Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
Biosci Rep ; 40(4)2020 04 30.
Article in English | MEDLINE | ID: mdl-32314787

ABSTRACT

DNA polymerase δ (Polδ) is a highly processive essential replicative DNA polymerase. In humans, the Polδ holoenzyme consists of p125, p50, p68 and p12 subunits and recently, we showed that the p12 subunit exists as a dimer. Extensive biochemical studies suggest that all the subunits of Polδ interact with the processivity factor proliferating cell nuclear antigen (PCNA) to carry out a pivotal role in genomic DNA replication. While PCNA-interacting protein motif (PIP) motifs in p68, p50 and p12 have been mapped, same in p125, the catalytic subunit of the holoenzyme, remains elusive. Therefore, in the present study by using multiple approaches we have conclusively mapped a non-canonical PIP motif from residues 999VGGLLAFA1008 in p125, which binds to the inter-domain-connecting loop (IDCL) of PCNA with high affinity. Collectively, including previous studies, we conclude that similar to Saccharomyces cerevisiae Polδ, each of the human Polδ subunits possesses motif to interact with PCNA and significantly contributes toward the processive nature of this replicative DNA polymerase.


Subject(s)
DNA Polymerase III/genetics , DNA Replication , Proliferating Cell Nuclear Antigen/metabolism , Protein Interaction Domains and Motifs/genetics , Animals , CHO Cells , Cricetulus , DNA Polymerase III/isolation & purification , DNA Polymerase III/metabolism , Mutagenesis, Site-Directed , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Surface Plasmon Resonance
2.
J Chromatogr A ; 1602: 341-349, 2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31204039

ABSTRACT

Protein purification by affinity chromatography relies primarily on the interaction of a fused-tag to the protein of interest. Here, we describe a tag-free affinity method that employs functional selection interactions to a broad range of proteins. To achieve this, we coupled human DNA-clamp proliferating cell nuclear antigen (PCNA) that interacts with over one hundred proteins to an agarose resin. We demonstrate the versatility of our PCNA-Agarose column at various chromatographic steps by purifying PCNA-binding proteins that are involved in DNA Replication (DNA polymerase δ, flap endonuclease 1 and DNA ligase 1), translesion DNA synthesis (DNA polymerases eta, kappa and iota) and genome stability (p15). We also show the competence of the PCNA-Agarose column to purify non-PCNA binding proteins by fusing the PCNA-binding motif of human p21 as an affinity tag. Finally, we establish that our PCNA-Agarose column is a suitable analytical method for characterizing the binding strength of PCNA-binding proteins. The conservation and homology of PCNA-like clamps will allow for the immediate extension of our method to other species.


Subject(s)
Chromatography, Affinity/methods , Proliferating Cell Nuclear Antigen/isolation & purification , Sepharose/chemistry , Buffers , DNA Polymerase III/isolation & purification , DNA Repair , DNA Replication , Humans , Protein Binding , Recombinant Proteins/isolation & purification , Resins, Synthetic/chemistry
3.
J Biol Chem ; 294(11): 3947-3956, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30655288

ABSTRACT

Human DNA polymerase δ is essential for DNA replication and acts in conjunction with the processivity factor proliferating cell nuclear antigen (PCNA). In addition to its catalytic subunit (p125), pol δ comprises three regulatory subunits (p50, p68, and p12). PCNA interacts with all of these subunits, but only the interaction with p68 has been structurally characterized. Here, we report solution NMR-, isothermal calorimetry-, and X-ray crystallography-based analyses of the p12-PCNA interaction, which takes part in the modulation of the rate and fidelity of DNA synthesis by pol δ. We show that p12 binds with micromolar affinity to the classical PIP-binding pocket of PCNA via a highly atypical PIP box located at the p12 N terminus. Unlike the canonical PIP box of p68, the PIP box of p12 lacks the conserved glutamine; binds through a 2-fork plug made of an isoleucine and a tyrosine residue at +3 and +8 positions, respectively; and is stabilized by an aspartate at +6 position, which creates a network of intramolecular hydrogen bonds. These findings add to growing evidence that PCNA can bind a diverse range of protein sequences that may be broadly grouped as PIP-like motifs as has been previously suggested.


Subject(s)
DNA Polymerase III/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Amino Acid Motifs , Calorimetry , Catalytic Domain , DNA Polymerase III/chemistry , DNA Polymerase III/isolation & purification , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/isolation & purification
4.
J Am Chem Soc ; 139(50): 18339-18348, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29166001

ABSTRACT

A [4Fe4S]2+ cluster in the C-terminal domain of the catalytic subunit of the eukaryotic B-family DNA polymerases is essential for the formation of active multi-subunit complexes. Here we use a combination of electrochemical and biochemical methods to assess the redox activity of the [4Fe4S]2+ cluster in Saccharomyces cerevisiae polymerase (Pol) δ, the lagging strand DNA polymerase. We find that Pol δ bound to DNA is indeed redox-active at physiological potentials, generating a DNA-mediated signal electrochemically with a midpoint potential of 113 ± 5 mV versus NHE. Moreover, biochemical assays following electrochemical oxidation of Pol δ reveal a significant slowing of DNA synthesis that can be fully reversed by reduction of the oxidized form. A similar result is apparent with photooxidation using a DNA-tethered anthraquinone. These results demonstrate that the [4Fe4S] cluster in Pol δ can act as a redox switch for activity, and we propose that this switch can provide a rapid and reversible way to respond to replication stress.


Subject(s)
DNA Polymerase III/metabolism , Iron-Sulfur Proteins/metabolism , Saccharomyces cerevisiae/enzymology , DNA Polymerase III/isolation & purification , Iron-Sulfur Proteins/chemistry , Oxidation-Reduction
5.
PLoS One ; 11(12): e0169259, 2016.
Article in English | MEDLINE | ID: mdl-28036377

ABSTRACT

We describe a gentle and rapid method to purify the intact multiprotein DNA replication complex using free flow electrophoresis (FFE). In particular, we applied FFE to purify the human cell DNA synthesome, which is a multiprotein complex that is fully competent to carry-out all phases of the DNA replication process in vitro using a plasmid containing the simian virus 40 (SV40) origin of DNA replication and the viral large tumor antigen (T-antigen) protein. The isolated native DNA synthesome can be of use in studying the mechanism by which mammalian DNA replication is carried-out and how anti-cancer drugs disrupt the DNA replication or repair process. Partially purified extracts from HeLa cells were fractionated in a native, liquid based separation by FFE. Dot blot analysis showed co-elution of many proteins identified as part of the DNA synthesome, including proliferating cell nuclear antigen (PCNA), DNA topoisomerase I (topo I), DNA polymerase δ (Pol δ), DNA polymerase ɛ (Pol ɛ), replication protein A (RPA) and replication factor C (RFC). Previously identified DNA synthesome proteins co-eluted with T-antigen dependent and SV40 origin-specific DNA polymerase activity at the same FFE fractions. Native gels show a multiprotein PCNA containing complex migrating with an apparent relative mobility in the megadalton range. When PCNA containing bands were excised from the native gel, mass spectrometric sequencing analysis identified 23 known DNA synthesome associated proteins or protein subunits.


Subject(s)
DNA-Directed DNA Polymerase/analysis , DNA-Directed DNA Polymerase/isolation & purification , Electrophoresis/methods , Multienzyme Complexes/analysis , Multienzyme Complexes/isolation & purification , Antigens, Viral, Tumor/genetics , Cell Extracts/chemistry , Cell Line, Tumor , DNA Polymerase I/isolation & purification , DNA Polymerase II/isolation & purification , DNA Polymerase III/isolation & purification , DNA Replication , DNA Topoisomerases, Type I/isolation & purification , HeLa Cells , Humans , Proliferating Cell Nuclear Antigen/analysis , Replication Origin/genetics , Replication Protein A/isolation & purification , Replication Protein C/isolation & purification , Simian virus 40/genetics
6.
Genet Mol Res ; 15(2)2016 May 23.
Article in English | MEDLINE | ID: mdl-27323084

ABSTRACT

The filamentous fungus Moniliophthora perniciosa is a basidiomycota that causes the witches' broom disease in cocoa trees (Theobroma cacao L.). The mitochondrial DNA polymerase of M. perniciosa (MpmitDNApol) is classified within the B family of DNA polymerases, which can be found in viruses and cellular organelles. Using virtual screening processes, accessing KEGG, PubChem, and ZINC databases, we selected the 27 best putative nucleoside viral-like polymerase inhibitors to test against MpmitDNApol. We used Autodock Vina to perform docking simulations of the selected molecules and to return energy values in several ligand conformations. Then, we used Pymol v1.7.4.4 to check the stereochemistry of chiral carbons, hydrogen bonding receptors, absence or presence of hydrogen, sub and superstructure, numbers of rings, rotatable bonds, and donor groups. We selected the Entecavir Hydrate, a drug used to control hepatitis B; subsequently AMBER 14 was used to describe the behavior of polymerase-entecavir complex after setting up 3500 ps of simulation in water at a temperature of 300 K. From the simulation, a graph of Potential Energy was generated revealing that the ligand remains in the catalytic site after 3500 ps with a final energy of -612,587.4214 kcal/mol.


Subject(s)
Basidiomycota/genetics , DNA Polymerase III/genetics , Fungi/genetics , Plant Diseases/genetics , Computational Biology , DNA Polymerase III/isolation & purification , Mitochondria/genetics , Plant Diseases/virology , User-Computer Interface
7.
Nucleic Acids Res ; 44(15): 7242-50, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27185888

ABSTRACT

The intolerance of DNA polymerase δ (Polδ) to incorrect base pairing contributes to its extremely high accuracy during replication, but is believed to inhibit translesion synthesis (TLS). However, chicken DT40 cells lacking the POLD3 subunit of Polδ are deficient in TLS. Previous genetic and biochemical analysis showed that POLD3 may promote lesion bypass by Polδ itself independently of the translesion polymerase Polζ of which POLD3 is also a subunit. To test this hypothesis, we have inactivated Polδ proofreading in pold3 cells. This significantly restored TLS in pold3 mutants, enhancing dA incorporation opposite abasic sites. Purified proofreading-deficient human Polδ holoenzyme performs TLS of abasic sites in vitro much more efficiently than the wild type enzyme, with over 90% of TLS events resulting in dA incorporation. Furthermore, proofreading deficiency enhances the capability of Polδ to continue DNA synthesis over UV lesions both in vivo and in vitro These data support Polδ contributing to TLS in vivo and suggest that the mutagenesis resulting from loss of Polδ proofreading activity may in part be explained by enhanced lesion bypass.


Subject(s)
DNA Polymerase III/metabolism , DNA Replication , DNA/biosynthesis , DNA/chemistry , Alleles , Cell Line , DNA Damage , DNA Polymerase III/chemistry , DNA Polymerase III/genetics , DNA Polymerase III/isolation & purification , Holoenzymes/chemistry , Holoenzymes/genetics , Holoenzymes/isolation & purification , Holoenzymes/metabolism , Humans , Immunoglobulins/genetics , Ultraviolet Rays
8.
Malar J ; 15: 116, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26911594

ABSTRACT

BACKGROUND: Emergence of drug-resistant Plasmodium falciparum has created an urgent need for new drug targets. DNA polymerase δ is an essential enzyme required for chromosomal DNA replication and repair, and therefore may be a potential target for anti-malarial drug development. However, little is known of the characteristics and function of this P. falciparum enzyme. METHODS: The coding sequences of DNA polymerase δ catalytic subunit (PfPolδ-cat), DNA polymerase δ small subunit (PfPolδS) and proliferating cell nuclear antigen (PfPCNA) from chloroquine- and pyrimethamine-resistant P. falciparum strain K1 were amplified, cloned into an expression vector and expressed in Escherichia coli. The recombinant proteins were analysed by SDS-PAGE and identified by LC-MS/MS. PfPolδ-cat was biochemically characterized. The roles of PfPolδS and PfPCNA in PfPolδ-cat function were investigated. In addition, inhibitory effects of 11 compounds were tested on PfPolδ-cat activity and on in vitro parasite growth using SYBR Green I assay. RESULTS: The purified recombinant protein PfPolδ-cat, PfPolδS and PfPCNA showed on SDS-PAGE the expected size of 143, 57 and 34 kDa, respectively. Predicted amino acid sequence of the PfPolδ-cat and PfPolδS had 59.2 and 24.7 % similarity respectively to that of the human counterpart. The PfPolδ-cat possessed both DNA polymerase and 3'-5' exonuclease activities. It used both Mg(2+) and Mn(2+) as cofactors and was inhibited by high KCl salt (>200 mM). PfPolδS stimulated PfPolδ-cat activity threefolds and up to fourfolds when PfPCNA was included in the assay. Only two compounds were potent inhibitors of PfPolδ-cat, namely, butylphenyl-dGTP (BuPdGTP; IC50 of 38 µM) and 7-acetoxypentyl-(3, 4 dichlorobenzyl) guanine (7-acetoxypentyl-DCBG; IC50 of 55 µM). The latter compound showed higher inhibition on parasite growth (IC50 of 4.1 µM). CONCLUSIONS: Recombinant PfPolδ-cat, PfPolδS and PfPCNA were successfully expressed and purified. PfPolS and PfPCNA increased DNA polymerase activity of PfPolδ-cat. The high sensitivity of PfPolδ to BuPdGTP can be used to differentiate parasite enzyme from mammalian and human counterparts. Interestingly, 7-acetoxypentyl-DCBG showed inhibitory effects on both enzyme activity and parasite growth. Thus, 7-acetoxypentyl-DCBG is a potential candidate for future development of a new class of anti-malarial agents targeting parasite replicative DNA polymerase.


Subject(s)
DNA Polymerase III/chemistry , DNA Polymerase III/metabolism , Plasmodium falciparum/enzymology , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Antimalarials/pharmacology , Cells, Cultured , DNA Polymerase III/genetics , DNA Polymerase III/isolation & purification , Drug Resistance , Erythrocytes/parasitology , Humans , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/isolation & purification , Proliferating Cell Nuclear Antigen/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
9.
Chembiochem ; 13(16): 2439-44, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23023962

ABSTRACT

Thermostable bacterial polymerases like Taq, Therminator and Vent exo(-) are able to perform DNA synthesis by using modified DNA precursors, a property that is exploited in several therapeutic and biotechnological applications. Viral polymerases are also known to accept modified substrates, and this has proven crucial in the development of antiviral therapies. However, non-thermostable polymerases of bacterial origin, or engineered variants, that have similar substrate tolerance and could be used for synthetic biology purposes remain to be identified. We have identified the α subunit of Escherichia coli polymerase III (Pol III α) as a bacterial polymerase that is able to recognise and process as substrates several pyrophosphate-modified dATP analogues in place of its natural substrate dATP for template-directed DNA synthesis. A number of dATP analogues featuring a modified pyrophosphate group were able to serve as substrates during enzymatic DNA synthesis by Pol III α. Features such as the presence of potentially chelating chemical groups and the size and spatial flexibility of the chemical structure seem to be of major importance for the modified leaving group to play its role during the enzymatic reaction. In addition, we could establish that if the pyrophosphate group is altered, deoxynucleotide incorporation proceeds with an efficiency varying with the nature of the nucleobase. Our results represent a great step towards the achievement of a system of artificial DNA synthesis hosted by E. coli and involving the use of altered nucleotide precursors for nucleic acid synthesis.


Subject(s)
DNA Polymerase III/metabolism , Deoxyribonucleotides/chemistry , Escherichia coli/enzymology , DNA/biosynthesis , DNA Polymerase III/chemistry , DNA Polymerase III/isolation & purification , Deoxyribonucleotides/metabolism , Kinetics , Substrate Specificity
10.
PLoS One ; 7(6): e39156, 2012.
Article in English | MEDLINE | ID: mdl-22723953

ABSTRACT

Mammalian DNA polymerase δ (Pol δ), a four-subunit enzyme, plays a crucial and versatile role in DNA replication and DNA repair processes. We have reconstituted human Pol δ complexes in insect cells infected with a single baculovirus into which one or more subunits were assembled. This system allowed for the efficient expression of the tetrameric Pol δ holoenzyme, the p125/p50 core dimer, the core+p68 trimer and the core+p12 trimer, as well as the p125 catalytic subunit. These were isolated in milligram amounts with reproducible purity and specific activities by a highly standardized protocol. We have systematically compared their activities in order to gain insights into the roles of the p12 and p68 subunits, as well as their responses to PCNA. The relative specific activities (apparent k(cat)) of the Pol δ holoenzyme, core+p68, core+p12 and p125/p50 core were 100, 109, 40, and 29. The corresponding apparent K(d)'s for PCNA were 7.1, 8.7, 9.3 and 73 nM. Our results support the hypothesis that Pol δ interacts with PCNA through multiple interactions, and that there may be a redundancy in binding interactions that may permit Pol δ to adopt flexible configurations with PCNA. The abilities of the Pol δ complexes to fully extend singly primed M13 DNA were examined. All the subassemblies except the core+p68 were defective in their abilities to completely extend the primer, showing that the p68 subunit has an important function in synthesis of long stretches of DNA in this assay. The core+p68 trimer could be reconstituted by addition of p12.


Subject(s)
Baculoviridae/genetics , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , Baculoviridae/metabolism , Calpain/metabolism , DNA Polymerase III/isolation & purification , DNA Replication/physiology , Enzyme Activation , Gene Expression , HEK293 Cells , HeLa Cells , Holoenzymes/genetics , Holoenzymes/isolation & purification , Holoenzymes/metabolism , Humans , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Plasmids/genetics , Polydeoxyribonucleotides/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Protein Stability , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
11.
PLoS One ; 6(7): e22224, 2011.
Article in English | MEDLINE | ID: mdl-21789240

ABSTRACT

Eukaryotic DNA polymerase δ (pol δ) plays a crucial role in chromosomal DNA replication and various DNA repair processes. It is thought to consist of p125, p66 (p68), p50 and p12 subunits. However, rigorous isolation of mammalian pol δ from natural sources has usually yielded two-subunit preparations containing only p125 and p50 polypeptides. While recombinant pol δ isolated from infected insect cells have some problems of consistency in the quality of the preparations, and the yields are much lower. To address these deficiencies, we have constructed recombinant BmNPV baculoviruses using MultiBac system. This method makes the generation of recombinant forms of pol δ containing mutations in any one of the subunits or combinations thereof extremely facile. From about 350 infected larvae, we obtained as much as 4 mg of pol δ four-subunit complex. Highly purified enzyme behaved like the one of native form by rigorous characterization and comparison of its activities on poly(dA)/oligo(dT) template-primer and singly primed M13 DNA, and its homogeneity on FPLC gel filtration. In vitro base excision repair (BER) assays showed that pol δ plays a significant role in uracil-intiated BER and is more likely to mediate LP BER, while the trimer lacking p12 is more likely to mediate SN BER. It seems likely that loss of p12 modulates the rate of SN BER and LP BER during the repair process. Thus, this work provides a simple, fast, reliable and economic way for the large-scale production of human DNA polymerase δ with a high activity and purity, setting up a new platform for our further research on the biochemical properties of pol δ, its regulation and the integration of its functions, and how alterations in pol δ function could contribute to the etiology of human cancer or other diseases that can result from loss of genomic stability.


Subject(s)
Bioreactors , Biotechnology/methods , Bombyx/metabolism , DNA Polymerase III/biosynthesis , Recombinant Proteins/biosynthesis , Animals , Baculoviridae/genetics , Blotting, Western , Bombyx/virology , Chromatography, Affinity , DNA Polymerase III/isolation & purification , DNA Repair/genetics , Gene Expression , Genetic Vectors/genetics , HeLa Cells , Hemolymph/metabolism , Humans , Larva/virology , Molecular Weight , Protein Subunits/metabolism , Recombinant Proteins/isolation & purification , Recombination, Genetic/genetics , Subcellular Fractions/metabolism
12.
DNA Repair (Amst) ; 10(5): 497-505, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21429821

ABSTRACT

Elucidating the sources of genetic variation within microsatellite alleles has important implications for understanding the etiology of human diseases. Mismatch repair is a well described pathway for the suppression of microsatellite instability. However, the cellular polymerases responsible for generating microsatellite errors have not been fully described. We address this gap in knowledge by measuring the fidelity of recombinant yeast polymerase δ (Pol δ) and ɛ (Pol ɛ) holoenzymes during synthesis of a [GT/CA] microsatellite. The in vitro HSV-tk forward assay was used to measure DNA polymerase errors generated during gap-filling of complementary GT(10) and CA(10)-containing substrates and ∼90 nucleotides of HSV-tk coding sequence surrounding the microsatellites. The observed mutant frequencies within the microsatellites were 4 to 30-fold higher than the observed mutant frequencies within the coding sequence. More specifically, the rate of Pol δ and Pol ɛ misalignment-based insertion/deletion errors within the microsatellites was ∼1000-fold higher than the rate of insertion/deletion errors within the HSV-tk gene. Although the most common microsatellite error was the deletion of a single repeat unit, ∼ 20% of errors were deletions of two or more units for both polymerases. The differences in fidelity for wild type enzymes and their exonuclease-deficient derivatives were ∼2-fold for unit-based microsatellite insertion/deletion errors. Interestingly, the exonucleases preferentially removed potentially stabilizing interruption errors within the microsatellites. Since Pol δ and Pol ɛ perform not only the bulk of DNA replication in eukaryotic cells but also are implicated in performing DNA synthesis associated with repair and recombination, these results indicate that microsatellite errors may be introduced into the genome during multiple DNA metabolic pathways.


Subject(s)
DNA Polymerase III/metabolism , DNA Polymerase II/metabolism , Microsatellite Repeats/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Base Sequence , DNA Mismatch Repair/genetics , DNA Polymerase II/genetics , DNA Polymerase II/isolation & purification , DNA Polymerase III/genetics , DNA Polymerase III/isolation & purification , DNA Replication/genetics , Holoenzymes , Humans , Molecular Sequence Data , Mutation/genetics , Simplexvirus/genetics , Thymidine Kinase/genetics
13.
PLoS Negl Trop Dis ; 4(8): e786, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20706627

ABSTRACT

BACKGROUND: Eukaryotic family A DNA polymerases are involved in mitochondrial DNA replication or translesion DNA synthesis. Here, we present evidence that the sole family A DNA polymerase from the parasite protozoan E. histolytica (EhDNApolA) localizes to the nucleus and that its biochemical properties indicate that this DNA polymerase may be involved in translesion DNA synthesis. METHODOLOGY AND RESULTS: EhDNApolA is the sole family A DNA polymerase in E. histolytica. An in silico analysis places family A DNA polymerases from the genus Entamoeba in a separate branch of a family A DNA polymerases phylogenetic tree. Biochemical studies of a purified recombinant EhDNApolA demonstrated that this polymerase is active in primer elongation, is poorly processive, displays moderate strand displacement, and does not contain 3'-5' exonuclease or editing activity. Importantly, EhDNApolA bypasses thymine glycol lesions with high fidelity, and confocal microscopy demonstrates that this polymerase is translocated into the nucleus. These data suggest a putative role of EhDNApolA in translesion DNA synthesis in E. histolytica. CONCLUSION: This is the first report of the biochemical characterization of a DNA polymerase from E. histolytica. EhDNApolA is a family A DNA polymerase that is grouped into a new subfamily of DNA polymerases with translesion DNA synthesis capabilities similar to DNA polymerases from subfamily nu.


Subject(s)
DNA Polymerase III/metabolism , Entamoeba histolytica/enzymology , Protozoan Proteins/metabolism , Thymine/analogs & derivatives , Cell Nucleus/chemistry , DNA Polymerase III/isolation & purification , Models, Molecular , Phylogeny , Protein Structure, Tertiary , Protozoan Proteins/isolation & purification , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Homology , Thymine/metabolism
14.
Methods Mol Biol ; 521: 345-59, 2009.
Article in English | MEDLINE | ID: mdl-19563116

ABSTRACT

This chapter summarizes isolation procedures of four recombinant human proteins crucial for DNA replication: (a) the replicative DNA polymerase (pol) delta, (b) proliferating cell nuclear antigen (PCNA), (c) replication protein A (RP-A), and (d) replication factor C (RF-C). Pol delta is a four-subunit enzyme essential for replication of the lagging strand and possibly of the leading strand. PCNA is a central player important for coordination of the complex network of proteins interacting at the replication fork. RP-A is single-strand DNA-binding protein involved in DNA replication, DNA repair, DNA recombination, and checkpoint control. RF-C as a clamp loader is required for loading of PCNA onto double-stranded DNA and therefore enables PCNA-dependent elongation by pol delta and pol epsilon. To reconstitute the intact pol delta and RF-C, a baculovirus expression system is used, where insect cells are infected with baculoviruses, each coding for one of the four or five subunits of pol delta or RF-C, respectively. We also present two easy methods to isolate the homotrimeric human PCNA and the heterotrimeric human RP-A from an Escherichia coli expression system.


Subject(s)
DNA Polymerase III/isolation & purification , DNA Replication/physiology , Proliferating Cell Nuclear Antigen/isolation & purification , Replication Protein A/isolation & purification , Replication Protein C/isolation & purification , Animals , Baculoviridae/genetics , Cell Line , Chromatography, Affinity/methods , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , Escherichia coli/genetics , Humans , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Protein Subunits , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Replication Protein A/genetics , Replication Protein A/metabolism , Replication Protein C/genetics , Replication Protein C/metabolism , Spodoptera/genetics
15.
Methods Mol Med ; 142: 25-36, 2008.
Article in English | MEDLINE | ID: mdl-18437303

ABSTRACT

The need for new drugs to treat infections caused by antibiotic-resistant bacterial strains has prompted many studies to identify novel targets in pathogenic bacteria. Among the three DNA polymerases expressed by bacteria, one of these, designated pol III, is responsible for DNA replication and growth of bacteria and, therefore, warrants consideration as a drug target. However, the pol III enzymes of Gram-positive and Gram-negative species are quite different, and the Gram-positive enzyme pol IIIC has been more extensively studied as a drug target than the Gram-negative enzyme pol IIIE.DNA polymerases are unique enzymes with respect to the five substrates (four dNTPs, one of which is radiolabeled, and primer:template DNA) that they typically utilize. Variations of the assay, e.g., by leaving out one dNTP but allowing measurable incorporation of the remaining substrates, or use of homopolymer primer:templates, may be used to simplify the assay or to obtain mechanistic information about inhibitors. Use of gel analysis of primer extension assays can also be applied to study alternate substrates of DNA polymerases. Methods to isolate pol IIIC from Gram-positive bacterial cells and to clone and express the polC gene are described in this chapter. In addition, the assay conditions commonly used to identify and study the mechanism of inhibitors of pol IIIC are emphasized.


Subject(s)
Bacillus subtilis/enzymology , DNA Polymerase III/antagonists & inhibitors , DNA Polymerase III/metabolism , Enzyme Inhibitors/analysis , Animals , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Cattle , Cloning, Molecular , DNA/analysis , DNA Polymerase III/genetics , DNA Polymerase III/isolation & purification , DNA Primers/analysis , DNA Primers/chemical synthesis
16.
J Virol ; 81(11): 5777-87, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17360744

ABSTRACT

Adeno-associated virus (AAV) replicates its DNA by a modified rolling-circle mechanism that exclusively uses leading strand displacement synthesis. To identify the enzymes directly involved in AAV DNA replication, we fractionated adenovirus-infected crude extracts and tested them in an in vitro replication system that required the presence of the AAV-encoded Rep protein and the AAV origins of DNA replication, thus faithfully reproducing in vivo viral DNA replication. Fractions that contained replication factor C (RFC) and proliferating cell nuclear antigen (PCNA) were found to be essential for reconstituting AAV DNA replication. These could be replaced by purified PCNA and RFC to retain full activity. We also found that fractions containing polymerase delta, but not polymerase epsilon or alpha, were capable of replicating AAV DNA in vitro. This was confirmed when highly purified polymerase delta complex purified from baculovirus expression clones was used. Curiously, as the components of the DNA replication system were purified, neither the cellular single-stranded DNA binding protein (RPA) nor the adenovirus-encoded DNA binding protein was found to be essential for DNA replication; both only modestly stimulated DNA synthesis on an AAV template. Also, in addition to polymerase delta, RFC, and PCNA, an as yet unidentified factor(s) is required for AAV DNA replication, which appeared to be enriched in adenovirus-infected cells. Finally, the absence of any apparent cellular DNA helicase requirement led us to develop an artificial AAV replication system in which polymerase delta, RFC, and PCNA were replaced with T4 DNA polymerase and gp32 protein. This system was capable of supporting AAV DNA replication, demonstrating that under some conditions the Rep helicase activity can function to unwind duplex DNA during strand displacement synthesis.


Subject(s)
DNA Polymerase III/isolation & purification , DNA Polymerase III/physiology , DNA, Viral/biosynthesis , Dependovirus/genetics , Animals , Cell Line , Cellulose/analogs & derivatives , Cellulose/chemistry , Chromatography, Agarose , Humans , Mice , Proliferating Cell Nuclear Antigen/isolation & purification , Proliferating Cell Nuclear Antigen/physiology , Replication Protein C/isolation & purification , Replication Protein C/physiology
17.
Cell Cycle ; 5(4): 428-36, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16479163

ABSTRACT

Using a yeast two-hybrid screening technique and the p50 subunit of human DNA polymerase delta (pol delta) as a bait, p21 was found to interact with the p50 subunit of pol delta. A direct interaction between p21 and p50 was confirmed by using ELISA and pull-down assays with purified proteins. The interaction sites between p50 and p21 were mapped by pull down assays with GST deletion mutants. Residues 127-193 constitute the primary interaction region on p50 to which p21 binds, while p50 binds to the C-terminal 26 residues of p21. A histone kinase activity was associated with the highly purified calf thymus pol delta and addition of purified recombinant p21 inhibited the kinase activity in a dose dependent manner. p50 is phosphorylated in vivo and can be phosphorylated by CDK2/cyclinA in vitro. In vivo evidence of p21 association with p50 was obtained by coimmunoprecipitation using MCF7 cells. It was also shown that the association of p21 with p50 and other components of the pol delta complex increased in MCF7 cells treated with adriamycin. Our results suggested that p50 might target or anchor p21 to pol delta complex upon certain DNA damage such as adriamycin treatment.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Polymerase III/chemistry , DNA Polymerase III/metabolism , Protein Subunits/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclins/metabolism , DNA Polymerase III/isolation & purification , Doxorubicin/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Phosphoproteins/metabolism , Phosphorylation , Protamine Kinase/metabolism , Protein Binding/drug effects , Protein Interaction Mapping , Protein Subunits/chemistry
18.
J Biol Chem ; 280(35): 31303-13, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-15995249

ABSTRACT

Nuclear DNA helicase II (NDH II), alternatively named RNA helicase A, is involved in transcription and RNA processing. Here, we report that NDH II interacts with the Werner syndrome helicase WRN, an enzyme associated with premature aging and predisposition to tumorigenesis. NDH II was co-purified with WRN, DNA polymerase delta, and replication protein A (70 kDa) during several steps of conventional column chromatography. Co-immunoprecipitations revealed an association between NDH II, WRN, and polymerase delta. We demonstrate a direct protein-protein interaction between WRN and NDH II that is mediated by the N-terminal double-strand RNA-binding domain II and C-terminal RGG box of NDH II and the N-terminal exonuclease domain of WRN. WRN inhibited the DNA-dependent NTPase and DNA helicase activities of NDH II. On the other hand, the 3' --> 5' exonuclease activity of WRN was increased by the presence of NDH II. NDH II directly stimulated the exonuclease domain of WRN, whereas the exonuclease domain of WRN suppressed the DNA-dependent (but not RNA-dependent) ATPase activity of NDH II. These results suggest that the double-strand RNA-binding domain II and RGG box of NDH II together form a protein-protein interaction surface that contacts the exonuclease domain of WRN. Furthermore, NDH II enhanced the degradation of D-loop DNA by the WRN exonuclease. Taken together, these results suggest that NDH II plays a role in promoting the DNA processing function of WRN, which in turn might be necessary for maintaining genomic stability.


Subject(s)
DNA Helicases/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , DEAD-box RNA Helicases , DNA/chemistry , DNA/metabolism , DNA Helicases/genetics , DNA Helicases/isolation & purification , DNA Polymerase III/genetics , DNA Polymerase III/isolation & purification , DNA Polymerase III/metabolism , Enzyme Activation , Exodeoxyribonucleases , HeLa Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Neoplasm Proteins/genetics , Neoplasm Proteins/isolation & purification , Protein Structure, Tertiary , RecQ Helicases , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Werner Syndrome/enzymology , Werner Syndrome/physiopathology , Werner Syndrome Helicase
19.
Acta Crystallogr D Biol Crystallogr ; 59(Pt 7): 1192-9, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12832762

ABSTRACT

The beta subunit of the Escherichia coli replicative DNA polymerase III holoenzyme is the sliding clamp that interacts with the alpha (polymerase) subunit to maintain the high processivity of the enzyme. The beta protein is a ring-shaped dimer of 40.6 kDa subunits whose structure has previously been determined at a resolution of 2.5 A [Kong et al. (1992), Cell, 69, 425-437]. Here, the construction of a new plasmid that directs overproduction of beta to very high levels and a simple procedure for large-scale purification of the protein are described. Crystals grown under slightly modified conditions diffracted to beyond 1.9 A at 100 K at a synchrotron source. The structure of the beta dimer solved at 1.85 A resolution shows some differences from that reported previously. In particular, it was possible at this resolution to identify residues that differed in position between the two subunits in the unit cell; side chains of these and some other residues were found to occupy alternate conformations. This suggests that these residues are likely to be relatively mobile in solution. Some implications of this flexibility for the function of beta are discussed.


Subject(s)
DNA Polymerase III/chemistry , Protein Subunits/chemistry , Amino Acid Sequence , Crystallography, X-Ray/methods , DNA Polymerase III/isolation & purification , DNA Polymerase III/metabolism , DNA Replication , Dimerization , Escherichia coli Proteins/chemistry , Motion , Pliability , Protein Binding , Protein Conformation , Sequence Alignment
20.
Protein Expr Purif ; 27(1): 90-7, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12509989

ABSTRACT

Enterococcus faecalis (Ef) dnaE and polC, the respective genes encoding the DNA replication-specific DNA polymerase III E and DNA polymerase III C, were cloned and engineered for expression in Escherichia coli as hexahistidine (his6)-tagged recombinant proteins. Each gene expressed a catalytically active DNA polymerase of the expected molecular weight. The recombinant polymerases were purified and each was characterized with respect to catalytic properties, inhibitor sensitivity, and recognition by specific antibody raised against the corresponding DNA polymerase III of the model Gram-positive (Gr(+)) organism, Bacillus subtilis (Bs). In conclusion, the properties of each Enterococcus polymerase enzymes were similar to those of the respective B. subtilis enzymes.


Subject(s)
Bacterial Proteins , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , DNA-Directed DNA Polymerase/genetics , Enterococcus faecalis/enzymology , Genes, Bacterial/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA Polymerase III/chemistry , DNA Polymerase III/isolation & purification , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/isolation & purification , DNA-Directed DNA Polymerase/metabolism , Enterococcus faecalis/genetics , Enzyme Inhibitors/pharmacology , Escherichia coli , Gene Expression , Guanosine Triphosphate/analogs & derivatives , Guanosine Triphosphate/pharmacology , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...