Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.451
Filter
1.
G3 (Bethesda) ; 14(9)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39120426

ABSTRACT

Whole genome duplications are implicated in genome instability and tumorigenesis. Human and yeast polyploids exhibit increased replication stress and chromosomal instability, both hallmarks of cancer. In this study, we investigate the transcriptional response of Schizosaccharomyces pombe to increased ploidy generally, and in response to treatment with the genotoxin methyl methanesulfonate (MMS). We find that treatment of MMS induces upregulation of genes involved in general response to genotoxins, in addition to cell cycle regulatory genes. Downregulated genes are enriched in transport and sexual reproductive pathways. We find that the diploid response to MMS is muted compared to the haploid response, although the enriched pathways remain largely the same. Overall, our data suggests that the global S. pombe transcriptome doubles in response to increased ploidy but undergoes modest transcriptional changes in both unperturbed and genotoxic stress conditions.


Subject(s)
DNA Damage , Diploidy , Gene Expression Regulation, Fungal , Haploidy , Schizosaccharomyces , Schizosaccharomyces/genetics , Schizosaccharomyces/drug effects , Methyl Methanesulfonate/pharmacology , Transcriptome , Transcription, Genetic , Gene Expression Profiling , Mutagens/toxicity , Mutagens/pharmacology
2.
DNA Repair (Amst) ; 141: 103732, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39094381

ABSTRACT

The human DNA repair enzyme AlkB homologue-2 (ALKBH2) repairs methyl adducts from genomic DNA and is overexpressed in several cancers. However, there are no known inhibitors available for this crucial DNA repair enzyme. The aim of this study was to examine whether the first-generation HIV protease inhibitors having strong anti-cancer activity can be repurposed as inhibitors of ALKBH2. We selected four such inhibitors and performed in vitro binding analysis against ALKBH2 based on alterations of its intrinsic tryptophan fluorescence and differential scanning fluorimetry. The effect of these HIV protease inhibitors on the DNA repair activity of ALKBH2 was also evaluated. Interestingly, we observed that one of the inhibitors, ritonavir, could inhibit ALKBH2-mediated DNA repair significantly via competitive inhibition and sensitized cancer cells to alkylating agent methylmethane sulfonate (MMS). This work may provide new insights into the possibilities of utilizing HIV protease inhibitor ritonavir as a DNA repair antagonist.


Subject(s)
AlkB Homolog 2, Alpha-Ketoglutarate-Dependent Dioxygenase , DNA Repair , HIV Protease Inhibitors , Methyl Methanesulfonate , Ritonavir , Humans , AlkB Homolog 2, Alpha-Ketoglutarate-Dependent Dioxygenase/metabolism , Ritonavir/pharmacology , HIV Protease Inhibitors/pharmacology , Methyl Methanesulfonate/pharmacology , DNA Damage , Alkylation , Cell Line, Tumor
3.
PLoS Genet ; 20(7): e1011181, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39074150

ABSTRACT

When replication forks encounter damaged DNA, cells utilize damage tolerance mechanisms to allow replication to proceed. These include translesion synthesis at the fork, postreplication gap filling, and template switching via fork reversal or homologous recombination. The extent to which these different damage tolerance mechanisms are utilized depends on cell, tissue, and developmental context-specific cues, the last two of which are poorly understood. To address this gap, we have investigated damage tolerance responses in Drosophila melanogaster. We report that tolerance of DNA alkylation damage in rapidly dividing larval tissues depends heavily on translesion synthesis. Furthermore, we show that the REV1 protein plays a multi-faceted role in damage tolerance in Drosophila. Larvae lacking REV1 are hypersensitive to methyl methanesulfonate (MMS) and have highly elevated levels of γ-H2Av (Drosophila γ-H2AX) foci and chromosome aberrations in MMS-treated tissues. Loss of the REV1 C-terminal domain (CTD), which recruits multiple translesion polymerases to damage sites, sensitizes flies to MMS. In the absence of the REV1 CTD, DNA polymerases eta and zeta become critical for MMS tolerance. In addition, flies lacking REV3, the catalytic subunit of polymerase zeta, require the deoxycytidyl transferase activity of REV1 to tolerate MMS. Together, our results demonstrate that Drosophila prioritize the use of multiple translesion polymerases to tolerate alkylation damage and highlight the critical role of REV1 in the coordination of this response to prevent genome instability.


Subject(s)
DNA Damage , DNA Repair , DNA Replication , DNA-Directed DNA Polymerase , Drosophila Proteins , Drosophila melanogaster , Methyl Methanesulfonate , Nucleotidyltransferases , Animals , Drosophila melanogaster/genetics , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Methyl Methanesulfonate/pharmacology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Alkylation , DNA Repair/genetics , DNA Replication/genetics , Larva/genetics , Histones/metabolism , Histones/genetics
4.
Chem Res Toxicol ; 37(5): 814-823, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38652696

ABSTRACT

The major product of DNA-methylating agents, N7-methyl-2'-deoxyguanosine (MdG), is a persistent lesion in vivo, but it is not believed to have a large direct physiological impact. However, MdG reacts with histone proteins to form reversible DNA-protein cross-links (DPCMdG), a family of DNA lesions that can significantly threaten cell survival. In this paper, we developed a tandem mass spectrometry method for quantifying the amounts of MdG and DPCMdG in nuclear DNA by taking advantage of their chemical lability and the concurrent release of N7-methylguanine. Using this method, we determined that DPCMdG is formed in less than 1% yield based upon the levels of MdG in methyl methanesulfonate (MMS)-treated HeLa cells. Despite its low chemical yield, DPCMdG contributes to MMS cytotoxicity. Consequently, cells that lack efficient DPC repair by the DPC protease SPRTN are hypersensitive to MMS. This investigation shows that the downstream chemical and biochemical effects of initially formed DNA damage can have significant biological consequences. With respect to MdG formation, the initial DNA lesion is only the beginning.


Subject(s)
DNA , Deoxyguanosine , Methyl Methanesulfonate , Humans , HeLa Cells , DNA/metabolism , DNA/chemistry , DNA/drug effects , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Deoxyguanosine/chemistry , Methyl Methanesulfonate/chemistry , Methyl Methanesulfonate/pharmacology , Tandem Mass Spectrometry , Cell Survival/drug effects , DNA Damage/drug effects , Cross-Linking Reagents/chemistry , DNA-Binding Proteins
5.
G3 (Bethesda) ; 14(3)2024 03 06.
Article in English | MEDLINE | ID: mdl-38261383

ABSTRACT

We performed a functional analysis of two potential partners of ASF1, a highly conserved histone chaperone that plays a crucial role in the sexual development and DNA damage resistance in the ascomycete Sordaria macrospora. ASF1 is known to be involved in nucleosome assembly and disassembly, binding histones H3 and H4 during transcription, replication and DNA repair and has direct and indirect roles in histone recycling and modification as well as DNA methylation, acting as a chromatin modifier hub for a large network of chromatin-associated proteins. Here, we functionally characterized two of these proteins, RTT109 and CHK2. RTT109 is a fungal-specific histone acetyltransferase, while CHK2 is an ortholog to PRD-4, a checkpoint kinase of Neurospora crassa that performs similar cell cycle checkpoint functions as yeast RAD53. Through the generation and characterization of deletion mutants, we discovered striking similarities between RTT109 and ASF1 in terms of their contributions to sexual development, histone acetylation, and protection against DNA damage. Phenotypic observations revealed a developmental arrest at the same stage in Δrtt109 and Δasf1 strains, accompanied by a loss of H3K56 acetylation, as detected by western blot analysis. Deletion mutants of rtt109 and asf1 are sensitive to the DNA damaging agent methyl methanesulfonate, but not hydroxyurea. In contrast, chk2 mutants are fertile and resistant to methyl methanesulfonate, but not hydroxyurea. Our findings suggest a close functional association between ASF1 and RTT109 in the context of development, histone modification, and DNA damage response, while indicating a role for CHK2 in separate pathways of the DNA damage response.


Subject(s)
Histones , Saccharomyces cerevisiae Proteins , Sordariales , Histones/metabolism , Methyl Methanesulfonate/pharmacology , Molecular Chaperones/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , DNA Repair , DNA Damage , Chromatin/genetics , Chromatin/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Histone Acetyltransferases/metabolism , Acetylation
6.
Cells ; 12(15)2023 07 31.
Article in English | MEDLINE | ID: mdl-37566058

ABSTRACT

The comet assay in Drosophila has been used in the last few years to study DNA damage responses (DDR) in different repair-mutant strains and to compare them to analyze DNA repair. We have used this approach to study interactions between DNA repair pathways in vivo. Additionally, we have implemented an ex vivo comet assay, in which nucleoids from treated and untreated cells were incubated ex vivo with cell-free protein extracts from individuals with distinct repair capacities. Four strains were used: wild-type OregonK (OK), nucleotide excision repair mutant mus201, dmPolQ protein mutant mus308, and the double mutant mus201;mus308. Methyl methanesulfonate (MMS) was used as a genotoxic agent. Both approaches were performed with neuroblasts from third-instar larvae; they detected the effects of the NER and dmPolQ pathways on the DDR to MMS and that they act additively in this response. Additionally, the ex vivo approach quantified that mus201, mus308, and the double mutant mus201;mus308 strains presented, respectively, 21.5%, 52.9%, and 14.8% of OK strain activity over MMS-induced damage. Considering the homology between mammals and Drosophila in repair pathways, the detected additive effect might be extrapolated even to humans, demonstrating that Drosophila might be an excellent model to study interactions between repair pathways.


Subject(s)
Drosophila melanogaster , Drosophila , Humans , Animals , Comet Assay , Drosophila/genetics , Drosophila melanogaster/genetics , DNA Repair , DNA Damage , Methyl Methanesulfonate/pharmacology , Mammals/genetics
7.
Mol Microbiol ; 120(4): 587-607, 2023 10.
Article in English | MEDLINE | ID: mdl-37649278

ABSTRACT

Saccharomyces cerevisiae Pso2/SNM1 is essential for DNA interstrand crosslink (ICL) repair; however, its mechanism of action remains incompletely understood. While recent work has revealed that Pso2/Snm1 is dual-localized in the nucleus and mitochondria, it remains unclear whether cell-intrinsic and -extrinsic factors regulate its subcellular localization and function. Herein, we show that Pso2 undergoes ubiquitination and phosphorylation, but not SUMOylation, in unstressed cells. Unexpectedly, we found that methyl methanesulfonate (MMS), rather than ICL-forming agents, induced robust SUMOylation of Pso2 on two conserved residues, K97 and K575, and that SUMOylation markedly increased its abundance in the mitochondria. Reciprocally, SUMOylation had no discernible impact on Pso2 translocation to the nucleus, despite the presence of steady-state levels of SUMOylated Pso2 across the cell cycle. Furthermore, substitution of the invariant residues K97 and K575 by arginine in the Pso2 SUMO consensus motifs severely impaired SUMOylation and abolished its translocation to the mitochondria of MMS-treated wild type cells, but not in unstressed cells. We demonstrate that whilst Siz1 and Siz2 SUMO E3 ligases catalyze Pso2 SUMOylation, the former plays a dominant role. Notably, we found that the phenotypic characteristics of the SUMOylation-defective mutant Pso2K97R/K575R closely mirrored those observed in the Pso2Δ petite mutant. Additionally, leveraging next-generation sequencing analysis, we demonstrate that Pso2 mitigates MMS-induced damage to mitochondrial DNA (mtDNA). Viewed together, our work offers previously unknown insights into the link between genotoxic stress-induced SUMOylation of Pso2 and its preferential targeting to the mitochondria, as well as its role in attenuating MMS-induced mtDNA damage.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Humans , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Methyl Methanesulfonate/pharmacology , Methyl Methanesulfonate/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Sumoylation , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Endodeoxyribonucleases/metabolism , DNA Damage , Mitochondria/metabolism , Translocation, Genetic , Ubiquitin-Protein Ligases/metabolism
8.
Elife ; 122023 06 06.
Article in English | MEDLINE | ID: mdl-37278514

ABSTRACT

The replication checkpoint is essential for accurate DNA replication and repair, and maintenance of genomic integrity when a cell is challenged with genotoxic stress. Several studies have defined the complement of proteins that change subcellular location in the budding yeast Saccharomyces cerevisiae following chemically induced DNA replication stress using methyl methanesulfonate (MMS) or hydroxyurea (HU). How these protein movements are regulated remains largely unexplored. We find that the essential checkpoint kinases Mec1 and Rad53 are responsible for regulating the subcellular localization of 159 proteins during MMS-induced replication stress. Unexpectedly, Rad53 regulation of the localization of 52 proteins is independent of its known kinase activator Mec1, and in some scenarios independent of Tel1 or the mediator proteins Rad9 and Mrc1. We demonstrate that Rad53 is phosphorylated and active following MMS exposure in cells lacking Mec1 and Tel1. This noncanonical mode of Rad53 activation depends partly on the retrograde signaling transcription factor Rtg3, which also facilitates proper DNA replication dynamics. We conclude that there are biologically important modes of Rad53 protein kinase activation that respond to replication stress and operate in parallel to Mec1 and Tel1.


Subject(s)
Protein Serine-Threonine Kinases , Saccharomyces cerevisiae Proteins , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Saccharomyces cerevisiae/metabolism , Phosphorylation , DNA Damage , Methyl Methanesulfonate/pharmacology , DNA Replication
9.
Int J Mol Sci ; 23(14)2022 Jul 07.
Article in English | MEDLINE | ID: mdl-35886903

ABSTRACT

The infection of a mammalian host by the pathogenic fungus Candida albicans involves fungal resistance to reactive oxygen species (ROS)-induced DNA damage stress generated by the defending macrophages or neutrophils. Thus, the DNA damage response in C. albicans may contribute to its pathogenicity. Uncovering the transcriptional changes triggered by the DNA damage-inducing agent MMS in many model organisms has enhanced the understanding of their DNA damage response processes. However, the transcriptional regulation triggered by MMS remains unclear in C. albicans. Here, we explored the global transcription profile in response to MMS in C. albicans and identified 306 defined genes whose transcription was significantly affected by MMS. Only a few MMS-responsive genes, such as MGT1, DDR48, MAG1, and RAD7, showed potential roles in DNA repair. GO term analysis revealed that a large number of induced genes were involved in antioxidation responses, and some downregulated genes were involved in nucleosome packing and IMP biosynthesis. Nevertheless, phenotypic assays revealed that MMS-induced antioxidation gene CAP1 and glutathione metabolism genes GST2 and GST3 showed no direct roles in MMS resistance. Furthermore, the altered transcription of several MMS-responsive genes exhibited RAD53-related regulation. Intriguingly, the transcription profile in response to MMS in C. albicans shared a limited similarity with the pattern in S. cerevisiae, including COX17, PRI2, and MGT1. Overall, C. albicans cells exhibit global transcriptional changes to the DNA damage agent MMS; these findings improve our understanding of this pathogen's DNA damage response pathways.


Subject(s)
Candida albicans , Methyl Methanesulfonate , Actin Capping Proteins/genetics , Actin Capping Proteins/metabolism , Animals , Candida albicans/drug effects , Candida albicans/genetics , Candida albicans/metabolism , DNA Damage/drug effects , DNA-Binding Proteins/metabolism , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Mammals/metabolism , Methyl Methanesulfonate/pharmacology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
10.
Elife ; 112022 03 15.
Article in English | MEDLINE | ID: mdl-35289750

ABSTRACT

DNA base damage arises frequently in living cells and needs to be removed by base excision repair (BER) to prevent mutagenesis and genome instability. Both the formation and repair of base damage occur in chromatin and are conceivably affected by DNA-binding proteins such as transcription factors (TFs). However, to what extent TF binding affects base damage distribution and BER in cells is unclear. Here, we used a genome-wide damage mapping method, N-methylpurine-sequencing (NMP-seq), and characterized alkylation damage distribution and BER at TF binding sites in yeast cells treated with the alkylating agent methyl methanesulfonate (MMS). Our data show that alkylation damage formation was mainly suppressed at the binding sites of yeast TFs ARS binding factor 1 (Abf1) and rDNA enhancer binding protein 1 (Reb1), but individual hotspots with elevated damage levels were also found. Additionally, Abf1 and Reb1 binding strongly inhibits BER in vivo and in vitro, causing slow repair both within the core motif and its adjacent DNA. Repair of ultraviolet (UV) damage by nucleotide excision repair (NER) was also inhibited by TF binding. Interestingly, TF binding inhibits a larger DNA region for NER relative to BER. The observed effects are caused by the TF-DNA interaction, because damage formation and BER can be restored by depletion of Abf1 or Reb1 protein from the nucleus. Thus, our data reveal that TF binding significantly modulates alkylation base damage formation and inhibits repair by the BER pathway. The interplay between base damage formation and BER may play an important role in affecting mutation frequency in gene regulatory regions.


Subject(s)
DNA Repair , Transcription Factors , DNA , DNA Damage , Methyl Methanesulfonate/pharmacology , Transcription Factors/genetics
11.
Genes Cells ; 27(5): 331-344, 2022 May.
Article in English | MEDLINE | ID: mdl-35194903

ABSTRACT

Base excision repair (BER) removes damaged bases by generating single-strand breaks (SSBs), gap-filling by DNA polymerase ß (POLß), and resealing SSBs. A base-damaging agent, methyl methanesulfonate (MMS) is widely used to study BER. BER increases cellular tolerance to MMS, anti-cancer base-damaging drugs, temozolomide, carmustine, and lomustine, and to clinical poly(ADP ribose)polymerase (PARP) poisons, olaparib and talazoparib. The poisons stabilize PARP1/SSB complexes, inhibiting access of BER factors to SSBs. PARP1 and XRCC1 collaboratively promote SSB resealing by recruiting POLß to SSBs, but XRCC1-/- cells are much more sensitive to MMS than PARP1-/- cells. We recently report that the PARP1 loss in XRCC1-/- cells restores their MMS tolerance and conclude that XPCC1 facilitates the release of PARP1 from SSBs by maintaining its autoPARylation. We here show that the PARP1 loss in XRCC1-/- cells also restores their tolerance to the three anti-cancer base-damaging drugs, although they and MMS induce different sets of base damage. We reveal the synthetic lethality of the XRCC1-/- mutation, but not POLß-/- , with olaparib and talazoparib, indicating that XRCC1 is a unique BER factor in suppressing toxic PARP1/SSB complex and can suppress even when PARP1 catalysis is inhibited. In conclusion, XRCC1 suppresses the PARP1/SSB complex via PARP1 catalysis-dependent and independent mechanisms.


Subject(s)
Poisons , Poly(ADP-ribose) Polymerases , Adenosine Diphosphate Ribose , Alkylating Agents , DNA , DNA Damage , DNA Repair , Methyl Methanesulfonate/pharmacology , Phthalazines , Piperazines , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Temozolomide/pharmacology
12.
Protein Expr Purif ; 189: 105967, 2022 01.
Article in English | MEDLINE | ID: mdl-34481085

ABSTRACT

Recombinases are responsible for homologous recombination (HR), proper genome maintenance, and accurate deoxyribonucleic acid (DNA) duplication. Moreover, HR plays a determining role in DNA transaction processes such as DNA replication, repair, recombination, and transcription. Staphylococcus aureus, an opportunistic pathogen, usually causes respiratory infections such as sinusitis, skin infections, and food poisoning. To date, the role of the RecA gene product in S. aureus remains obscure. In this study, we attempted to map the functional properties of the RecA protein. S. aureus expresses the recA gene product in vivo upon exposure to the DNA-damaging agents, ultraviolet radiation, and methyl methanesulfonate. The recombinant purified S. aureus RecA protein displayed strong single-stranded DNA affinity compared to feeble binding to double-stranded DNA. Interestingly, the RecA protein is capable of invasion and formed displacement loops and readily performed strand-exchange activities with an oligonucleotide-based substrate. Notably, the S. aureus RecA protein hydrolyzed the DNA-dependent adenosine triphosphate and cleaved LexA, showing the conserved function of coprotease. This study provides the functional characterization of the S. aureus RecA protein and sheds light on the canonical processes of homologous recombination, which are conserved in the gram-positive foodborne pathogen S. aureus.


Subject(s)
Bacterial Proteins/metabolism , DNA, Single-Stranded/genetics , Rec A Recombinases/genetics , Recombinational DNA Repair , Serine Endopeptidases/metabolism , Staphylococcus aureus/genetics , Adenosine Triphosphate/metabolism , Cloning, Molecular , DNA/genetics , DNA/metabolism , DNA Damage , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , DNA, Single-Stranded/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Kinetics , Methyl Methanesulfonate/pharmacology , Protein Binding , Protein Transport , Rec A Recombinases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Staphylococcus aureus/radiation effects , Thermodynamics , Ultraviolet Rays/adverse effects
13.
Elife ; 102021 11 01.
Article in English | MEDLINE | ID: mdl-34723799

ABSTRACT

Three-methyl cytosine (3meC) are toxic DNA lesions, blocking base pairing. Bacteria and humans express members of the AlkB enzymes family, which directly remove 3meC. However, other organisms, including budding yeast, lack this class of enzymes. It remains an unanswered evolutionary question as to how yeast repairs 3meC, particularly in single-stranded DNA. The yeast Shu complex, a conserved homologous recombination factor, aids in preventing replication-associated mutagenesis from DNA base damaging agents such as methyl methanesulfonate (MMS). We found that MMS-treated Shu complex-deficient cells exhibit a genome-wide increase in A:T and G:C substitutions mutations. The G:C substitutions displayed transcriptional and replicational asymmetries consistent with mutations resulting from 3meC. Ectopic expression of a human AlkB homolog in Shu-deficient yeast rescues MMS-induced growth defects and increased mutagenesis. Thus, our work identifies a novel homologous recombination-based mechanism mediated by the Shu complex for coping with alkylation adducts.


Subject(s)
Homologous Recombination/drug effects , Methyl Methanesulfonate/pharmacology , Mutagens/pharmacology , Saccharomyces cerevisiae/genetics , Alkylation , Mutagenesis , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
14.
Sci Rep ; 11(1): 21791, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34750454

ABSTRACT

The inducible Di-Cre system was used to delete the putative ubiquitin-conjugating enzyme 13 gene (ubc13) of Plasmodium falciparum to study its role in ubiquitylation and the functional consequence during the parasite asexual blood stage. Deletion resulted in a significant reduction of parasite growth in vitro, reduced ubiquitylation of the Lys63 residue of ubiquitin attached to protein substrates, and an increased sensitivity of the parasite to both the mutagen, methyl methanesulfonate and the antimalarial drug dihydroartemisinin (DHA), but not chloroquine. The parasite was also sensitive to the UBC13 inhibitor NSC697923. The data suggest that this gene does code for an ubiquitin conjugating enzyme responsible for K63 ubiquitylation, which is important in DNA repair pathways as was previously demonstrated in other organisms. The increased parasite sensitivity to DHA in the absence of ubc13 function indicates that DHA may act primarily through this pathway and that inhibitors of UBC13 may both enhance the efficacy of this antimalarial drug and directly inhibit parasite growth.


Subject(s)
Antimalarials/pharmacology , Artemisinins/pharmacology , Methyl Methanesulfonate/pharmacology , Mutagens/pharmacology , Plasmodium falciparum/drug effects , Protozoan Proteins/genetics , Ubiquitin-Conjugating Enzymes/genetics , DNA Damage/drug effects , Gene Knockdown Techniques , Humans , Nitrofurans/pharmacology , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protein Structure, Tertiary , Sequence Alignment , Sulfones/pharmacology
15.
PLoS One ; 16(9): e0257473, 2021.
Article in English | MEDLINE | ID: mdl-34529719

ABSTRACT

The major human apurinic/apyrimidinic (AP) site endonuclease, APEX1, is a central player in the base excision DNA repair (BER) pathway and has a role in the regulation of DNA binding by transcription factors. In vertebrates, APEX1 knockouts are embryonic lethal, and only a handful of knockout cell lines are known. To facilitate studies of multiple functions of this protein in human cells, we have used the CRISPR/Cas9 system to knock out the APEX1 gene in a widely used non-cancer hypotriploid HEK 293FT cell line. Two stable knockout lines were obtained, one carrying two single-base deletion alleles and one single-base insertion allele in exon 3, another homozygous in the single-base insertion allele. Both mutations cause a frameshift that leads to premature translation termination before the start of the protein's catalytic domain. Both cell lines totally lacked the APEX1 protein and AP site-cleaving activity, and showed significantly lower levels of the APEX1 transcript. The APEX1-null cells were unable to support BER on uracil- or AP site-containing substrates. Phenotypically, they showed a moderately increased sensitivity to methyl methanesulfonate (MMS; ~2-fold lower EC50 compared with wild-type cells), and their background level of natural AP sites detected by the aldehyde-reactive probe was elevated ~1.5-2-fold. However, the knockout lines retained a nearly wild-type sensitivity to oxidizing agents hydrogen peroxide and potassium bromate. Interestingly, despite the increased MMS cytotoxicity, we observed no additional increase in AP sites in knockout cells upon MMS treatment, which could indicate their conversion into more toxic products in the absence of repair. Overall, the relatively mild cell phenotype in the absence of APEX1-dependent BER suggests that mammalian cells possess mechanisms of tolerance or alternative repair of AP sites. The knockout derivatives of the extensively characterized HEK 293FT cell line may provide a valuable tool for studies of APEX1 in DNA repair and beyond.


Subject(s)
DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , CRISPR-Cas Systems/genetics , Cell Cycle Checkpoints , DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Gene Editing , HEK293 Cells , Humans , Hydrogen Peroxide/chemistry , Methyl Methanesulfonate/pharmacology , Phenotype , RNA, Guide, Kinetoplastida/metabolism
16.
PLoS One ; 16(6): e0252587, 2021.
Article in English | MEDLINE | ID: mdl-34061890

ABSTRACT

Living organisms are continuously under threat from a vast array of DNA-damaging agents, which impact genome DNA. DNA replication machinery stalls at damaged template DNA. The stalled replication fork is restarted via bypass replication by translesion DNA-synthesis polymerases, including the Y-family polymerases Polη, Polι, and Polκ, which possess the ability to incorporate nucleotides opposite the damaged template. To investigate the division of labor among these polymerases in vivo, we generated POLη-/-, POLι-/-, POLκ-/-, double knockout (KO), and triple knockout (TKO) mutants in all combinations from human TK6 cells. TKO cells exhibited a hypersensitivity to ultraviolet (UV), cisplatin (CDDP), and methyl methanesulfonate (MMS), confirming the pivotal role played by these polymerases in bypass replication of damaged template DNA. POLη-/- cells, but not POLι-/- or POLκ-/- cells, showed a strong sensitivity to UV and CDDP, while TKO cells showed a slightly higher sensitivity to UV and CDDP than did POLη-/- cells. On the other hand, TKO cells, but not all single KO cells, exhibited a significantly higher sensitivity to MMS than did wild-type cells. Consistently, DNA-fiber assay revealed that Polη plays a crucial role in bypassing lesions caused by UV-mimetic agent 4-nitroquinoline-1-oxide and CDDP, while all three polymerases play complementary roles in bypassing MMS-induced damage. Our findings indicate that the three Y-family polymerases play distinctly different roles in bypass replication, according to the type of DNA damage generated on the template strand.


Subject(s)
DNA Damage , DNA-Directed DNA Polymerase/metabolism , DNA/metabolism , Cell Line , Cisplatin/pharmacology , DNA/genetics , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair , DNA Replication , DNA-Directed DNA Polymerase/deficiency , DNA-Directed DNA Polymerase/genetics , Gene Knockout Techniques , Humans , Methyl Methanesulfonate/pharmacology , Ultraviolet Rays , DNA Polymerase iota
17.
Mol Biol Rep ; 48(5): 4107-4119, 2021 May.
Article in English | MEDLINE | ID: mdl-34075539

ABSTRACT

DNA damage response (DDR) pathways are initiated to prevent mutations from being passed on in the event of DNA damage. Mutations in DDR proteins can contribute to the development and maintenance of cancer cells, but many mutations observed in human tumors have not been functionally characterized. Because a proper response to DNA damage is fundamental to living organisms, DDR proteins and processes are often highly conserved. The goal of this project was to use Saccharomyces cerevisiae as a model for functional screening of human cancer mutations in conserved DDR proteins. After comparing the cancer mutation frequency and conservation of DDR proteins, Mre11 was selected for functional screening. A subset of mutations in conserved residues was analyzed by structural modeling and screened for functional effects in yeast Mre11. Yeast expressing wild type or mutant Mre11 were then assessed for DNA damage sensitivity using hydroxyurea (HU) and methyl methanesulfonate (MMS). The results were further validated in human cancer cells. The N-terminal point mutations tested in yeast Mre11 do not confer sensitivity to DNA damage sensitivity, suggesting that these residues are dispensable for yeast Mre11 function and may have conserved sequence without conserved function. However, a mutation near the capping domain associated with breast and colorectal cancers compromises Mre11 function in both yeast and human cells. These results provide novel insight into the function of this conserved capping domain residue and demonstrate a framework for yeast-based screening of cancer mutations.


Subject(s)
Adenocarcinoma/genetics , Breast Neoplasms/genetics , DNA Damage/genetics , DNA Repair/genetics , Early Detection of Cancer/methods , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/genetics , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/genetics , MRE11 Homologue Protein/chemistry , MRE11 Homologue Protein/genetics , Mutation Rate , Protein Domains/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Adenocarcinoma/pathology , Breast Neoplasms/pathology , DNA Damage/drug effects , Female , Humans , Hydroxyurea/pharmacology , MCF-7 Cells , Methyl Methanesulfonate/pharmacology , Microorganisms, Genetically-Modified , Saccharomyces cerevisiae/metabolism
18.
J Biosci ; 462021.
Article in English | MEDLINE | ID: mdl-33709962

ABSTRACT

The highly radiation-resistant bacterium Deinococcus radiodurans responds to gamma radiation or desiccation through the coordinated expression of genes belonging to Radiation and Desiccation Resistance/Response (RDR) regulon. RDR regulon is operated through cis-acting sequence RDRM (Radiation Desiccation Response Motif), trans-acting repressor DdrO and protease IrrE (also called PprI). The present study evaluated whether RDR regulon controls the response of D. radiodurans to various other DNA damaging stressors, to which it is resistant, such as UV rays, mitomycin C (MMC), methyl methanesulfonate (MMS), ethidium bromide (EtBr), etc. Activation of 3 RDR regulon genes (ddrB, gyrB and DR1143) was studied by tagging their promoter sequences with a highly sensitive GFP reporter. Here we demonstrated that all the DNA damaging stressors elicited activation of RDR regulon of D. radiodurans in a dose-dependent and RDRM-/IrrE-dependent manner. However, ROS-mediated indirect effects [induced by hydrogen peroxide (H2O2), methyl viologen (MV), heavy metal/metalloid (zinc or tellurite), etc.] did not activate RDR regulon. We also showed that level of activation was inversely proportional to cellular abundance of repressor DdrO. Our data strongly suggests that direct DNA damage activates RDR regulon in D. radiodurans.


Subject(s)
Bacterial Proteins/genetics , DNA Damage/radiation effects , Deinococcus/genetics , Radiation Tolerance/genetics , DNA Damage/drug effects , DNA Damage/genetics , Deinococcus/radiation effects , Gamma Rays/adverse effects , Gene Expression Regulation, Bacterial/radiation effects , Hydrogen Peroxide/pharmacology , Methyl Methanesulfonate/pharmacology , Nucleotide Motifs/radiation effects , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/radiation effects , Radiation Tolerance/drug effects , Ultraviolet Rays/adverse effects
19.
PLoS One ; 16(3): e0235303, 2021.
Article in English | MEDLINE | ID: mdl-33730086

ABSTRACT

Continuous culture systems allow for the controlled growth of microorganisms over a long period of time. Here, we develop a novel test for mutagenicity that involves growing yeast in continuous culture systems exposed to low levels of mutagen for a period of approximately 20 days. In contrast, most microorganism-based tests for mutagenicity expose the potential mutagen to the biological reporter at a high concentration of mutagen for a short period of time. Our test improves upon the sensitivity of the well-established Ames test by at least 20-fold for each of two mutagens that act by different mechanisms (the intercalator ethidium bromide and alkylating agent methyl methanesulfonate). To conduct the tests, cultures were grown in small, inexpensive continuous culture systems in media containing (potential) mutagen, and the resulting mutagenicity of the added compound was assessed via two methods: a canavanine-based plate assay and whole genome sequencing. In the canavanine-based plate assay, we were able to detect a clear relationship between the amount of mutagen and the number of canavanine-resistant mutant colonies over a period of one to three weeks of exposure. Whole genome sequencing of yeast grown in continuous culture systems exposed to methyl methanesulfonate demonstrated that quantification of mutations is possible by identifying the number of unique variants across each strain. However, this method had lower sensitivity than the plate-based assay and failed to distinguish the different concentrations of mutagen. In conclusion, we propose that yeast grown in continuous culture systems can provide an improved and more sensitive test for mutagenicity.


Subject(s)
Ethidium/pharmacology , Methyl Methanesulfonate/pharmacology , Saccharomyces cerevisiae/drug effects , Canavanine/pharmacology , Culture Media/chemistry , DNA, Fungal/chemistry , DNA, Fungal/metabolism , Mutagenicity Tests/instrumentation , Mutagenicity Tests/methods , Saccharomyces cerevisiae/genetics , Whole Genome Sequencing
20.
FEMS Yeast Res ; 21(2)2021 03 18.
Article in English | MEDLINE | ID: mdl-33606011

ABSTRACT

SLX4 is a scaffold to coordinate the action of structure-specific endonucleases that are required for homologous recombination and DNA repair. In view of ScSLX4 functions in the maintenance and stability of the genome in Saccharomyces cerevisiae, we have explored the roles of CaSLX4 in Candida albicans. Here, we constructed slx4Δ/Δ mutant and found that it exhibited increased sensitivity to the DNA damaging agent, methyl methanesulfonate (MMS) but not the DNA replication inhibitor, hydroxyurea (HU). Accordingly, RT-qPCR and western blotting analysis revealed the activation of SLX4 expression in response to MMS. The deletion of SLX4 resulted in a defect in the recovery from MMS-induced filamentation to yeast form and re-entry into the cell cycle. Like many other DNA repair genes, SLX4 expression was activated by the checkpoint kinase Rad53 under MMS-induced DNA damage. In addition, SLX4 was not required for the inactivation of the DNA damage checkpoint, as indicated by normal phosphorylation of Rad53 in slx4Δ/Δ cells. Therefore, our results demonstrate SLX4 plays an important role in cell recovery from MMS-induced DNA damage in C. albicans.


Subject(s)
Candida albicans/drug effects , Candida albicans/genetics , DNA Damage/drug effects , Endodeoxyribonucleases/genetics , Fungal Proteins/genetics , Methyl Methanesulfonate/pharmacology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Cell Cycle Checkpoints/drug effects , Endodeoxyribonucleases/metabolism , Fungal Proteins/metabolism , Hydroxyurea/pharmacology , Phosphorylation , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL