Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
DNA Repair (Amst) ; 133: 103605, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38042029

ABSTRACT

Oxidative damage is a major source of genomic instability in all organisms with the aerobic metabolism. 8-Oxoguanine (8-oxoG), an abundant oxidized purine, is mutagenic and must be controlled by a dedicated DNA repair system (GO system) that prevents G:C→T:A transversions through an easily formed 8-oxoG:A mispair. In some forms, the GO system is present in nearly all cellular organisms. However, recent studies uncovered many instances of viruses possessing non-canonical nucleotides in their genomes. The features of genome damage and maintenance in such cases of alternative genetic chemistry remain barely explored. In particular, 2,6-diaminopurine (Z nucleotide) completely substitutes for A in the genomes of some bacteriophages, which have evolved pathways for dZTP synthesis and specialized polymerases that prefer dZTP over dATP. Here we address the ability of the GO system enzymes to cope with oxidative DNA damage in the presence of Z in DNA. DNA polymerases of two different structural families (Klenow fragment and RB69 polymerase) were able to incorporate dZMP opposite to 8-oxoG in the template, as well as 8-oxodGMP opposite to Z in the template. Fpg, a 8-oxoguanine-DNA glycosylase that discriminates against 8-oxoG:A mispairs, also did not remove 8-oxoG from 8-oxoG:Z mispairs. However, MutY, a DNA glycosylase that excises A from pairs with 8-oxoG, had a significantly lower activity on Z:8-oxoG mispairs. Similar preferences were observed for Fpg and MutY from different bacterial species (Escherichia coli, Staphylococcus aureus and Lactococcus lactis). Overall, the relaxed control of 8-oxoG in the presence of the Z nucleotide may be a source of additional mutagenesis in the genomes of bacteriophages or bacteria that have survived the viral invasion.


Subject(s)
DNA Glycosylases , Nucleotides , Humans , Nucleotides/metabolism , DNA Repair , DNA Glycosylases/metabolism , Oxidative Stress , DNA Damage , Escherichia coli/metabolism , Genome, Viral
2.
Int J Mol Sci ; 24(11)2023 May 23.
Article in English | MEDLINE | ID: mdl-37298065

ABSTRACT

The protein encoded by the vaccinia virus D4R gene has base excision repair uracil-DNA N-glycosylase (vvUNG) activity and also acts as a processivity factor in the viral replication complex. The use of a protein unlike PolN/PCNA sliding clamps is a unique feature of orthopoxviral replication, providing an attractive target for drug design. However, the intrinsic processivity of vvUNG has never been estimated, leaving open the question whether it is sufficient to impart processivity to the viral polymerase. Here, we use the correlated cleavage assay to characterize the translocation of vvUNG along DNA between two uracil residues. The salt dependence of the correlated cleavage, together with the similar affinity of vvUNG for damaged and undamaged DNA, support the one-dimensional diffusion mechanism of lesion search. Unlike short gaps, covalent adducts partly block vvUNG translocation. Kinetic experiments show that once a lesion is found it is excised with a probability ~0.76. Varying the distance between two uracils, we use a random walk model to estimate the mean number of steps per association with DNA at ~4200, which is consistent with vvUNG playing a role as a processivity factor. Finally, we show that inhibitors carrying a tetrahydro-2,4,6-trioxopyrimidinylidene moiety can suppress the processivity of vvUNG.


Subject(s)
Uracil-DNA Glycosidase , Vaccinia virus , Uracil-DNA Glycosidase/genetics , Uracil-DNA Glycosidase/metabolism , Vaccinia virus/genetics , DNA/metabolism , Virus Replication , DNA Repair , Uracil/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , DNA Replication
3.
Int J Mol Sci ; 25(1)2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38203235

ABSTRACT

Apurinic/apyrimidinic (AP) sites are abundant DNA lesions generated both by spontaneous base loss and as intermediates of base excision DNA repair. In human cells, they are normally repaired by an essential AP endonuclease, APE1, encoded by the APEX1 gene. Other enzymes can cleave AP sites by either hydrolysis or ß-elimination in vitro, but it is not clear whether they provide the second line of defense in living cells. Here, we studied AP site repairs in APEX1 knockout derivatives of HEK293FT cells using a reporter system based on transcriptional mutagenesis in the enhanced green fluorescent protein gene. Despite an apparent lack of AP site-processing activity in vitro, the cells efficiently repaired the tetrahydrofuran AP site analog resistant to ß-elimination. This ability persisted even when the second AP endonuclease homolog, APE2, was also knocked out. Moreover, APEX1 null cells were able to repair uracil, a DNA lesion that is removed via the formation of an AP site. If AP site hydrolysis was chemically blocked, the uracil repair required the presence of NTHL1, an enzyme that catalyzes ß-elimination. Our results suggest that human cells possess at least two back-up AP site repair pathways, one of which is NTHL1-dependent.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase , DNA , Humans , DNA Damage/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Endonucleases , Excision Repair , Uracil
4.
Cells ; 11(20)2022 10 11.
Article in English | MEDLINE | ID: mdl-36291061

ABSTRACT

Proteins that recognize specific DNA sequences or structural elements often find their cognate DNA lesions in a processive mode, in which an enzyme binds DNA non-specifically and then slides along the DNA contour by one-dimensional diffusion. Opposite to the processive mechanism is distributive search, when an enzyme binds, samples and releases DNA without significant lateral movement. Many DNA glycosylases, the repair enzymes that excise damaged bases from DNA, use processive search to find their cognate lesions. Here, using a method based on correlated cleavage of multiply damaged oligonucleotide substrates we investigate the mechanism of lesion search by three structurally related DNA glycosylases-bacterial endonuclease VIII (Nei) and its mammalian homologs NEIL1 and NEIL2. Similarly to another homologous enzyme, bacterial formamidopyrimidine-DNA glycosylase, NEIL1 seems to use a processive mode to locate its targets. However, the processivity of Nei was notably lower, and NEIL2 exhibited almost fully distributive action on all types of substrates. Although one-dimensional diffusion is often regarded as a universal search mechanism, our results indicate that even proteins sharing a common fold may be quite different in the ways they locate their targets in DNA.


Subject(s)
DNA Glycosylases , Deoxyribonuclease (Pyrimidine Dimer) , Animals , Deoxyribonuclease (Pyrimidine Dimer)/genetics , Deoxyribonuclease (Pyrimidine Dimer)/metabolism , DNA-Formamidopyrimidine Glycosylase/genetics , DNA-Formamidopyrimidine Glycosylase/metabolism , DNA Repair , DNA Glycosylases/genetics , DNA/metabolism , Oligonucleotides , Mammals/metabolism
5.
Int J Mol Sci ; 23(13)2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35806289

ABSTRACT

Many proteins consist of two or more structural domains: separate parts that have a defined structure and function. For example, in enzymes, the catalytic activity is often localized in a core fragment, while other domains or disordered parts of the same protein participate in a number of regulatory processes. This situation is often observed in many DNA glycosylases, the proteins that remove damaged nucleobases thus initiating base excision DNA repair. This review covers the present knowledge about the functions and evolution of such noncatalytic parts in DNA glycosylases, mostly concerned with the human enzymes but also considering some unique members of this group coming from plants and prokaryotes.


Subject(s)
DNA Glycosylases , DNA/chemistry , DNA Damage , DNA Glycosylases/metabolism , DNA Repair , Humans
6.
Molecules ; 26(21)2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34771075

ABSTRACT

Uracil-DNA glycosylases are enzymes that excise uracil bases appearing in DNA as a result of cytosine deamination or accidental dUMP incorporation from the dUTP pool. The activity of Family 1 uracil-DNA glycosylase (UNG) activity limits the efficiency of antimetabolite drugs and is essential for virulence in some bacterial and viral infections. Thus, UNG is regarded as a promising target for antitumor, antiviral, antibacterial, and antiprotozoal drugs. Most UNG inhibitors presently developed are based on the uracil base linked to various substituents, yet new pharmacophores are wanted to target a wide range of UNGs. We have conducted virtual screening of a 1,027,767-ligand library and biochemically screened the best hits for the inhibitory activity against human and vaccinia virus UNG enzymes. Although even the best inhibitors had IC50 ≥ 100 µM, they were highly enriched in a common fragment, tetrahydro-2,4,6-trioxopyrimidinylidene (PyO3). In silico, PyO3 preferably docked into the enzyme's active site, and in kinetic experiments, the inhibition was better consistent with the competitive mechanism. The toxicity of two best inhibitors for human cells was independent of the presence of methotrexate, which is consistent with the hypothesis that dUMP in genomic DNA is less toxic for the cell than strand breaks arising from the massive removal of uracil. We conclude that PyO3 may be a novel pharmacophore with the potential for development into UNG-targeting agents.


Subject(s)
Enzyme Inhibitors/pharmacology , Pyrimidines/pharmacology , Uracil-DNA Glycosidase/antagonists & inhibitors , Vaccinia virus/enzymology , Enzyme Inhibitors/chemistry , Humans , Kinetics , Ligands , Molecular Docking Simulation , Molecular Structure , Pyrimidines/chemistry , Uracil-DNA Glycosidase/metabolism
7.
J Biol Chem ; 296: 100229, 2021.
Article in English | MEDLINE | ID: mdl-33361155

ABSTRACT

DNA of living cells is always exposed to damaging factors. To counteract the consequences of DNA lesions, cells have evolved several DNA repair systems, among which base excision repair is one of the most important systems. Many currently used antitumor drugs act by damaging DNA, and DNA repair often interferes with chemotherapy and radiotherapy in cancer cells. Tumors are usually extremely genetically heterogeneous, often bearing mutations in DNA repair genes. Thus, knowledge of the functionality of cancer-related variants of proteins involved in DNA damage response and repair is of great interest for personalization of cancer therapy. Although computational methods to predict the variant functionality have attracted much attention, at present, they are mostly based on sequence conservation and make little use of modern capabilities in computational analysis of 3D protein structures. We have used molecular dynamics (MD) to model the structures of 20 clinically observed variants of a DNA repair enzyme, 8-oxoguanine DNA glycosylase. In parallel, we have experimentally characterized the activity, thermostability, and DNA binding in a subset of these mutant proteins. Among the analyzed variants of 8-oxoguanine DNA glycosylase, three (I145M, G202C, and V267M) were significantly functionally impaired and were successfully predicted by MD. Alone or in combination with sequence-based methods, MD may be an important functional prediction tool for cancer-related protein variants of unknown significance.


Subject(s)
DNA Glycosylases/chemistry , DNA Repair , DNA, Neoplasm/chemistry , Guanine/analogs & derivatives , Mutation , Neoplasm Proteins/chemistry , Amino Acid Substitution , Binding Sites , DNA Damage , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Gene Expression , Guanine/chemistry , Guanine/metabolism , Humans , Kinetics , Leukemia/enzymology , Leukemia/genetics , Leukemia/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Molecular Dynamics Simulation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Principal Component Analysis , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Small Cell Lung Carcinoma/enzymology , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology
8.
Int J Mol Sci ; 21(9)2020 Apr 28.
Article in English | MEDLINE | ID: mdl-32354123

ABSTRACT

DNA glycosylases are enzymes that initiate the base excision repair pathway, a major biochemical process that protects the genomes of all living organisms from intrinsically and environmentally inflicted damage. Recently, base excision repair inhibition proved to be a viable strategy for the therapy of tumors that have lost alternative repair pathways, such as BRCA-deficient cancers sensitive to poly(ADP-ribose)polymerase inhibition. However, drugs targeting DNA glycosylases are still in development and so far have not advanced to clinical trials. In this review, we cover the attempts to validate DNA glycosylases as suitable targets for inhibition in the pharmacological treatment of cancer, neurodegenerative diseases, chronic inflammation, bacterial and viral infections. We discuss the glycosylase inhibitors described so far and survey the advances in the assays for DNA glycosylase reactions that may be used to screen pharmacological libraries for new active compounds.


Subject(s)
DNA Glycosylases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Bacterial Infections/drug therapy , Bacterial Infections/enzymology , DNA Repair , Drug Discovery , Enzyme Inhibitors/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/enzymology , Neoplasms/drug therapy , Neoplasms/enzymology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/enzymology , Small Molecule Libraries/therapeutic use , Virus Diseases/drug therapy , Virus Diseases/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...