and fails to support the enhancement of CPD repair by ectopic expression of DDB2 in SV40-transformed human cells. These results suggest that the physical interaction between DDB and XPA plays an important role in the DDB-mediated NER reaction.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , Xeroderma Pigmentosum Group A Protein/metabolism , Cell Line , Cell Line, Transformed , Cell Nucleus/metabolism , DNA Damage , Humans , Mutation, Missense , Protein Interaction Domains and Motifs , Pyrimidine Dimers/metabolism , Ultraviolet Rays , Xeroderma Pigmentosum Group A Protein/chemistry , Xeroderma Pigmentosum Group A Protein/genetics
16.
J Biol Chem ; 283(37): 25485-25491, 2008 Sep 12.
Article in English | MEDLINE | ID: mdl-18550533

ABSTRACT

The Chk1 kinase is highly conserved from yeast to humans and is well known to function in the cell cycle checkpoint induced by genotoxic or replication stress. The activation of Chk1 is achieved by ATR-dependent phosphorylation with the aid of additional factors. Robust genotoxic insults induce apoptosis instead of the cell cycle checkpoint, and some of the components in the ATR-Chk1 pathway are cleaved by active caspases, although it has been unclear whether the attenuation of the ATR-Chk1 pathway has some role in apoptosis induction. Here we show that Chk1 is activated by caspase-dependent cleavage when the cells undergo apoptosis. Treatment of chicken DT40 cells with various genotoxic agents, UV light, etoposide, or camptothecin induced Chk1 cleavage, which was inhibited by a pan-caspase inhibitor, benzyloxycarbonyl-VAD-fluoromethyl ketone. The cleavage of Chk1 was similarly observed in human Jurkat cells treated with a non-genotoxic apoptosis inducer, staurosporine. We have determined the cleavage site(s), Asp-299 in chicken and Asp-299 and Asp-351 in human cells. We further show that a truncated form of human Chk1 mimicking the N-terminal cleavage fragment (residues 1-299) possesses strikingly elevated kinase activity. Moreover, the ectopic expression of Chk1-(1-299) in human U2OS cells induces abnormal nuclear morphology with localized chromatin condensation and phosphorylation of histone H2AX. These results suggest that Chk1 is activated by caspase-mediated cleavage during apoptosis and might be implicated in enhancing apoptotic reactions rather than attenuating the ATR-Chk1 pathway.


Subject(s)
Apoptosis , Protein Kinases/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Camptothecin/pharmacology , Caspases/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Checkpoint Kinase 1 , Chickens , Cysteine Proteinase Inhibitors/pharmacology , Etoposide/pharmacology , Histones/metabolism , Humans , Jurkat Cells , Models, Biological , Phosphorylation , Protein Serine-Threonine Kinases/metabolism
17.
Biochem Biophys Res Commun ; 364(4): 771-7, 2007 Dec 28.
Article in English | MEDLINE | ID: mdl-17976535

ABSTRACT

DDB1 was originally identified as a heterodimeric complex with DDB2 and plays an accessory role in nucleotide excision repair. DDB1 also constitutes an E3 ubiquitin ligase complex together with Cul4A and Roc1 and acts as an adaptor, suggesting its multiple roles beyond DNA repair. We have generated a conditional DDB1-knockout mutant using a chicken B lymphocyte line DT40. Doxycycline-induced DDB1 depletion caused a severe growth defect followed by apoptotic cell death. Flow cytometric analyses revealed that cell cycle progression is initially retarded at all phases and subsequently impaired at S phase along with the appearance of sub-G1 population. Similarly, DDB1-knockdown in human U2OS cells by small interfering RNA exhibited a loss of clonogenic activity and perturbed cell cycle progression. These results demonstrate that the DDB1 gene is indispensable for cell viability in higher vertebrates and this conditional DDB1-knockout clone would be highly useful for the functional analysis of DDB1.


Subject(s)
Apoptosis , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , DNA-Binding Proteins/genetics , Growth Disorders/genetics , Growth Disorders/metabolism , Animals , Cell Line , Chickens , Gene Silencing
18.
Nucleic Acids Res ; 33(9): 2993-3001, 2005.
Article in English | MEDLINE | ID: mdl-15914671

ABSTRACT

DNA interstrand crosslinks (ICLs) represent a severe form of damage that blocks DNA metabolic processes and can lead to cell death or carcinogenesis. The repair of DNA ICLs in mammals is not well characterized. We have reported previously that a key protein complex of nucleotide excision repair (NER), XPA-RPA, recognizes DNA ICLs. We now report the use of triplex technology to direct a site-specific psoralen ICL to a target DNA substrate to determine whether the human global genome NER damage recognition complex, XPC-hHR23B, recognizes this lesion. Our results demonstrate that XPC-hHR23B recognizes psoralen ICLs, which have a structure fundamentally different from other lesions that XPC-hHR23B is known to bind, with high affinity and specificity. XPC-hHR23B and XPA-RPA protein complexes were also observed to bind psoralen ICLs simultaneously, demonstrating not only that psoralen ICLs are recognized by XPC-hHR23B alone, but also that XPA-RPA may interact cooperatively with XPC-hHR23B on damaged DNA, forming a multimeric complex. Since XPC-hHR23B and XPA-RPA participate in the recognition and verification of DNA damage, these results support the hypothesis that interplay between components of the global genome repair sub-pathway of NER is critical for the recognition of psoralen DNA ICLs in the mammalian genome.


Subject(s)
Cross-Linking Reagents/toxicity , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , DNA/metabolism , Ficusin/toxicity , DNA/chemistry , DNA Repair Enzymes , Humans , Kinetics , Protein Binding , Replication Protein A , Xeroderma Pigmentosum Group A Protein
19.
Biochem Biophys Res Commun ; 321(1): 183-91, 2004 Aug 13.
Article in English | MEDLINE | ID: mdl-15358233

ABSTRACT

Thymine glycol (Tg) is one of predominant oxidative DNA lesions caused by ionizing radiation and other oxidative stresses. Human NTH1 is a bifunctional enzyme with DNA glycosylase and AP lyase activities and removes Tg as the first step of base excision repair (BER). We have searched for the factors interacting with NTH1 by using a pull-down assay and found that GST-NTH1 fusion protein precipitates proliferating cell nuclear antigen (PCNA) and p53 as well as XPG from human cell-free extracts. GST-NTH1 also bound to recombinant FLAG-tagged XPG, PCNA, and (His)6-tagged p53 proteins, indicating direct protein-protein interaction between those proteins. Furthermore, His-p53 and FLAG-XPG, but not PCNA, stimulated the Tg DNA glycosylase/AP lyase activity of GST-NTH1 or NTH1. These results provide an insight into the positive regulation of BER reaction and also suggest a possible linkage between BER of Tg and other cellular mechanisms.


Subject(s)
DNA Repair/physiology , Deoxyribonuclease (Pyrimidine Dimer)/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Tumor Suppressor Protein p53/metabolism , Base Sequence , Cell Line , Cloning, Molecular , DNA Damage , DNA Primers , DNA Repair/genetics , Deoxyribonuclease (Pyrimidine Dimer)/genetics , Escherichia coli/genetics , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Kinetics , Proliferating Cell Nuclear Antigen/genetics , Recombinant Fusion Proteins/metabolism , Restriction Mapping , Tumor Suppressor Protein p53/genetics
20.
Genes Genet Syst ; 78(2): 169-77, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12773817

ABSTRACT

DDB (damaged DNA-binding protein) is a heterodimer, comprised of p48 (DDB2) and p127 (DDB1) subunits, which has a high affinity for a variety of DNA lesions including UV-photoproducts. The mutations in DDB2 gene have been found in a subset of xeroderma pigmentosum complementation group E patients. However, no natural mutation has been identified so far in the cDNA of human DDB1 and the precise roles of DDB1 are still unknown. We have cloned the DDB1 cDNA from the chicken B lymphocyte line DT40 and revealed an open reading frame of 3420 bp encoding a polypeptide of 1140 amino acids, which is identical in size to the orthologs of human, monkey, mouse, rat and Drosophila melanogaster in databases. The amino acid sequence deduced from the chicken DDB1 cDNA shows a high homology to the mammalian DDB1 orthologs (96-97% identity). Northern blot analysis using 5' portion of the chicken DDB1 cDNA as a probe detected a single transcript of ~ 4.3 kb in chicken DT40 cells as well as in human HeLa cells and mouse embryonic fibroblasts. Furthermore, the chicken DDB1 (tagged with enhanced GFP) transiently expressed in human cells mainly localized in the cytoplasm, and coexpression of human DDB2 dramatically changed the localization from the cytoplasm to nucleus. These results suggest that DDB1 is evolutionarily conserved in the primary structure and function, and may play a fundamental role in higher eukaryotes.


Subject(s)
Chickens/genetics , DNA-Binding Proteins/genetics , Amino Acid Sequence , Animals , Chickens/physiology , Cloning, Molecular , Conserved Sequence , DNA Damage , DNA Repair/physiology , DNA, Complementary , DNA-Binding Proteins/physiology , Genes, Reporter , HeLa Cells , Humans , Microscopy, Fluorescence , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...