Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(22)2020 Nov 23.
Article in English | MEDLINE | ID: mdl-33238630

ABSTRACT

MTH1 (MutT homolog 1) or NUDT1 (Nudix Hydrolase 1), also known as oxidized purine nucleoside triphosphatase, has potential as a biomarker for monitoring cancer progression and quantifying target engagement for relevant therapies. In this study, we validate one MTH1 inhibitor TH287 as a PET MTH1 radiotracer. TH287 was radiolabeled with tritium and the binding of [3H]TH287 to MTH1 was evaluated in live glioblastoma cells (U251MG) through saturation and competitive binding assays, together with in vitro enzymatic assays. Furthermore, TH287 was radiolabeled with carbon-11 for in vivo microPET studies. Saturation binding assays show that [3H]TH287 has a dissociation constant (Kd) of 1.97 ± 0.18 nM, Bmax of 2676 ± 122 fmol/mg protein for U251MG cells, and nH of 0.98 ± 0.02. Competitive binding assays show that TH287 (Ki: 3.04 ± 0.14 nM) has a higher affinity for MTH1 in U251MG cells compared to another well studied MTH1 inhibitor: (S)-crizotinib (Ki: 153.90 ± 20.48 nM). In vitro enzymatic assays show that TH287 has an IC50 of 2.2 nM in inhibiting MTH1 hydrolase activity and a Ki of 1.3 nM from kinetics assays, these results are consistent with our radioligand binding assays. Furthermore, MicroPET imaging shows that [11C]TH287 gets into the brain with rapid clearance from the brain, kidney, and heart. The results presented here indicate that radiolabeled TH287 has favorable properties to be a useful tool for measuring MTH1 in vitro and for further evaluation for in vivo PET imaging MTH1 of brain tumors and other central nervous system disorders.


Subject(s)
Biomarkers, Tumor/isolation & purification , DNA Repair Enzymes/genetics , Glioblastoma/diagnostic imaging , Phosphoric Monoester Hydrolases/genetics , Pyrimidines/pharmacology , Animals , Biomarkers, Tumor/metabolism , Brain/diagnostic imaging , Brain/metabolism , Cell Line, Tumor , Crizotinib/pharmacology , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/isolation & purification , Glioblastoma/genetics , Glioblastoma/pathology , Heart/diagnostic imaging , Humans , Kidney/diagnostic imaging , Kidney/metabolism , Mice , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/isolation & purification , Positron Emission Tomography Computed Tomography , Pyrimidines/chemistry
2.
Int J Biol Macromol ; 160: 903-914, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32502608

ABSTRACT

Uracil-DNA glycosylases (UDGs) are conserved DNA-repair enzymes that can be found in many species, including herpesviruses. Since they play crucial roles for efficient viral DNA replication in herpesviruses, they have been considered as potential antiviral targets. In our previous work, Staphylococcus aureus SAUGI was identified as a DNA mimic protein that targets UDGs from S. aureus, human, Herpes simplex virus (HSV) and Epstein-Barr virus (EBV). Interestingly, SAUGI has the strongest inhibitory effects with EBVUDG. Here, we determined complex structures of SAUGI with EBVUDG and another γ-herpesvirus UDG from Kaposi's sarcoma-associated herpesvirus (KSHVUDG), which SAUGI fails to effectively inhibit. Structural analysis of the SAUGI/EBVUDG complex suggests that the additional interaction between SAUGI and the leucine loop may explain why SAUGI shows the highest binding capacity with EBVUDG. In contrast, SAUGI appears to make only partial contacts with the key components responsible for the compression and stabilization of the DNA backbone in the leucine loop extension of KSHVUDG. The findings in this study provide a molecular explanation for the differential inhibitory effects and binding strengths that SAUGI has on these two UDGs, and the structural basis of the differences should be helpful in developing inhibitors that would interfere with viral DNA replication.


Subject(s)
DNA Repair Enzymes/chemistry , Gammaherpesvirinae/enzymology , Uracil-DNA Glycosidase/chemistry , Amino Acid Substitution , DNA Repair Enzymes/isolation & purification , DNA Repair Enzymes/metabolism , DNA Replication , Models, Molecular , Molecular Conformation , Protein Binding , Recombinant Proteins , Structure-Activity Relationship , Uracil-DNA Glycosidase/isolation & purification , Uracil-DNA Glycosidase/metabolism
3.
Biosens Bioelectron ; 150: 111865, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31740260

ABSTRACT

We develop a low-background electrochemical biosensor for one-step detection of uracil DNA glycosylase (UDG) based on the host-guest interaction and iron-embedded nitrogen-rich carbon nanotube (Fe-N-C) that mimics enzyme-mediated electrocatalysis to achieve signal amplification. In this work, Fe-N-C is initially immobilized on a glassy carbon electrode, followed by the immobilization of ß-cyclodextrin (ß-CD). We construct the signal probes by assembling the methylene blue (MB)-labeled hairpin DNAs onto the surface of Au nanoparticles (AuNPs) to form the MB-hairpin/AuNP probes. Due to the steric effect of AuNPs and the stem-loop structure of hairpin DNA, MB is prevented from entering the cavity of ß-CD on the electrode. In contrast, UDG enables the removal of uracil from the U•A pairs in the stem of hairpin DNA probe to generate apurinic/apyrimidinic (AP) sites, leading to the assembly of MB-hairpin/AuNP probes on the electrode based on host-guest reaction between ß-CD and MB. Meanwhile, L-cysteine (RSH) is oxidized by O2 to disulfide L-cystine (RSSR) and H2O2. In the presence of H2O2, Fe-N-C catalyzes the oxidation of MB to generate an amplified electrochemical signal. Notably, the Fe-N-C-catalyzed oxidation of MB is mediated by the oxidation of RSH by O2 instead of external H2O2, greatly simplifying the experimental procedures and improving the electrochemical signal. Due to the introduction of host-guest recognition, this electrochemical biosensor displays a low-background signal and high signal-to-noise ratio, enabling the one-step sensitive measurement of UDG with a detection limit of 7.4 × 10-5 U mL-1. Moreover, this biosensor can measure UDG in crude cell extracts and screen the inhibitors, providing a new platform for biomedical research.


Subject(s)
Biosensing Techniques , DNA Repair Enzymes/isolation & purification , DNA Repair/genetics , Electrochemical Techniques , DNA Repair Enzymes/genetics , Gold/chemistry , Hydrogen Peroxide/chemistry , Iron/chemistry , Metal Nanoparticles/chemistry , Methylene Blue/chemistry , Nanotubes, Carbon/chemistry
4.
Cell Death Dis ; 10(6): 434, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31164636

ABSTRACT

Human mutT homolog 1(MTH1), the oxidized dNTP pool sanitizer enzyme, has been reported to be highly expressed in various malignant tumors. However, the oncogenic role of MTH1 in gastric cancer remains to be determined. In the current study, we found that MTH1 was overexpressed in human gastric cancer tissues and cells. Using an in vitro MTH1 inhibitor screening system, the compounds available in our laboratory were screened and the small molecules containing 5-cyano-6-phenylpyrimidine structure were firstly found to show potently and specifically inhibitory effect on MTH1, especially compound MI-743 with IC50 = 91.44 ± 1.45 nM. Both molecular docking and target engagement experiments proved that MI-743 can directly bind to MTH1. Moreover, MI-743 could not only inhibit cell proliferation in up to 16 cancer cell lines, especially gastric cancer cells HGC-27 and MGC-803, but also significantly induce MTH1-related 8-oxo-dG accumulation and DNA damage. Furthermore, the growth of xenograft tumours derived by injection of MGC-803 cells in nude mice was also significantly inhibited by MI-743 treatment. Importantly, MTH1 knockdown by siRNA in those two gastric cancer cells exhibited the similar findings. Our findings indicate that MTH1 is highly expressed in human gastric cancer tissues and cell lines. Small molecule MI-743 with 5-cyano-6-phenylpyrimidine structure may serve as a novel lead compound targeting the overexpressed MTH1 for gastric cancer treatment.


Subject(s)
8-Hydroxy-2'-Deoxyguanosine/metabolism , Antineoplastic Agents/pharmacology , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/metabolism , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Stomach Neoplasms/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , DNA Damage/drug effects , DNA Repair Enzymes/genetics , DNA Repair Enzymes/isolation & purification , Female , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Docking Simulation , Mutation , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/isolation & purification , Stomach Neoplasms/genetics , Transplantation, Heterologous
5.
Methods Mol Biol ; 1999: 225-244, 2019.
Article in English | MEDLINE | ID: mdl-31127580

ABSTRACT

DNA double-strand breaks (DSBs) are a potentially lethal DNA lesions that disrupt both the physical and genetic continuity of the DNA duplex. Homologous recombination (HR) is a universally conserved genome maintenance pathway that initiates via nucleolytic processing of the broken DNA ends (resection). Eukaryotic DNA resection is catalyzed by the resectosome-a multicomponent molecular machine consisting of the nucleases DNA2 or Exonuclease 1 (EXO1), Bloom's helicase (BLM), the MRE11-RAD50-NBS1 (MRN) complex, and additional regulatory factors. Here, we describe methods for purification and single-molecule imaging and analysis of EXO1, DNA2, and BLM. We also describe how to adapt resection assays to the high-throughput single-molecule DNA curtain assay. By organizing hundreds of individual molecules on the surface of a microfluidic flowcell, DNA curtains visualize protein complexes with the required spatial and temporal resolution to resolve the molecular choreography during critical DNA-processing reactions.


Subject(s)
Microfluidic Analytical Techniques/methods , Recombinational DNA Repair , Single Molecule Imaging/methods , DNA Breaks, Double-Stranded , DNA Helicases/analysis , DNA Helicases/genetics , DNA Helicases/isolation & purification , DNA Repair Enzymes/analysis , DNA Repair Enzymes/genetics , DNA Repair Enzymes/isolation & purification , Exodeoxyribonucleases/analysis , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/isolation & purification , Microscopy, Fluorescence/methods , Quantum Dots/chemistry , RecQ Helicases/genetics , RecQ Helicases/isolation & purification , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
6.
Oncol Rep ; 41(3): 1851-1862, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30569141

ABSTRACT

Human MutT homolog 1 (MTH1) hydrolyses oxidised nucleotide triphosphates, thereby preventing them from being incorporated into DNA; MTH1 has been found to be elevated in many types of cancers, including lung, stomach cancer, melanoma and breast cancer. Thus, tumour­targeted hMTH1 may be valuable for developing novel anticancer therapies. In the present study, we prepared human MTH1 protein and its monoclonal antibody (mAb). The hMTH1 gene was cloned into the prokaryotic expression vector pET28a and optimally expressed in the E. coli Transetta (DE3) strain. Using an Ni­NTA column and a G­50 gel filtration column, 20.1 mg of active hMTH1 was obtained from 1,000 ml of bacterial culture, and the purity was over 98%, as detected by high­performance liquid chromatography (HPLC). The half maximal inhibitory concentration (IC50) of TH287 (hMTH1 inhibitor) was determined to be 3.53±0.47 nM using the recombinant hMTH1 protein (rhMTH1). The enzyme activity assay showed the Michaelis constant (Km) and the catalytic constant (kcat) of the protein were 106.13±48.83 µM and 3.64±0.58 sec­1, respectively. The anti­hMTH1 mAb was obtained via the hybridoma technique and validated by western blot analysis. In addition, an immunofluorescence assay (IFA) and ELISA determined that the mAb could efficiently bind to natural hMTH1 expressed on the human breast cancer cell line MCF­7. Taken together, the results showed the rhMTH1 is an active protein and has practical applications for inhibitor selection, and our prepared hMTH1 mAb will provide a valuable tool for the further characterisation of hMTH1 and antitumour medicinal development in future.


Subject(s)
Antibodies, Monoclonal/immunology , DNA Repair Enzymes/immunology , DNA Repair Enzymes/metabolism , Drug Development/methods , Phosphoric Monoester Hydrolases/immunology , Phosphoric Monoester Hydrolases/metabolism , Antibodies, Monoclonal/isolation & purification , Cell Line, Tumor , Cloning, Molecular , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/isolation & purification , Drug Screening Assays, Antitumor/methods , Enzyme Assays/methods , Humans , Oxidation-Reduction , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/isolation & purification , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
7.
Nat Commun ; 9(1): 3877, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250067

ABSTRACT

PAXX is a recently identified component of the nonhomologous end joining (NHEJ) DNA repair pathway. The molecular mechanisms of PAXX action remain largely unclear. Here we characterise the interactomes of PAXX and its paralogs, XLF and XRCC4, to show that these factors share the ability to interact with DNA polymerase λ (Pol λ), stimulate its activity and are required for recruitment of Pol λ to laser-induced DNA damage sites. Stimulation of Pol λ activity by XRCC4 paralogs requires a direct interaction between the SP/8 kDa domain of Pol λ and their N-terminal head domains to facilitate recognition of the 5' end of substrate gaps. Furthermore, PAXX and XLF collaborate with Pol λ to promote joining of incompatible DNA ends and are redundant in supporting Pol λ function in vivo. Our findings identify Pol λ as a novel downstream effector of PAXX function and show XRCC4 paralogs act in synergy to regulate polymerase activity in NHEJ.


Subject(s)
DNA End-Joining Repair/physiology , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , DNA Breaks, Double-Stranded/radiation effects , DNA Repair Enzymes/genetics , DNA Repair Enzymes/isolation & purification , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/isolation & purification , HEK293 Cells , Humans , Lasers/adverse effects , Mutagenesis, Site-Directed , Protein Binding/physiology , Protein Domains/physiology , Protein Interaction Mapping/methods , Protein Interaction Maps/physiology , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Tandem Mass Spectrometry/methods
8.
Methods Enzymol ; 600: 25-66, 2018.
Article in English | MEDLINE | ID: mdl-29458761

ABSTRACT

Accurate repair of DNA double-strand breaks (DSBs) is carried out by homologous recombination. In order to repair DNA breaks by the recombination pathway, the 5'-terminated DNA strand at DSB sites must be first nucleolytically processed to produce 3'-overhang. The process is termed DNA end resection and involves the interplay of several nuclease complexes. DNA end resection commits DSB repair to the recombination pathway including a process termed single-strand annealing, as resected DNA ends are generally nonligatable by the competing nonhomologous end-joining machinery. Biochemical reconstitution experiments provided invaluable mechanistic insights into the DNA end resection pathways. In this chapter, we describe preparation procedures of key proteins involved in DNA end resection in human cells, including the MRE11-RAD50-NBS1 complex, phosphorylated variant of CtIP, the DNA2 nuclease-helicase with its helicase partners Bloom (BLM) or Werner (WRN), as well as the single-stranded DNA-binding protein replication protein A. The availability of recombinant DNA end resection factors will help to further elucidate resection mechanisms and regulatory processes that may involve novel protein partners and posttranslational modifications.


Subject(s)
Cell Culture Techniques/methods , DNA Breaks, Double-Stranded , Enzyme Assays/methods , Recombinant Proteins/isolation & purification , Recombinational DNA Repair , Acid Anhydride Hydrolases , Animals , Baculoviridae/genetics , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , Cell Culture Techniques/instrumentation , Cell Cycle Proteins/isolation & purification , Cell Cycle Proteins/metabolism , DNA Helicases/isolation & purification , DNA Helicases/metabolism , DNA Repair Enzymes/isolation & purification , DNA Repair Enzymes/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases , Enzyme Assays/instrumentation , Humans , MRE11 Homologue Protein/isolation & purification , MRE11 Homologue Protein/metabolism , Nuclear Proteins/isolation & purification , Nuclear Proteins/metabolism , RecQ Helicases/isolation & purification , RecQ Helicases/metabolism , Recombinant Proteins/metabolism , Replication Protein A/isolation & purification , Replication Protein A/metabolism , Sf9 Cells , Spodoptera , Transfection/methods , Werner Syndrome Helicase/isolation & purification , Werner Syndrome Helicase/metabolism
9.
Methods Enzymol ; 600: 67-106, 2018.
Article in English | MEDLINE | ID: mdl-29458776

ABSTRACT

DNA end resection initiates the largely accurate repair of DNA double-strand breaks (DSBs) by homologous recombination. Specifically, recombination requires the formation of 3' overhangs at DSB sites, which is carried out by nucleases that specifically degrade 5'-terminated DNA. In most cases, DNA end resection is a two-step process, comprising of initial short-range followed by more processive long-range resection. In this chapter, we describe selected assays that reconstitute both the short- and long-range pathways. First, we define methods to study the exonuclease and endonuclease activities of the MRE11-RAD50-NBS1 (MRN) complex in conjunction with phosphorylated cofactor CtIP. This reaction is particularly important to initiate processing of DNA breaks and to recruit components belonging to the subsequent long-range pathway. Next, we describe assays that reconstitute the concerted reactions of Bloom (BLM) or Werner (WRN) helicases that function together with the DNA2 nuclease-helicase, and which are as a complex capable to resect DNA of kilobases in length. The reconstituted reactions allow us to understand how the resection pathways function at the molecular level. The assays will be invaluable to define regulatory mechanisms and to identify inhibitory compounds, which may be valuable in cancer therapy.


Subject(s)
Cell Culture Techniques/methods , DNA Breaks, Double-Stranded , Enzyme Assays/methods , Recombinant Proteins/isolation & purification , Recombinational DNA Repair , Acid Anhydride Hydrolases , Animals , Baculoviridae/genetics , Buffers , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , Cell Culture Techniques/instrumentation , Cell Cycle Proteins/isolation & purification , Cell Cycle Proteins/metabolism , DNA Helicases/isolation & purification , DNA Helicases/metabolism , DNA Repair Enzymes/isolation & purification , DNA Repair Enzymes/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , Electrophoresis, Polyacrylamide Gel/instrumentation , Electrophoresis, Polyacrylamide Gel/methods , Endodeoxyribonucleases , Enzyme Assays/instrumentation , Humans , MRE11 Homologue Protein/isolation & purification , MRE11 Homologue Protein/metabolism , Nuclear Proteins/isolation & purification , Nuclear Proteins/metabolism , Oligonucleotides/metabolism , RecQ Helicases/isolation & purification , RecQ Helicases/metabolism , Recombinant Proteins/metabolism , Replication Protein A/isolation & purification , Replication Protein A/metabolism , Sf9 Cells , Spodoptera , Transfection/methods , Werner Syndrome Helicase/isolation & purification , Werner Syndrome Helicase/metabolism
10.
Article in English | MEDLINE | ID: mdl-23295485

ABSTRACT

Human MTH1 (hMTH1) is an enzyme that hydrolyses several oxidized purine nucleoside triphosphates to their corresponding nucleoside monophosphates. Crystallographic studies have shown that the accurate mode of interaction between 8-oxoguanine and hMTH1 cannot be understood without determining the positions of the H atoms, as can be observed in neutron and/or ultrahigh-resolution X-ray diffraction studies. The hMTH1 protein prepared in the original expression system from Escherichia coli did not appear to be suitable for obtaining high-quality crystals because the hMTH1 protein had heterogeneous N-termini of Met1 and Gly2 that resulted from N-terminal Met excision by methionine aminopeptidase from the E. coli host. To obtain homogeneous hMTH1, the Gly at the second position was replaced by Lys. As a result, mutant hMTH1 protein [hMTH1(G2K)] with a homogeneous N-terminus could be prepared and high-quality crystals which diffracted to near 1.1 Šresolution using synchrotron radiation were produced. The new crystals belonged to space group P2(1)2(1)2(1), with unit-cell parameters a = 46.36, b = 47.58, c = 123.89 Å.


Subject(s)
DNA Repair Enzymes/chemistry , Phosphoric Monoester Hydrolases/chemistry , Amino Acid Substitution , Crystallization/methods , Crystallography, X-Ray , DNA Repair Enzymes/genetics , DNA Repair Enzymes/isolation & purification , Glycine/chemistry , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/metabolism , Humans , Lysine/chemistry , Mutation , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/isolation & purification , Protein Conformation
11.
Nucleic Acids Res ; 40(22): 11571-82, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23042675

ABSTRACT

During mammalian base excision repair (BER) of lesion-containing DNA, it is proposed that toxic strand-break intermediates generated throughout the pathway are sequestered and passed from one step to the next until repair is complete. This stepwise process is termed substrate channeling. A working model evaluated here is that a complex of BER factors may facilitate the BER process. FLAG-tagged DNA polymerase (pol) ß was expressed in mouse fibroblasts carrying a deletion in the endogenous pol ß gene, and the cell extract was subjected to an 'affinity-capture' procedure using anti-FLAG antibody. The pol ß affinity-capture fraction (ACF) was found to contain several BER factors including polymerase-1, X-ray cross-complementing factor1-DNA ligase III and enzymes involved in processing 3'-blocked ends of BER intermediates, e.g. polynucleotide kinase and tyrosyl-DNA phosphodiesterase 1. In contrast, DNA glycosylases, apurinic/aprymidinic endonuclease 1 and flap endonuclease 1 and several other factors involved in BER were not present. Some of the BER factors in the pol ß ACF were in a multi-protein complex as observed by sucrose gradient centrifugation. The pol ß ACF was capable of substrate channeling for steps in vitro BER and was proficient in in vitro repair of substrates mimicking a 3'-blocked topoisomerase I covalent intermediate or an oxidative stress-induced 3'-blocked intermediate.


Subject(s)
DNA Polymerase beta/metabolism , DNA Repair Enzymes/metabolism , DNA Repair , Animals , Cell Extracts , DNA/metabolism , DNA Contamination , DNA Repair Enzymes/isolation & purification , Fibroblasts/metabolism , Mice , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/isolation & purification
12.
J Biol Chem ; 287(31): 26254-67, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22692201

ABSTRACT

Human SNM1A and SNM1B/Apollo have both been implicated in the repair of DNA interstrand cross-links (ICLs) by cellular studies, and SNM1B is also required for telomere protection. Here, we describe studies on the biochemical characterization of the SNM1A and SNM1B proteins. The results reveal some fundamental differences in the mechanisms of the two proteins. Both SNM1A and SNM1B digest double-stranded and single-stranded DNA with a 5'-to-3' directionality in a reaction that is stimulated by divalent cations, and both nucleases are inhibited by the zinc chelator o-phenanthroline. We find that SNM1A has greater affinity for single-stranded DNA over double-stranded DNA that is not observed with SNM1B. Although both proteins demonstrate a low level of processivity on low molecular weight DNA oligonucleotide substrates, when presented with high molecular weight DNA, SNM1A alone is rendered much more active, being capable of digesting kilobase-long stretches of DNA. Both proteins can digest past ICLs induced by the non-distorting minor groove cross-linking agent SJG-136, albeit with SNM1A showing a greater capacity to achieve this. This is consistent with the proposal that SNM1A and SNM1B might exhibit some redundancy in ICL repair. Together, our work establishes differences in the substrate selectivities of SNM1A and SNM1B that are likely to be relevant to their in vivo roles and which might be exploited in the development of selective inhibitors.


Subject(s)
DNA Repair Enzymes/chemistry , DNA-Binding Proteins/chemistry , Nuclear Proteins/chemistry , Cell Cycle Proteins , Chelating Agents/chemistry , DNA/chemistry , DNA Cleavage , DNA Damage , DNA Repair Enzymes/biosynthesis , DNA Repair Enzymes/isolation & purification , DNA, Single-Stranded/chemistry , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/isolation & purification , Enzyme Assays , Enzyme Inhibitors/chemistry , Escherichia coli , Exodeoxyribonucleases , Fluorescein/chemistry , Fluorescent Dyes/chemistry , Humans , Hydrogen-Ion Concentration , Hydrolysis , Magnesium/chemistry , Nuclear Proteins/biosynthesis , Nuclear Proteins/isolation & purification , Plasmids/chemistry , Protein Binding , RNA/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Substrate Specificity
13.
Article in English | MEDLINE | ID: mdl-22232169

ABSTRACT

Cockayne syndrome protein A is one of the main components in mammalian transcription coupled repair. Here, the overproduction, purification and crystallization of human Cockayne syndrome protein A in complex with its interacting partner DNA damage binding protein 1 are reported. The complex was coproduced in insect cells, copurified and crystallized using sitting drops with PEG 3350 and sodium citrate as crystallizing agents. The crystals had unit-cell parameters a = b = 142.03, c = 250.19 Å and diffracted to 2.9 Å resolution on beamline ID14-1 at the European Synchrotron Radiation Facility.


Subject(s)
DNA Repair Enzymes/chemistry , DNA-Binding Proteins/chemistry , Transcription Factors/chemistry , Crystallization , Crystallography, X-Ray , DNA Repair Enzymes/isolation & purification , DNA-Binding Proteins/isolation & purification , Humans , Protein Binding , Transcription Factors/isolation & purification
14.
DNA Repair (Amst) ; 11(1): 46-52, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22079122

ABSTRACT

Exocyclic ethenobases are highly mutagenic DNA lesions strongly implicated in inflammation and vinyl chloride-induced carcinogenesis. While the alkyladenine DNA glycosylase, AAG (or MPG), binds the etheno lesions 1,N(6)-ethenoadenine (ɛA) and 3,N(4)-ethenocytosine (ɛC) with high affinity, only ɛA can be excised to initiate base excision repair. Here, we discover that the human AlkB homolog 2 (ALKBH2) dioxygenase enzyme catalyzes direct reversal of ɛC lesions in both double- and single-stranded DNA with comparable efficiency to canonical ALKBH2 substrates. Notably, we find that in vitro, the non-enzymatic binding of AAG to ɛC specifically blocks ALKBH2-catalyzed repair of ɛC but not that of methylated ALKBH2 substrates. These results identify human ALKBH2 as a repair enzyme for mutagenic ɛC lesions and highlight potential consequences for substrate-binding overlap between the base excision and direct reversal DNA repair pathways.


Subject(s)
Cytosine/analogs & derivatives , DNA Glycosylases/metabolism , DNA Repair Enzymes/metabolism , DNA Repair , Dioxygenases/metabolism , AlkB Homolog 2, Alpha-Ketoglutarate-Dependent Dioxygenase , Cytosine/metabolism , DNA Damage , DNA Repair Enzymes/isolation & purification , DNA Restriction Enzymes/metabolism , Dioxygenases/isolation & purification , Humans , Substrate Specificity
15.
Exp Oncol ; 33(3): 121-5, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21956462

ABSTRACT

UNLABELLED: Proteins tightly bound to DNA (TBP) comprise a group of proteins that remain bound to DNA even after harsh deproteinization procedures. The amount of these proteins is 20-100 µg for mg of DNA depending on eukaryotic source. This experimental paper examines the possibility to use some TBP for clinical biomarker discovery, e.g. for identification of prognostic and diagnostic cancer markers. The main aim of this study was to designate differences between tightly DNA binding protein patterns extracted from rat liver and rat experimental hepatomas (Zajdela ascites hepatoma and hepatoma G-27) and to evaluate possibility that some of these proteins may be used as biomarkers for cell cancer transformation. METHODS: We used proteomics aproach as a tool for comparison of pattern of TBP from rat experimental hepatomas and normal liver cells. Combination of 2DE fractionation with mass spectrometry (MALDI TOF-MS) suitable for parallel profiling of complex TBP mixtures. RESULTS: Intriguingly 2DE protein maps of TBP from rat liver and rat experimental hepatomas (Zajdela acites hepatoma and hepatoma G-27) were quite different. We identified 9 proteins, some of them shared in all TBP patterns. Among identified tightly bound to DNA proteins there were three proteins considered as nuclear matrix proteins (lamin B1, scaffold attachment factor B1, heterogeneous nuclear ribonucleoprotein). Also we identified DNA repair protein RAD50, coiled-coil domain-containing protein 41, structural maintenance of chromosomes protein1A and some ATP -dependent RNA helicases indicating that TBP are of interest with respect to their potential involvement in the topological organization and/ or function of genomic DNA. CONCLUSIONS: We suppose that proteomic approach for TBP identification may be promising in development of biomarkers, also obtained results may be valuable for further understanding TBP functions in genome.


Subject(s)
Biomarkers, Tumor/isolation & purification , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , Liver Neoplasms, Experimental/metabolism , Animals , Biomarkers, Tumor/metabolism , DNA Repair Enzymes/isolation & purification , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , Liver/metabolism , Liver Neoplasms, Experimental/genetics , Nuclear Matrix , Nuclear Matrix-Associated Proteins/isolation & purification , Nuclear Matrix-Associated Proteins/metabolism , Prognosis , Protein Binding , Proteomics , RNA Helicases/isolation & purification , RNA Helicases/metabolism , Rats , Rats, Wistar , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
16.
Anal Biochem ; 413(2): 179-84, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21329650

ABSTRACT

The ability to monitor and characterize DNA mismatch repair activity in various mammalian cells is important for understanding mechanisms involved in mutagenesis and tumorigenesis. Since mismatch repair proteins recognize mismatches containing both normal and chemically altered or damaged bases, in vitro assays must accommodate a variety of mismatches in different sequence contexts. Here we describe the construction of DNA mismatch substrates containing G:T or O(6)meG:T mismatches, the purification of recombinant native human MutSα (MSH2-MSH6) and MutLα (MLH1-PMS2) proteins, and in vitro mismatch repair and excision assays that can be adapted to study mismatch repair in nuclear extracts from mismatch repair proficient and deficient cells.


Subject(s)
DNA Mismatch Repair , DNA Repair Enzymes/metabolism , Recombinant Proteins/metabolism , Base Sequence , Cell Line , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/isolation & purification , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , Humans , MutL Proteins , Polynucleotides/chemistry , Polynucleotides/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
17.
Genes Genet Syst ; 85(4): 287-95, 2010.
Article in English | MEDLINE | ID: mdl-21178309

ABSTRACT

The oxidized nucleotide precursors 7, 8-dihydro-8-oxo-dGTP (8-oxo-dGTP) and 1, 2-dihydro-2-oxo-dATP (2-oxo-dATP) are readily incorporated into nascent DNA strands during replication, which would cause base substitution mutations. E. coli MutT and human homologue hMTH1 hydrolyze 8-oxo-dGTP, thereby preventing mutations. In this study, we searched for hMTH1 homologues in the ascidian Ciona intestinalis using the NCBI-BLAST database. Among several candidates, we focused on one open reading frame, designated as CiMutT, because of its high degree of identity (41.7%) and similarity (58.3%) to the overall amino acid sequence of hMTH1, including the Nudix box. CiMutT significantly suppressed the mutator activity of E. coli mutT mutant. Purified CiMutT had a pyrophosphohydrolase activity that hydrolyzed 8-oxo-dGTP to 8-oxo-dGMP and inorganic pyrophosphate. It had a pH optimum of 9.5 and Mg(++) requirement with optimal activity at 5 mM. The activity of CiMutT for 8-oxo-dGTP was comparable to that of hMTH1, while it was 100-fold lower for 2-oxo-dATP than that of hMTH1. These facts indicate that CiMutT is a functional homologue of E. coli MutT. In addition, the enzyme hydrolyzed all four of the unoxidized nucleoside triphosphates, with a preference for dATP. The specific activity for 8-oxo-dGTP was greater than that for unoxidized dATP and dGTP. These results suggest that CiMutT has the potential to prevent mutations by 8-oxo-dGTP in C. intestinalis.


Subject(s)
Ciona intestinalis/enzymology , DNA Repair Enzymes/biosynthesis , Phosphoric Monoester Hydrolases/biosynthesis , Amino Acid Sequence , Animals , DNA Repair , DNA Repair Enzymes/genetics , DNA Repair Enzymes/isolation & purification , Deoxyadenine Nucleotides/metabolism , Deoxyguanine Nucleotides/metabolism , Escherichia coli Proteins/genetics , Humans , Molecular Sequence Data , Mutation , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/isolation & purification , Pyrophosphatases/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Sequence Alignment
18.
Structure ; 18(11): 1431-42, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-21070942

ABSTRACT

DNA ligase IV (LigIV) is critical for nonhomologous end joining (NHEJ), the major DNA double-strand break (DSB) repair pathway in human cells, and LigIV activity is regulated by XRCC4 and XLF (XRCC4-like factor) interactions. Here, we employ small angle X-ray scattering (SAXS) data to characterize three-dimensional arrangements in solution for full-length XRCC4, XRCC4 in complex with LigIV tandem BRCT domains and XLF, plus the XRCC4·XLF·BRCT2 complex. XRCC4 forms tetramers mediated through a head-to-head interface, and the XRCC4 C-terminal coiled-coil region folds back on itself to support this interaction. The interaction between XLF and XRCC4 is also mediated via head-to-head interactions. In the XLF·XRCC4·BRCT complex, alternating repeating units of XLF and XRCC4·BRCT place the BRCT domain on one side of the filament. Collective results identify XRCC4 and XLF filaments suitable to align DNA molecules and function to facilitate LigIV end joining required for DSB repair in vivo.


Subject(s)
DNA Ligases/chemistry , DNA Repair Enzymes/chemistry , DNA-Binding Proteins/chemistry , Models, Molecular , Molecular Dynamics Simulation , Protein Conformation , Chromatography, Gel , DNA Ligase ATP , DNA Ligases/isolation & purification , DNA Ligases/metabolism , DNA Repair Enzymes/isolation & purification , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , Humans , Scattering, Small Angle
19.
Proc Natl Acad Sci U S A ; 107(31): 13660-5, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20616048

ABSTRACT

ATR kinase is a critical upstream regulator of the checkpoint response to various forms of DNA damage. Previous studies have shown that ATR is recruited via its binding partner ATR-interacting protein (ATRIP) to replication protein A (RPA)-covered single-stranded DNA (RPA-ssDNA) generated at sites of DNA damage where ATR is then activated by TopBP1 to phosphorylate downstream targets including the Chk1 signal transducing kinase. However, this critical feature of the human ATR-initiated DNA damage checkpoint signaling has not been demonstrated in a defined system. Here we describe an in vitro checkpoint system in which RPA-ssDNA and TopBP1 are essential for phosphorylation of Chk1 by the purified ATR-ATRIP complex. Checkpoint defective RPA mutants fail to activate ATR kinase in this system, supporting the conclusion that this system is a faithful representation of the in vivo reaction. Interestingly, we find that an alternative form of RPA (aRPA), which does not support DNA replication, can substitute for the checkpoint function of RPA in vitro, thus revealing a potential role for aRPA in the activation of ATR kinase. We also find that TopBP1 is recruited to RPA-ssDNA in a manner dependent on ATRIP and that the N terminus of TopBP1 is required for efficient recruitment and activation of ATR kinase.


Subject(s)
Cell Cycle Proteins/metabolism , DNA, Single-Stranded/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Replication Protein A/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/isolation & purification , Adaptor Proteins, Signal Transducing/metabolism , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/metabolism , Cell Cycle Proteins/isolation & purification , Checkpoint Kinase 1 , DNA Repair Enzymes/isolation & purification , DNA Repair Enzymes/metabolism , DNA, Single-Stranded/chemistry , DNA-Binding Proteins/metabolism , Enzyme Activation , HeLa Cells , Humans , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/isolation & purification
20.
Anal Chem ; 82(7): 2784-96, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20225856

ABSTRACT

Many cellular processes are driven by protein complexes. Although the identification of protein components in such complexes has become almost a routine matter, accurate determination of their stoichiometry within a protein complex is still a challenge. We have established a method to determine the stoichiometries of protein complexes using absolute quantification (AQUA) with the help of synthetic standard peptides in combination with multiple reaction monitoring (MRM). Our approach is exemplified by the analysis of the human spliceosomal hPrp19/CDC5L complex, which consists of seven individual proteins and plays a crucial role in the assembly of the fully catalytically active spliceosome during pre-mRNA splicing. We evaluated several conditions for complete hydrolysis of the protein complex and found that the denaturing conditions under which hydrolysis is performed are absolutely crucial for accurately determining protein stoichiometries within this complex. In addition, we tested the suitability of different AQUA peptides and further compared different MS techniques to read out the relative signal intensities that were then used in absolute quantification. Our analyses revealed that dependent on the denaturing conditions different stoichiometries within the complex were obtained. The most consistent results were obtained by enzymatic hydrolysis in the presence of acetonitrile in combination with MRM.


Subject(s)
Multiprotein Complexes/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Amino Acid Sequence , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/isolation & purification , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/isolation & purification , Humans , Hydrolysis , Nuclear Proteins/chemistry , Nuclear Proteins/isolation & purification , RNA Splicing Factors , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/isolation & purification , Spliceosomes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL