Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(39): e2314904120, 2023 Sep 26.
Article in English | MEDLINE | ID: mdl-37722041
2.
Antioxidants (Basel) ; 9(8)2020 Jul 23.
Article in English | MEDLINE | ID: mdl-32717933

ABSTRACT

Fibrocellular membrane or epiretinal membrane (ERM) forms on the surface of the inner limiting membrane (ILM) in the inner retina and alters the structure and function of the retina. ERM formation is frequently observed in ocular inflammatory conditions, such as proliferative diabetic retinopathy (PDR) and retinal detachment (RD). Although peeling of the ERM is used as a surgical intervention, it can inadvertently distort the retina. Our goal is to design alternative strategies to tackle ERMs. As a first step, we sought to determine the composition of the ERMs by identifying the constituent cell-types and gene expression signature in patient samples. Using ultrastructural microscopy and immunofluorescence analyses, we found activated microglia, astrocytes, and Müller glia in the ERMs from PDR and RD patients. Moreover, oxidative stress and inflammation associated gene expression was significantly higher in the RD and PDR membranes as compared to the macular hole samples, which are not associated with inflammation. We specifically detected differential expression of hypoxia inducible factor 1-α (HIF1-α), proinflammatory cytokines, and Notch, Wnt, and ERK signaling pathway-associated genes in the RD and PDR samples. Taken together, our results provide new information to potentially develop methods to tackle ERM formation.

3.
Front Neurosci ; 14: 611904, 2020.
Article in English | MEDLINE | ID: mdl-33384581

ABSTRACT

Parkinson's disease, diabetic retinopathy, hyperoxia induced retinopathy, and neuronal damage resulting from ischemia are among the notable neurodegenerative diseases in which oxidative stress occurs shortly before the onset of neurodegeneration. A shared feature of these diseases is the depletion of OXR1 (oxidation resistance 1) gene products shortly before the onset of neurodegeneration. In animal models of these diseases, restoration of OXR1 has been shown to reduce or eliminate the deleterious effects of oxidative stress induced cell death, delay the onset of symptoms, and reduce overall severity. Moreover, increasing OXR1 expression in cells further increases oxidative stress resistance and delays onset of disease while showing no detectable side effects. Thus, restoring or increasing OXR1 function shows promise as a therapeutic for multiple neurodegenerative diseases. This review examines the role of OXR1 in oxidative stress resistance and its impact on neurodegenerative diseases. We describe the potential of OXR1 as a therapeutic in light of our current understanding of its function at the cellular and molecular level and propose a possible cascade of molecular events linked to OXR1's regulatory functions.

4.
BMC Mol Biol ; 17(1): 20, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27578267

ABSTRACT

BACKGROUND: Cells respond to numerous internal and external stresses, such as heat, cold, oxidative stress, DNA damage, and osmotic pressure changes. In most cases, the primary response to stress is transcriptional induction of genes that assist the cells in tolerating the stress and facilitate the repair of the cellular damage. However, when the transcription machinery itself is stressed, responding by such standard mechanisms may not be possible. RESULTS: In this study, we demonstrate that depletion or inactivation of RNA polymerase II (RNAPII) changes the preferred polyadenylation site usage for several transcripts, and leads to increased transcription of a specific subset of genes. Surprisingly, depletion of RNA polymerase I (RNAPI) also promotes altered polyadenylation site usage, while depletion of RNA polymerase III (RNAPIII) does not appear to have an impact. CONCLUSIONS: Our results demonstrate that stressing the transcription machinery by depleting either RNAPI or RNAPII leads to a novel transcriptional response that results in induction of specific mRNAs and altered polyadenylation of many of the induced transcripts.


Subject(s)
Alternative Splicing , Polyadenylation , RNA Polymerase II/genetics , RNA, Messenger/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Transcription, Genetic , Gene Deletion
5.
Mol Cell Biochem ; 412(1-2): 165-71, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26708217

ABSTRACT

Reactive oxygen species are a by-product of aerobic metabolism that can damage lipid, proteins, and nucleic acids. Oxidative damage to DNA is especially critical, because it can lead to cell death or mutagenesis. Previously we reported that the yeast sub1 deletion mutant is sensitive to hydrogen peroxide treatment and that the human SUB1 can complement the sensitivity of the yeast sub1 mutant. In this study, we find that Sub1 protects DNA from oxidative damage in vivo and in vitro. We demonstrate that transcription of SUB1 mRNA is induced by oxidative stress and that the sub1Δ mutant has an increased number of chromosomal DNA strand breaks after peroxide treatment. We further demonstrate that purified Sub1 protein can protect DNA from oxidative damage in vitro, using the metal ion catalyzed oxidation assay.


Subject(s)
DNA Damage , DNA-Binding Proteins/physiology , Oxidative Stress , Transcription Factors/physiology , Humans
6.
J Interferon Cytokine Res ; 35(3): 186-99, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25330068

ABSTRACT

We demonstrate that interferon (IFN)-ß-1b induces an alternative-start transcript containing the C-terminal TLDc domain of nuclear receptor coactivator protein 7 (NCOA7), a member of the OXR family of oxidation resistance proteins. IFN-ß-1b induces NCOA7-AS (alternative start) expression in peripheral blood mononuclear cells (PBMCs) obtained from healthy individuals and multiple sclerosis patients and human fetal brain cells, astrocytoma, neuroblastoma, and fibrosarcoma cells. NCOA7-AS is a previously undocumented IFN-ß-inducible gene that contains only the last 5 exons of full-length NCOA7 plus a unique first exon (exon 10a) that is not found in longer forms of NCOA7. This exon encodes a domain closely related to an important class of bacterial aldo-keto oxido-reductase proteins that play a critical role in regulating redox activity. We demonstrate that NCOA7-AS is induced by IFN and LPS, but not by oxidative stress and exhibits, independently, oxidation resistance activity. We further demonstrate that induction of NCOA7-AS by IFN is dependent on IFN-receptor activation, the Janus kinase-signal transducers and activators of transcription (JAK-STAT) signaling pathway, and a canonical IFN-stimulated response element regulatory sequence upstream of exon 10a. We describe a new role for IFN-ßs involving a mechanism of action that leads to an increase in resistance to inflammation-mediated oxidative stress.


Subject(s)
Immunotherapy/methods , Interferon-beta/metabolism , Leukocytes, Mononuclear/physiology , Multiple Sclerosis/therapy , Nuclear Receptor Coactivators/metabolism , Protein Isoforms/metabolism , Amino Acid Sequence , Cell Line , Humans , Janus Kinases/metabolism , Molecular Sequence Data , Multiple Sclerosis/immunology , Nuclear Receptor Coactivators/genetics , Oxidation-Reduction , Oxidative Stress , Protein Isoforms/genetics , Protein Structure, Tertiary/genetics , Receptors, Interferon/metabolism , Regulatory Elements, Transcriptional/genetics , STAT Transcription Factors/metabolism , Signal Transduction
7.
PLoS One ; 8(3): e58015, 2013.
Article in English | MEDLINE | ID: mdl-23554872

ABSTRACT

Non homologous end joining (NHEJ) is an important process that repairs double strand DNA breaks (DSBs) in eukaryotic cells. Cells defective in NHEJ are unable to join chromosomal breaks. Two different NHEJ assays are typically used to determine the efficiency of NHEJ. One requires NHEJ of linearized plasmid DNA transformed into the test organism; the other requires NHEJ of a single chromosomal break induced either by HO endonuclease or the I-SceI restriction enzyme. These two assays are generally considered equivalent and rely on the same set of NHEJ genes. PC4 is an abundant DNA binding protein that has been suggested to stimulate NHEJ. Here we tested the role of PC4's yeast homolog SUB1 in repair of DNA double strand breaks using different assays. We found SUB1 is required for NHEJ repair of DSBs in plasmid DNA, but not in chromosomal DNA. Our results suggest that these two assays, while similar are not equivalent and that repair of plasmid DNA requires additional factor(s) that are not required for NHEJ repair of chromosomal double-strand DNA breaks. Possible roles for Sub1 proteins in NHEJ of plasmid DNA are discussed.


Subject(s)
Chromosome Breakage , Chromosomes, Fungal/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair/physiology , DNA, Fungal/metabolism , DNA-Binding Proteins/metabolism , Plasmids/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Chromosomes, Fungal/genetics , DNA, Fungal/genetics , DNA-Binding Proteins/genetics , Plasmids/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics
8.
Nucleic Acids Res ; 41(5): 3104-14, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23355614

ABSTRACT

Alternative polyadenylation (APA) is conserved in all eukaryotic cells. Selective use of polyadenylation sites appears to be a highly regulated process and contributes to human pathogenesis. In this article we report that the yeast RPB2 gene is alternatively polyadenylated, producing two mRNAs with different lengths of 3'UTR. In normally growing wild-type cells, polyadenylation preferentially uses the promoter-proximal poly(A) site. After UV damage transcription of RPB2 is initially inhibited. As transcription recovers, the promoter-distal poly(A) site is preferentially used instead, producing more of a longer form of RPB2 mRNA. We show that the relative increase in the long RPB2 mRNA is not caused by increased mRNA stability, supporting the preferential usage of the distal poly(A) site during transcription recovery. We demonstrate that the 3'UTR of RPB2 is sufficient for this UV-induced regulation of APA. We present evidence that while transcription initiation rates do not seem to influence selection of the poly(A) sites of RPB2, the rate of transcription elongation is an important determinant.


Subject(s)
DNA Damage , Polyadenylation/radiation effects , RNA Polymerase II/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , 3' Untranslated Regions , Base Sequence , RNA Polymerase II/metabolism , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/radiation effects , Saccharomyces cerevisiae Proteins/metabolism , Transcription Elongation, Genetic , Transcription Initiation, Genetic , Ultraviolet Rays
9.
BMC Mol Biol ; 13: 26, 2012 Aug 08.
Article in English | MEDLINE | ID: mdl-22873401

ABSTRACT

BACKGROUND: The human OXR1 gene belongs to a class of genes with conserved functions that protect cells from reactive oxygen species (ROS). The gene was found using a screen of a human cDNA library by its ability to suppress the spontaneous mutator phenotype of an E. coli mutH nth strain. The function of OXR1 is unknown. The human and yeast genes are induced by oxidative stress and targeted to the mitochondria; the yeast gene is required for resistance to hydrogen peroxide. Multiple spliced isoforms are expressed in a variety of human tissues, including brain. RESULTS: In this report, we use a papillation assay that measures spontaneous mutagenesis of an E. coli mutM mutY strain, a host defective for oxidative DNA repair. Papillation frequencies with this strain are dependent upon a G→T transversion in the lacZ gene (a mutation known to occur as a result of oxidative damage) and are suppressed by in vivo expression of human OXR1. N-terminal, C-terminal and internal deletions of the OXR1 gene were constructed and tested for suppression of the mutagenic phenotype of the mutM mutY strain. We find that the TLDc domain, encoded by the final four exons of the OXR1 gene, is not required for papillation suppression in E. coli. Instead, we show that the protein segment encoded by exon 8 of OXR1 is responsible for the suppression of oxidative damage in E. coli. CONCLUSION: The protein segment encoded by OXR1 exon 8 plays an important role in the anti-oxidative function of the human OXR1 protein. This result suggests that the TLDc domain, found in OXR1 exons 12-16 and common in many proteins with nuclear function, has an alternate (undefined) role other than oxidative repair.


Subject(s)
Exons/genetics , Gene Expression Regulation/genetics , Mitochondria/metabolism , Oxidative Stress/genetics , Proteins/genetics , Proteins/metabolism , DNA Primers/genetics , Escherichia coli , Gene Library , Humans , Mitochondrial Proteins , Mutagenesis , Plasmids/genetics
10.
DNA Repair (Amst) ; 10(9): 934-41, 2011 Sep 05.
Article in English | MEDLINE | ID: mdl-21788159

ABSTRACT

aidB is one of the four genes of E. coli that is induced by alkylating agents and regulated by Ada protein. Three genes (ada, alkA, and alkB) encode DNA repair proteins that remove or repair alkylated bases. However, the role of AidB remains unclear despite extensive efforts to determine its function in cells exposed to alkylating agents. The E. coli AidB protein was identified as a component of the protein complex that assembles at strong promoters. We demonstrate that AidB protein preferentially binds to UP elements, AT rich transcription enhancer sequences found upstream of many highly expressed genes, several DNA repair genes, and housekeeping genes. AidB allows efficient transcription from promoters containing an UP element upon exposure to a DNA methylating agent and protects downstream genes from DNA damage. The DNA binding domain is required to target AidB to specific genes preferentially protecting them from alkylation damage. However, deletion of AidB's DNA binding domain does not prevent its antimutagenic activity, instead this deletion appears to allow AidB to function as a cytoplasmic alkylation resistance protein. Our studies identify the role of AidB in alkylating agent exposed cells and suggest a new cellular strategy in which a subset of the genome is preferentially protected from damage by alkylating agents.


Subject(s)
DNA Damage/genetics , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Alkylating Agents/pharmacology , Alkylation/drug effects , DNA Damage/drug effects , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Escherichia coli/drug effects , Escherichia coli Proteins/metabolism , Transcription, Genetic
11.
Methods Mol Biol ; 477: 331-42, 2008.
Article in English | MEDLINE | ID: mdl-19082958

ABSTRACT

In order to develop a more complete understanding of the genes required for resistance to oxidative DNA damage, we devised methods to identify genes that can prevent or repair oxidative DNA damage. These methods use the oxidative mutator phenotype of a repair deficient E. coli strain to measure the antimutator effect resulting from the expression of human cDNAs. The method can be adapted to characterize the function, and to determine the active site domains, of putative antimutator genes. Since bacteria do not contain subcellular compartments, genes that function in mitochondria, the cytoplasm, or the nucleus can be identified. Methods to determine the localization of genes in their normal host organism are also described.


Subject(s)
Genes , Genomics/methods , Oxidation-Reduction , Biological Assay , DNA, Complementary/genetics , Fluorescent Antibody Technique , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins , Mutagenesis/drug effects , Mutation/genetics , Organic Chemicals/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Proteins/metabolism , Reactive Oxygen Species/metabolism , Staining and Labeling
12.
BMC Cell Biol ; 8: 13, 2007 Mar 28.
Article in English | MEDLINE | ID: mdl-17391516

ABSTRACT

BACKGROUND: The NCOA7 gene product is an estrogen receptor associated protein that is highly similar to the human OXR1 gene product, which functions in oxidation resistance. OXR genes are conserved among all sequenced eukaryotes from yeast to humans. In this study we examine if NCOA7 has an oxidation resistance function similar to that demonstrated for OXR1. We also examine NCOA7 expression in response to oxidative stress and its subcellular localization in human cells, comparing these properties with those of OXR1. RESULTS: We find that NCOA7, like OXR1 can suppress the oxidative mutator phenotype when expressed in an E. coli strain that exhibits an oxidation specific mutator phenotype. Moreover, NCOA7's oxidation resistance function requires expression of only its carboxyl-terminal domain and is similar in this regard to OXR1. We find that, in human cells, NCOA7 is constitutively expressed and is not induced by oxidative stress and appears to localize to the nucleus following estradiol stimulation. These properties of NCOA7 are in striking contrast to those of OXR1, which is induced by oxidative stress, localizes to mitochondria, and appears to be excluded, or largely absent from nuclei. CONCLUSION: NCOA7 most likely arose from duplication. Like its homologue, OXR1, it is capable of reducing the DNA damaging effects of reactive oxygen species when expressed in bacteria, indicating the protein has an activity that can contribute to oxidation resistance. Unlike OXR1, it appears to localize to nuclei and interacts with the estrogen receptor. This raises the possibility that NCOA7 encodes the nuclear counterpart of the mitochondrial OXR1 protein and in mammalian cells it may reduce the oxidative by-products of estrogen metabolite-mediated DNA damage.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Oxidative Stress , Proteins/chemistry , Cell Nucleus/metabolism , Conserved Sequence , DNA Damage , Escherichia coli , Eukaryotic Cells , Gene Expression Regulation/drug effects , Humans , Hydrogen Peroxide/pharmacology , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Proteins , Multigene Family , Nuclear Receptor Coactivators , Protein Structure, Tertiary/physiology , Proteins/genetics , Proteins/physiology , Tumor Cells, Cultured
13.
DNA Repair (Amst) ; 6(6): 733-41, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17292678

ABSTRACT

DNA repair generally functions to improve survival and reduce mutagenesis of cells that have suffered DNA damage. In this study we examine the role of nucleotide excision repair (NER) and base excision repair (BER) in recovery, mutagenesis and DNA repair in response to DNA damage inflicted by the mustard compounds, sulfur mustard (SM) and chloroethyl ethyl sulfide (CEES) in bacteria and mammalian cells. SM and CEES are compared because SM produces cross-links and monoadducts, whereas CEES produces only monoadducts that are similar to those produced by SM, thus allowing the examination of which types of lesions may be responsible for the effects seen. We find that the presence of a functional NER pathway increases survival and reduces mutagenesis, whereas the presence of a functional BER pathway reduces survival, increases mutagenesis, and decreases repair. The deleterious effects of BER appear to be due to an interaction between the DNA glycosylases and the lesions produced by SM and CEES. Possible mechanisms for BER-mediated sensitization by glycosylase action on mustard lesions are discussed.


Subject(s)
DNA Damage , DNA Repair , Mustard Gas/analogs & derivatives , Mustard Gas/toxicity , Animals , Cell Survival , DNA Glycosylases/metabolism , Escherichia coli/metabolism , Luciferases/metabolism , Models, Chemical , Models, Molecular , Molecular Conformation , Mutagenesis
14.
J Virol ; 80(12): 5862-74, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16731925

ABSTRACT

Infected cells recognize viral replication as a DNA damage stress and elicit a DNA damage response that ultimately induces apoptosis as part of host immune surveillance. Here, we demonstrate a novel mechanism where the murine gamma herpesvirus 68 (gammaHV68) latency-associated, anti-interferon M2 protein inhibits DNA damage-induced apoptosis by interacting with the DDB1/COP9/cullin repair complex and the ATM DNA damage signal transducer. M2 expression constitutively induced DDB1 nuclear localization and ATM kinase activation in the absence of DNA damage. Activated ATM subsequently induced Chk activation and p53 phosphorylation and stabilization without eliciting H2AX phosphorylation and MRN recruitment to foci upon DNA damage. Consequently, M2 expression inhibited DNA repair, rendered cells resistant to DNA damage-induced apoptosis, and induced a G(1) cell cycle arrest. Our results suggest that gammaHV68 M2 blocks apoptosis-mediated intracellular innate immunity, which might ultimately contribute to its role in latent infection.


Subject(s)
DNA Damage , Rhadinovirus/physiology , Signal Transduction , Viral Matrix Proteins/physiology , Virus Latency , Active Transport, Cell Nucleus , Animals , Apoptosis , Cell Cycle , Cell Line , DNA Repair , DNA-Binding Proteins/metabolism , G1 Phase , Herpesviridae Infections , Humans , Mice , Tumor Virus Infections
15.
Mol Cell Biol ; 24(13): 6084-93, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15199162

ABSTRACT

Human positive cofactor 4 (PC4) is a transcriptional coactivator with a highly conserved single-strand DNA (ssDNA) binding domain of unknown function. We identified PC4 as a suppressor of the oxidative mutator phenotype of the Escherichia coli fpg mutY mutant and demonstrate that this suppression requires its ssDNA binding activity. Saccharomyces cerevisiae mutants lacking their PC4 ortholog Sub1 are sensitive to hydrogen peroxide and exhibit spontaneous and peroxide-induced hypermutability. PC4 expression suppresses the peroxide sensitivity of the yeast sub1Delta mutant, suggesting that the human protein has a similar function. A role for yeast and human proteins in DNA repair is suggested by the demonstration that Sub1 acts in a peroxide resistance pathway involving Rad2 and by the physical interaction of PC4 with the human Rad2 homolog XPG. We show that XPG recruits PC4 to a bubble-containing DNA substrate with a resulting displacement of XPG and formation of a PC4-DNA complex. We discuss the possible requirement for PC4 in either global or transcription-coupled repair of oxidative DNA damage to mediate the release of XPG bound to its substrate.


Subject(s)
DNA Damage , Mutagenesis , Repressor Proteins/physiology , Trans-Activators/physiology , Cell Death , DNA/metabolism , DNA Repair , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Endodeoxyribonucleases/metabolism , Endonucleases , Humans , Immediate-Early Proteins , Membrane Proteins , Nuclear Proteins , Oxidative Stress , Protein Binding , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors
16.
Mol Cell Biol ; 24(8): 3180-7, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15060142

ABSTRACT

Reactive oxygen species (ROS) are critical molecules produced as a consequence of aerobic respiration. It is essential for cells to control the production and activity of such molecules in order to protect the genome and regulate cellular processes such as stress response and apoptosis. Mitochondria are the major source of ROS within the cell, and as a result, numerous proteins have evolved to prevent or repair oxidative damage in this organelle. The recently discovered OXR1 gene family represents a set of conserved eukaryotic genes. Previous studies of the yeast OXR1 gene indicate that it functions to protect cells from oxidative damage. In this report, we show that human and yeast OXR1 genes are induced by heat and oxidative stress and that their proteins localize to the mitochondria and function to protect against oxidative damage. We also demonstrate that mitochondrial localization is required for Oxr1 protein to prevent oxidative damage.


Subject(s)
Gene Expression Regulation , Mitochondria/metabolism , Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Genetic Complementation Test , HeLa Cells , Hot Temperature , Humans , Hydrogen Peroxide/metabolism , Mitochondrial Proteins , Oxidants/metabolism , Oxidation-Reduction , Oxidative Stress , Proteins/genetics , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Tissue Distribution
17.
Carcinogenesis ; 24(3): 589-93, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12663522

ABSTRACT

To investigate the possible role of glycosylase action in causing tumor resistance, a full-length, histidine-tagged human alkyladenine glycosylase has been purified from the cloned human gene contained in a pTrc99A vector propagated in a tag alkA mutant Escherichia coli. This human enzyme releases both 3-methyladenine and 7-methylguanine from methylated DNA but in contrast to previous studies of the bacterial AlkA glycosylase, it does not release any adducts from [(3)H]chloroethylnitrosourea-modified DNA. This finding suggests that the alkyladenine DNA glycosylase-dependent resistance to the toxic effects of the chloroethylnitrosoureas reported previously in the literature may occur by a mechanism other than through direct glycosylase action.


Subject(s)
DNA Glycosylases , N-Glycosyl Hydrolases/metabolism , Nitrosourea Compounds/pharmacology , Base Sequence , DNA Primers , Humans , Substrate Specificity
18.
J Bacteriol ; 185(5): 1701-4, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12591888

ABSTRACT

A major role of the methyl-directed mismatch repair (MMR) system of Escherichia coli is to repair postreplicative errors. In this report, we provide evidence that MMR also acts on oxidized DNA, preventing mutagenesis. When cells deficient in MMR are grown anaerobically, spontaneous mutation frequencies are reduced compared with those of the same cells grown aerobically. In addition, we show that a dam mutant has an increased sensitivity to hydrogen peroxide treatment that can be suppressed by mutations that inactivate MMR. In a dam mutant, MMR is not targeted to newly replicated DNA strands and therefore mismatches are converted to single- and double-strand DNA breaks. Thus, base pairs containing oxidized bases will be converted to strand breaks if they are repaired by MMR. This is demonstrated by the increased peroxide sensitivity of a dam mutant and the finding that the sensitivity can be suppressed by mutations inactivating MMR. We demonstrate further that this repair activity results from MMR recognition of base pairs containing 8-oxoguanine (8-oxoG) based on the finding that overexpression of the MutM oxidative repair protein, which repairs 8-oxoG, can suppress the mutH-dependent increase in transversion mutations. These findings demonstrate that MMR has the ability to prevent oxidative mutagenesis either by removing 8-oxoG directly or by removing adenine misincorporated opposite 8-oxoG or both.


Subject(s)
Base Pair Mismatch , DNA Repair Enzymes , DNA Repair , Escherichia coli Proteins , Escherichia coli/genetics , Guanine/analogs & derivatives , Anaerobiosis , Cell Division , DNA/metabolism , DNA Damage/drug effects , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Formamidopyrimidine Glycosylase , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Escherichia coli/drug effects , Escherichia coli/metabolism , Gene Expression Regulation , Guanine/metabolism , Hydrogen Peroxide/pharmacology , Mutation , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/metabolism , Oxidation-Reduction , Oxidative Stress
19.
DNA Repair (Amst) ; 1(7): 507-16, 2002 Jul 17.
Article in English | MEDLINE | ID: mdl-12509225

ABSTRACT

The alkyladenine DNA glycosylase (AAG) has been cloned from mouse and humans. AAG knock out mouse cells are sensitized to a variety of alkylating and cross-linking agents suggesting AAG is active on a variety of substrates. In humans, two isoforms have been characterized that are generated by alternative splicing and contain either exon 1a or 1b (hAAG1 or hAAG2). In this study, we examine the ability of the both known isoforms of human AAG (hAAG) to contribute to survival of Escherichia coli from treatments with simple alkylating agents and cross-linking alkylating agents. Our results show that hAAG is effective at repairing methyl lesions when expressed in E. coli, but is unable to afford increased resistance to alkylating agents producing larger alkyl lesions such as ethyl lesions or lesions produced by the cross-linking alkylating agents N,N'-bis-chloroethyl-N-nitrosourea (BCNU), N-(2-chloroethyl)-N-nitrosourea (CNU) or mitomycin C. In the case of CNU, expression of hAAG causes increased sensitivity rather than resistance, suggesting deleterious effects of hAAG activity. We also demonstrate that there are no apparent differences between the two isoforms of hAAG when recovery from damage produced by all alkylating agents is tested.


Subject(s)
DNA Glycosylases , Escherichia coli/enzymology , N-Glycosyl Hydrolases/genetics , N-Glycosyl Hydrolases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Alkylating Agents/metabolism , Alkylation , DNA Repair/genetics , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Methylation/drug effects , Substrate Specificity/genetics , Substrate Specificity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...