Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 15(1): 141-58, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24366067

ABSTRACT

Global genome nucleotide excision repair (GG-NER) is responsible for identifying and removing bulky adducts from non-transcribed DNA that result from damaging agents such as UV radiation and cisplatin. Xeroderma pigmentosum complementation group C (XPC) is one of the essential damage recognition proteins of the GG-NER pathway and its dysfunction results in xeroderma pigmentosum (XP), a disorder involving photosensitivity and a predisposition to cancer. To better understand the identification of DNA damage by XPC in the context of chromatin and the role of XPC in the pathogenesis of XP, we characterized the interactome of XPC using a high throughput yeast two-hybrid screening. Our screening showed 49 novel interactors of XPC involved in DNA repair and replication, proteolysis and post-translational modifications, transcription regulation, signal transduction, and metabolism. Importantly, we validated the XPC-OTUD4 interaction by co-IP and provided evidence that OTUD4 knockdown in human cells indeed affects the levels of ubiquitinated XPC, supporting a hypothesis that the OTUD4 deubiquitinase is involved in XPC recycling by cleaving the ubiquitin moiety. This high-throughput characterization of the XPC interactome provides a resource for future exploration and suggests that XPC may have many uncharacterized cellular functions.


Subject(s)
Xeroderma Pigmentosum/metabolism , DNA Repair , HCT116 Cells , Humans , Protein Interaction Maps , RNA Interference , RNA, Small Interfering/metabolism , Sequence Analysis, DNA , Two-Hybrid System Techniques , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitination , Xeroderma Pigmentosum/chemistry
2.
Anal Chem ; 85(14): 6732-9, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23789926

ABSTRACT

Interstrand cross-links (ICLs) are highly toxic DNA lesions that block transcription and replication by preventing strand separation. ICL-inducing agents were among the earliest and are still the most widely used forms of chemotherapeutic drugs. Because of the repair of DNA ICLs, the therapeutic efficacy of the DNA cross-linking agents is often reduced by the development of chemoresistance in patients. Thus, it is very important to understand how various DNA ICLs are repaired. Such studies are currently hampered by the lack of an analytical method for monitoring directly the repair of DNA ICLs in cells. Here we report a high-performance liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) method, together with the isotope dilution technique, for assessing the repair of 8-methoxypsoralen (8-MOP)-induced DNA ICLs, as well as monoadducts (MAs), in cultured mammalian cells. We found that, while there were substantial decreases in the levels of ICL and MAs in repair-competent cells 24 h after 8-MOP/UVA treatment, there was little repair of 8-MOP-ICLs and -MAs in xeroderma pigmentosum, complementation group A-deficient human skin fibroblasts and excision repair cross-complementing rodent repair deficiency, complementation group 1-deficient Chinese hamster ovary cells over a 24 h period. This result provided unequivocal evidence supporting the notion that the 8-MOP photoadducts are substrates for nucleotide excision repair in mammalian cells. This is one of the first few reports about the application of LC-MS/MS for assessing the repair of DNA ICLs. The analytical method developed here, when combined with genetic manipulation, will also facilitate the assessment of the roles of other DNA repair pathways in removing these DNA lesions, and the method can also be generally applicable for investigating the repair of other types of DNA ICLs in mammalian cells.


Subject(s)
Cross-Linking Reagents/analysis , DNA Adducts/analysis , Methoxsalen/analysis , Tandem Mass Spectrometry/methods , Animals , CHO Cells , Cells, Cultured , Chromatography, Liquid/methods , Cricetinae , Cricetulus , Cross-Linking Reagents/chemistry , DNA Adducts/chemistry , HEK293 Cells , Humans , Mass Spectrometry/methods , Methoxsalen/chemistry , Xeroderma Pigmentosum/chemistry
3.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 2): 237-46, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23385459

ABSTRACT

XPB is a DNA-dependent helicase and a subunit of the TFIIH complex required for both transcription and DNA repair. XPB contains four domains: an N-terminal domain, two conserved helicase domains (HD1 and HD2) and a C-terminal extension. The C-terminal extension is important for DNA repair since the phosphorylation of Ser751 inhibits 5'-incision by ERCC1-XPF endonuclease. A disease-causing frameshift mutation (XP11BE) that changes the last 42 amino acids of XPB causes manifestations including impaired DNA repair and deficient transcription. Here, the crystal structure of the C-terminal half of XPB (residues 494-782) is reported at 1.8 Šresolution. The structure contained the conserved XPB HD2 and a C-terminal extension which shares structural similarity with RIG-I, leading to a structural model of the XPF-XPB-DNA complex for 5' incision during DNA repair. A mutation mimicking the XP11BE mutation produced the much less soluble mutant XPBm(494-781). Western blotting results confirmed that the intracellular levels of XPB and other TFIIH subunits in XP11BE patient cells were much lower than those from the healthy parents. Together, these results indicate that the XP11BE mutation not only divests the XPF-interaction motif, impairing DNA repair, but also reduces XPB solubility, leading to a lower intracellular level of TFIIH and deficient transcription.


Subject(s)
DNA Helicases/chemistry , DNA Repair/genetics , Frameshift Mutation , Peptide Fragments/chemistry , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/genetics , Cells, Cultured , Crystallization , DEAD Box Protein 58 , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/genetics , DNA Helicases/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Female , Humans , Male , Peptide Fragments/genetics , Protein Structure, Tertiary/genetics , Receptors, Immunologic , Xeroderma Pigmentosum/chemistry
4.
Photochem Photobiol ; 89(3): 649-54, 2013.
Article in English | MEDLINE | ID: mdl-23331297

ABSTRACT

The UVA is currently thought to be carcinogenic because, similar to UVB, it induces the formation of cyclobutane pyrimidine dimers (CPDs). Various drugs have been reported to cause photosensitive drug eruptions as an adverse effect. Although the precise mechanism of photosensitive drug eruption remains to be elucidated, it is generally accepted that free radicals and other reactive molecules generated via UV-irradiated drugs play important roles in the pathogenesis of photosensitive drug eruptions. The waveband of concern for photo-reactive drugs is UVA-visible light, but some extend into the UVB region. We tested whether photosensitive drugs could enhance CPD formation after UVA exposure by using isolated DNA in the presence of several reported photosensitive drugs using high-performance liquid chromatography. We found that the diuretic agent hydrochlorothiazide (HCT) significantly enhanced the production of TT dimers over a wide range of UVA. Furthermore, we investigated whether UVA plus HCT could enhance CPD production in xeroderma pigmentosum model mice defective in nucleotide excision repair. Immunofluorescence studies showed that CPD formation in the skin significantly increased after 365 nm narrow-band UVA irradiation in the presence of HCT, compared with that in wild-type mice. HCT could be used with caution because of its enhancement of UVA-induced DNA damage.


Subject(s)
DNA Repair/genetics , DNA/chemistry , Diuretics/adverse effects , Hydrochlorothiazide/adverse effects , Photosensitizing Agents/adverse effects , Pyrimidine Dimers/biosynthesis , Skin/drug effects , Xeroderma Pigmentosum/chemistry , Animals , DNA/metabolism , DNA Damage , Disease Models, Animal , Diuretics/chemistry , Hydrochlorothiazide/chemistry , Mice , Mice, Knockout , Photosensitizing Agents/chemistry , Pyrimidine Dimers/chemistry , Skin/chemistry , Skin/pathology , Skin/radiation effects , Ultraviolet Rays/adverse effects , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/pathology
5.
Am J Dermatopathol ; 35(1): 45-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22722466

ABSTRACT

Xeroderma pigmentosum (XP) is a heterogenous group of genetic diseases in which basal cell carcinoma (BCC) is the most common nonmelanoma skin cancer (NMSC) followed by squamous cell carcinoma (SCC). The aim of this study was to investigate the expression of matrix metalloproteinase (MMP)-13 and Ki-67 in SCC and BCC from patients with and without XP to elucidate their roles in the pathogenesis of these highly aggressive tumors in patients with XP. Immunolabeling using MMP-13 and Ki-67 antibodies was performed on tissue sections derived from skin biopsies of SCC and BCC of 15 patients with XP and 40 non-XP patients. There was no significant difference between XP and non-XP patients as regards MMP-13 expression by epithelial and stromal cells of SCC or BCC. Ki-67 expression in SCC and BCC of patients with XP was significantly higher than in non-XP patients. We concluded that the higher expression of Ki-67 in NMSC of patients with XP than of non-XP patients may reflect the growth and invasive capacity of these tumors in patients with XP. MMP-13 is expressed by tumor epithelial cells, stromal and inflammatory cells of NMSC of both XP and non-XP patients.


Subject(s)
Carcinoma, Basal Cell/chemistry , Carcinoma, Squamous Cell/chemistry , Ki-67 Antigen/analysis , Matrix Metalloproteinase 13/analysis , Skin Neoplasms/chemistry , Xeroderma Pigmentosum/chemistry , Adolescent , Biopsy , Carcinoma, Basal Cell/enzymology , Carcinoma, Basal Cell/immunology , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Child , Child, Preschool , Epithelial Cells/chemistry , Female , Humans , Immunohistochemistry , Male , Neoplasm Invasiveness , Skin Neoplasms/enzymology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Stromal Cells/chemistry , Xeroderma Pigmentosum/enzymology , Xeroderma Pigmentosum/immunology , Xeroderma Pigmentosum/pathology , Young Adult
6.
Biochemistry ; 45(50): 14965-79, 2006 Dec 19.
Article in English | MEDLINE | ID: mdl-17154534

ABSTRACT

XPC is a 940-residue multidomain protein critical for the sensing of aberrant DNA and initiation of global genome nucleotide excision repair. The C-terminal portion of XPC (residues 492-940; XPC-C) has critical interactions with DNA, RAD23B, CETN2, and TFIIH, whereas functional roles have not yet been assigned to the N-terminal portion (residues 1-491; XPC-N). In order to analyze the molecular basis for XPC function and mutational defects associated with xeroderma pigmentosum (XP) disease, a series of stable bacterially expressed N- and C-terminal fragments were designed on the basis of sequence analysis and produced for biochemical characterization. Limited proteolysis experiments combined with mass spectrometry revealed that the full XPC-C is stable but XPC-N is not. However, a previously unrecognized folded helical structural domain was found within XPC-N, XPC(156-325). Pull-down and protease protection assays demonstrated that XPC(156-325) physically interacts with the DNA repair factor XPA, establishing the first functional role for XPC-N. XPC-C exhibits binding characteristics of the full-length protein, including stimulation of DNA binding by physical interaction with RAD23B and CETN2. Analysis of an XPC missense mutation (Trp690Ser) found in certain patients with XP disease revealed that this mutation is associated with a diminished ability to bind DNA. Evidence of contributions to protein interactions from regions in both XPC-N and XPC-C along with recently recognized homologies to yeast PNGase prompted construction of a structural model of a folded XPC core. This model offers key insights into how domains from the two portions of the protein may cooperate in generating specific XPC functions.


Subject(s)
DNA-Binding Proteins/chemistry , DNA/chemistry , Mutation, Missense , Transcription Factors/chemistry , Binding Sites/genetics , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Xeroderma Pigmentosum/chemistry , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum/metabolism
7.
Biochemistry ; 45(11): 3653-63, 2006 Mar 21.
Article in English | MEDLINE | ID: mdl-16533048

ABSTRACT

Human centrin 2 is a component of the nucleotide excision repair system, as a subunit of the heterotrimer including xeroderma pigmentosum group C protein (XPC) and hHR23B. The C-terminal domain of centrin (C-HsCen2) binds strongly a peptide from the XPC protein (P1-XPC: N(847)-R(863)). Here, we characterize the solution Ca(2+)-dependent structural and molecular features of the C-HsCen2 in complex with P1-XPC, mainly using NMR spectroscopy and molecular modeling. The N-terminal half of the peptide, organized as an alpha helix is anchored into a deep hydrophobic cavity of the protein, because of three bulky hydrophobic residues in position 1-4-8 and electrostatic contacts with the centrin helix E. Investigation of the whole centrin interactions shows that the N-terminal domain of the protein is not involved in the complex formation and is structurally independent from the peptide-bound C-terminal domain. The complex may exist in three different binding conformations corresponding to zero, one, and two Ca(2+)-bound states, which may exchange with various rates and have distinct structural stability. The various features of the intermolecular interaction presented here constitute a centrin-specific mode for the target binding.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , Xeroderma Pigmentosum/metabolism , Amino Acid Motifs , Amino Acid Sequence , Calcium/metabolism , Calcium-Binding Proteins/chemistry , Cell Cycle Proteins/chemistry , DNA-Binding Proteins/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Temperature , Xeroderma Pigmentosum/chemistry
8.
Nucleic Acids Res ; 24(13): 2551-9, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8692695

ABSTRACT

The xeroderma pigmentosum syndrome complementation group C (XP-C) is due to a defect in the global genome repair subpathway of nucleotide excision repair (NER). The XPC protein is complexed with HHR23B, one of the two human homologs of the yeast NER protein, RAD23 (Masutani at al. (1994) EMBO J. 8, 1831-1843). Using heparin chromatography, gel filtration and native gel electrophoresis we demonstrate that the majority of HHR23B is in a free, non-complexed form, and that a minor fraction is tightly associated with XPC. In contrast, we cannot detect any bound HHR23A. Thus the HHR23 proteins may have an additional function independent of XPC. The fractionation behaviour suggests that the non-bound forms of the HHR23 proteins are not necessary for the core of the NER reaction. Although both HHR23 proteins share a high level of overall homology, they migrate very differently on native gels, pointing to a difference in conformation. Gel filtration suggests the XPC-HHR23B heterodimer resides in a high MW complex. However, immunodepletion studies starting from repair-competent Manley extracts fall to reveal a stable association of a significant fraction of the HHR23 proteins or the XPC-HHR23B complex with the basal transcription/repair factor TFIIH, or with the ERCC1 repair complex. Consistent with a function in repair or DNA/chromatin metabolism, immunofluorescence studies show all XPC, HHR23B and (the free) HHR23A to reside in the nucleus.


Subject(s)
DNA Repair , DNA-Binding Proteins/isolation & purification , Xeroderma Pigmentosum/chemistry , Amino Acid Sequence , Animals , CHO Cells , Cell Compartmentation , Cell Nucleus/chemistry , Cricetinae , DNA Repair Enzymes , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , HeLa Cells , Humans , Molecular Sequence Data , Protein Binding , Recombinant Proteins/isolation & purification
9.
J Invest Dermatol ; 99(5): 634-8, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1431227

ABSTRACT

In mammalian cells, 72-kD heat shock protein (HSP72) is the major stress-inducible protein that is thought to play a protective role against the various environmental stresses. In order to know the induction mechanism of HSP72, we examined the HSP72 in DNA repair-deficient xeroderma pigmentosum group A fibroblasts (XP2OSSV) and normal fibroblasts (WI38VA13) by the indirect immunofluorescence method using a monoclonal antibody specific for the inducible 72-kD protein. Heat-shock treatment of the same survival fraction (5% survival) induced HSP72 in xeroderma pigmentosum (XP) and normal cells. However, as compared with XP cells, normal cells showed the induction of HSP72 more rapidly and strongly. When XP and normal cells were irradiated with UVC at the same survival dose (10% survival), apparent induction of HSP72 was observed in both cell lines. In the case of UVC irradiation at the same dose (1.0 J/m2), though XP cells showed the induction of HSP72, HSP72 was not induced in normal cells. In both cell lines, heat-shock treatment caused more rapid induction of HSP72 than UV irradiation. These results suggest that the induction mechanism of HSP72 might be different between heat-shock treatment and UV irradiation. In addition, in the case of UV irradiation, the extent of DNA damage after DNA repair or the cell death might be involved in the induction of HSP72.


Subject(s)
Fibroblasts/chemistry , Heat-Shock Proteins/biosynthesis , Xeroderma Pigmentosum/chemistry , Cell Line, Transformed , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/radiation effects , Heat-Shock Proteins/radiation effects , Hot Temperature/therapeutic use , Humans , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...