141 results on '"Maizels N"'
Search Results
2. A Novel Target for Human Exonucleasel (hExol) in Class Switch Recombination.: 1
- Author
-
Vallur, A and Maizels, N.
- Published
- 2007
3. Preimmune diversification creates a repertoire while somatic hypermutation fine-tunes affinity —implications for the processes of mutation
- Author
-
Maizels, N.
- Published
- 1993
- Full Text
- View/download PDF
4. Isolation and detection of DNA-protein crosslinks in mammalian cells.
- Author
-
Torrecilla I, Ruggiano A, Kiianitsa K, Aljarbou F, Lascaux P, Hoslett G, Song W, Maizels N, and Ramadan K
- Subjects
- Animals, DNA genetics, DNA metabolism, DNA Replication, DNA Repair, Mammals genetics, DNA Damage, Proteins genetics
- Abstract
DNA-protein crosslinks (DPCs) are toxic DNA lesions wherein a protein is covalently attached to DNA. If not rapidly repaired, DPCs create obstacles that disturb DNA replication, transcription and DNA damage repair, ultimately leading to genome instability. The persistence of DPCs is associated with premature ageing, cancer and neurodegeneration. In mammalian cells, the repair of DPCs mainly relies on the proteolytic activities of SPRTN and the 26S proteasome, complemented by other enzymes including TDP1/2 and the MRN complex, and many of the activities involved are essential, restricting genetic approaches. For many years, the study of DPC repair in mammalian cells was hindered by the lack of standardised assays, most notably assays that reliably quantified the proteins or proteolytic fragments covalently bound to DNA. Recent interest in the field has spurred the development of several biochemical methods for DPC analysis. Here, we critically analyse the latest techniques for DPC isolation and the benefits and drawbacks of each. We aim to assist researchers in selecting the most suitable isolation method for their experimental requirements and questions, and to facilitate the comparison of results across different laboratories using different approaches., (© The Author(s) 2023. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
5. Topoisomerase Assays.
- Author
-
Nitiss JL, Kiianitsa K, Sun Y, Nitiss KC, and Maizels N
- Subjects
- Animals, DNA Cleavage, DNA Topoisomerases, Mice, Plasmids, DNA Topoisomerases, Type II metabolism, DNA, Superhelical
- Abstract
Topoisomerases are enzymes that play essential roles in DNA replication, transcription, chromosome segregation, and recombination. All cells have two major forms of DNA topoisomerases: type I enzymes, which make single-stranded cuts in DNA, and type II enzymes, which cut and decatenate double-stranded DNA. DNA topoisomerases are important targets of approved and experimental anti-cancer agents. Provided in this article are protocols to assess activities of topoisomerases and their inhibitors. Included are an assay for topoisomerase I activity based on relaxation of supercoiled DNA; an assay for topoisomerase II based on the decatenation of double-stranded DNA; and approaches for enriching and quantifying DNA-protein covalent complexes formed as obligatory intermediates in the reactions of type I and II topoisomerases with DNA; and assays for measuring DNA cleavage in vitro. Topoisomerases are not the only proteins that form covalent adducts with DNA in living cells, and the approaches described here are likely to find use in characterizing other protein-DNA adducts and exploring their utility as targets for therapy. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Assay of topoisomerase I activity Basic Protocol 2: Assay of topoisomerase II activity Basic Protocol 3: In vivo determination of topoisomerase covalent complexes using the in vivo complex of enzyme (ICE) assay Support Protocol 1: Preparation of mouse tissue for determination of topoisomerase covalent complexes using the ICE assay Support Protocol 2: Using recombinant topoisomerase standard for absolute quantification of cellular TOP2CC Basic Protocol 4: Quantification of topoisomerase-DNA covalent complexes by RADAR/ELISA: The rapid approach to DNA adduct recovery (RADAR) combined with the enzyme-linked immunosorbent assay (ELISA) Basic Protocol 5: Analysis of protein-DNA covalent complexes by RADAR/Western Support Protocol 3: Adduct-Seq to characterize adducted DNA Support Protocol 4: Nuclear fractionation and RNase treatment to reduce sample complexity Basic Protocol 6: Determination of DNA cleavage by purified topoisomerase I Basic Protocol 7: Determination of inhibitor effects on DNA cleavage by topoisomerase II using a plasmid linearization assay Alternate Protocol: Gel electrophoresis determination of topoisomerase II cleavage., (© 2021 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
6. Pathways and signatures of mutagenesis at targeted DNA nicks.
- Author
-
Zhang Y, Davis L, and Maizels N
- Subjects
- Cell Cycle genetics, DNA Breaks, Double-Stranded, DNA Helicases genetics, DNA Repair genetics, DNA-Binding Proteins genetics, DNA-Directed DNA Polymerase genetics, High-Throughput Nucleotide Sequencing, Humans, INDEL Mutation genetics, Mad2 Proteins genetics, Nucleotidyltransferases genetics, Signal Transduction genetics, DNA Polymerase theta, RNA, Guide, CRISPR-Cas Systems, BRCA2 Protein genetics, DNA Breaks, Single-Stranded, DNA Damage genetics, Mutagenesis genetics
- Abstract
Nicks are the most frequent form of DNA damage and a potential source of mutagenesis in human cells. By deep sequencing, we have identified factors and pathways that promote and limit mutagenic repair at a targeted nick in human cells. Mutations were distributed asymmetrically around the nick site. BRCA2 inhibited all categories of mutational events, including indels, SNVs and HDR. DNA2 and RPA promoted resection. DNA2 inhibited 1 bp deletions but contributed to longer deletions, as did REV7. POLQ stimulated SNVs. Parallel analysis of DSBs targeted to the same site identified similar roles for DNA2 and POLQ (but not REV7) in promoting deletions and for POLQ in stimulating SNVs. Insertions were infrequent at nicks, and most were 1 bp in length, as at DSBs. The translesion polymerase REV1 stimulated +1 insertions at one nick site but not another, illustrating the potential importance of sequence context in determining the outcome of mutagenic repair. These results highlight the potential for nicks to promote mutagenesis, especially in BRCA-deficient cells, and identify mutagenic signatures of DNA2, REV1, REV3, REV7 and POLQ., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
7. Treatment of human cells with 5-aza-dC induces formation of PARP1-DNA covalent adducts at genomic regions targeted by DNMT1.
- Author
-
Kiianitsa K, Zhang Y, and Maizels N
- Subjects
- Antimetabolites, Antineoplastic pharmacology, Antimetabolites, Antineoplastic therapeutic use, Azacitidine toxicity, Cells, Cultured, DNA drug effects, DNA metabolism, DNA Adducts chemistry, DNA Methylation, Hematologic Neoplasms genetics, Hematologic Neoplasms metabolism, Humans, K562 Cells, Azacitidine pharmacology, CpG Islands, DNA (Cytosine-5-)-Methyltransferase 1 metabolism, DNA Adducts metabolism, Hematologic Neoplasms drug therapy, Poly (ADP-Ribose) Polymerase-1 chemistry
- Abstract
The nucleoside analog 5-aza-2'-deoxycytidine (5-aza-dC) is used to treat some hematopoietic malignancies. The mechanism of cell killing depends upon DNMT1, but is otherwise not clearly defined. Here we show that PARP1 forms covalent DNA adducts in human lymphoblast or fibroblasts treated with 5-aza-dC. Some adducts recovered from 5-aza-dC-treated cells have undergone cleavage by apoptotic caspases 3/7. Mapping of PARP1-DNA adducts, by a new method, "Adduct-Seq", demonstrates adduct enrichment at CpG-dense genomic locations that are targets of maintenance methylation by DNMT1. Covalent protein-DNA adducts can arrest replication and induce apoptosis, and these results raise the possibility that induction of PARP1-DNA adducts may contribute to cell killing in response to treatment with 5-aza-dC., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
8. Rapid, direct detection of bacterial topoisomerase 1-DNA adducts by RADAR/ELISA.
- Author
-
Sinha D, Kiianitsa K, Sherman DR, and Maizels N
- Subjects
- Bacterial Proteins biosynthesis, Bacterial Proteins isolation & purification, DNA Adducts isolation & purification, DNA Topoisomerases, Type I isolation & purification, Escherichia coli chemistry, Escherichia coli genetics, Escherichia coli metabolism, Immunoblotting methods, Mycobacterium smegmatis chemistry, Mycobacterium smegmatis genetics, Mycobacterium smegmatis metabolism, Reproducibility of Results, Yersinia pestis genetics, Bacterial Proteins analysis, Cell Fractionation methods, DNA Adducts analysis, DNA Topoisomerases, Type I analysis, Enzyme-Linked Immunosorbent Assay methods, High-Throughput Screening Assays methods
- Abstract
Topoisomerases are proven drug targets, but antibiotics that poison bacterial Topoisomerase 1 (Top1) have yet to be discovered. We have developed a rapid and direct assay for quantification of Top1-DNA adducts that is suitable for high throughput assays. Adducts are recovered by "RADAR fractionation", a quick, convenient approach in which cells are lysed in chaotropic salts and detergent and nucleic acids and covalently bound adducts then precipitated with alcohol. Here we show that RADAR fractionation followed by ELISA immunodetection can quantify adducts formed by wild-type and mutant Top1 derivatives encoded by two different bacterial pathogens, Y. pestis and M. tuberculosis, expressed in E. coli or M. smegmatis, respectively. For both enzymes, quantification of adducts by RADAR/ELISA produces results comparable to the more cumbersome classical approach of CsCl density gradient fractionation. The experiments reported here establish that RADAR/ELISA assay offers a simple way to characterize Top1 mutants and analyze kinetics of adduct formation and repair. They also provide a foundation for discovery and optimization of drugs that poison bacterial Top1 using standard high-throughput approaches., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
9. POLQ suppresses interhomolog recombination and loss of heterozygosity at targeted DNA breaks.
- Author
-
Davis L, Khoo KJ, Zhang Y, and Maizels N
- Subjects
- CRISPR-Cas Systems, Cell Line, Tumor, DNA Breaks, Single-Stranded, DNA End-Joining Repair, DNA Ligases genetics, DNA Ligases metabolism, DNA-Directed DNA Polymerase genetics, Heterozygote, Humans, Loss of Heterozygosity, Mutation, Recombination, Genetic, DNA Polymerase theta, DNA Breaks, Double-Stranded, DNA-Directed DNA Polymerase metabolism, Recombinational DNA Repair
- Abstract
Interhomolog recombination (IHR) occurs spontaneously in somatic human cells at frequencies that are low but sufficient to ameliorate some genetic diseases caused by heterozygous mutations or autosomal dominant mutations. Here we demonstrate that DNA nicks or double-strand breaks (DSBs) targeted by CRISPR-Cas9 to both homologs can stimulate IHR and associated copy-neutral loss of heterozygosity (cnLOH) in human cells. The frequency of IHR is 10-fold lower at nicks than at DSBs, but cnLOH is evident in a greater fraction of recombinants. IHR at DSBs occurs predominantly via reciprocal end joining. At DSBs, depletion of POLQ caused a dramatic increase in IHR and in the fraction of recombinants exhibiting cnLOH, suggesting that POLQ promotes end joining in cis , which limits breaks available for recombination in trans These results define conditions that may produce cnLOH as a mutagenic signature in cancer and may, conversely, promote therapeutic correction of both compound heterozygous and dominant negative mutations associated with genetic disease., Competing Interests: The authors declare no competing interest.
- Published
- 2020
- Full Text
- View/download PDF
10. The "adductome": A limited repertoire of adducted proteins in human cells.
- Author
-
Kiianitsa K and Maizels N
- Subjects
- Cell Line, Cross-Linking Reagents chemistry, Cross-Linking Reagents pharmacology, DNA Topoisomerases, Type I chemistry, DNA-Binding Proteins analysis, Etoposide chemistry, Etoposide pharmacology, Formaldehyde chemistry, Formaldehyde pharmacology, High Mobility Group Proteins chemistry, Histones chemistry, Humans, Proteomics, RNA-Binding Proteins analysis, Topotecan chemistry, Topotecan pharmacology, DNA chemistry, DNA Adducts analysis, DNA-Binding Proteins chemistry, Mass Spectrometry, RNA-Binding Proteins chemistry
- Abstract
Proteins form adducts with nucleic acids in a variety of contexts, and these adducts may be cytotoxic if not repaired. Here we apply a proteomic approach to identification of proteins adducted to DNA or RNA in normally proliferating cells. This approach combines RADAR fractionation of proteins covalently bound to nucleic acids with quantitative mass spectrometry (MS). We demonstrate that "RADAR-MS" can quantify induction of TOP1- or TOP2-DNA adducts in cells treated with topotecan or etoposide, respectively, and also identify intermediates in physiological adduct repair. We validate RADAR-MS for discovery of previously unknown adducts by determining the repertoires of adducted proteins in two different normally proliferating human cell lines, CCRF-CEM T cells and GM639 fibroblasts. These repertoires are significantly similar with one another and exhibit robust correlations in their quantitative profiles (Spearman r = 0.52). A very similar repertoire is identified by the classical approach of CsCl buoyant density gradient centrifugation. We find that in normally proliferating human cells, the repertoire of adducted proteins - the "adductome" - is comprised of a limited number of proteins belonging to specific functional groups, and that it is greatly enriched for histones, HMG proteins and proteins involved in RNA splicing. Treatment with low concentrations of formaldehyde caused little change in the composition of the repertoire of adducted proteins, suggesting that reactive aldehydes generated by ongoing metabolic processes may contribute to protein adduction in normally proliferating cells. The identification of an endogenous adductome highlights the importance of adduct repair in maintaining genomic structure and the potential for deficiencies in adduct repair to contribute to cancer., Competing Interests: Declaration of Competing Interest The authors declare that there is no conflict of interest regarding the publication of this article., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
11. G-quadruplexes Sequester Free Heme in Living Cells.
- Author
-
Gray LT, Puig Lombardi E, Verga D, Nicolas A, Teulade-Fichou MP, Londoño-Vallejo A, and Maizels N
- Subjects
- Binding Sites, Cell Cycle Checkpoints drug effects, Cell Line, Tumor, Cell Survival drug effects, DNA, Catalytic metabolism, Heme chemistry, Humans, Iron metabolism, Ligands, Molecular Docking Simulation, Transcription, Genetic drug effects, Fused-Ring Compounds, G-Quadruplexes, Heme metabolism
- Abstract
Heme is an essential cofactor for many enzymes, but free heme is toxic and its levels are tightly regulated. G-quadruplexes bind heme avidly in vitro, raising the possibility that they may sequester heme in vivo. If so, then treatment that displaces heme from quadruplexes is predicted to induce expression of genes involved in iron and heme homeostasis. Here we show that PhenDC3, a G-quadruplex ligand structurally unrelated to heme, displaces quadruplex-bound heme in vitro and alters transcription in cultured human cells, upregulating genes that support heme degradation and iron homeostasis, and most strikingly causing a 30-fold induction of heme oxidase 1, the key enzyme in heme degradation. We propose that G-quadruplexes sequester heme to protect cells from the pathophysiological consequences of free heme., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
12. Activation-induced deaminase (AID) localizes to the nucleus in brief pulses.
- Author
-
Le Q and Maizels N
- Subjects
- B-Lymphocytes cytology, B-Lymphocytes enzymology, B-Lymphocytes ultrastructure, Cell Line, Cell Nucleus metabolism, Cytoplasm metabolism, Epigenesis, Genetic, Fibroblasts cytology, Fibroblasts enzymology, Fibroblasts ultrastructure, Genetic Variation, Humans, Immunoglobulins genetics, Microscopy, Fluorescence, Protein Transport, Cell Nucleus ultrastructure, Cytidine Deaminase metabolism, Cytoplasm ultrastructure, Single-Cell Analysis methods, Time-Lapse Imaging methods, Tumor Suppressor Protein p53 metabolism
- Abstract
Activation-induced deaminase (AID) converts C to U and 5-methyl-C to T. These mutagenic activities are critical to immunoglobulin (Ig) gene diversification and epigenetic reprogramming, but they must be tightly controlled to prevent compromising cell fitness. AID acts in the nucleus but localizes predominately to the cytoplasm. To address this apparent paradox, we have carried out time-lapse imaging of AID in single living B cells and fibroblasts. We demonstrate that AID enters the nucleus in brief (30 min) pulses, evident in about 10% of cells in the course of a single cell cycle (24 hr imaging). Pulses do not depend on AID catalytic activity, but they are coordinated with nuclear accumulation of P53. Pulsing may protect cells from pathologic consequences of excess exposure to AID, or enable AID to synchronize its activity with transcription of genes that are AID targets or with nuclear entry of factors that act at sites of AID-catalyzed DNA deamination to promote Ig gene diversification or epigenetic reprogramming., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
13. Increased levels of RECQ5 shift DNA repair from canonical to alternative pathways.
- Author
-
Olson HC, Davis L, Kiianitsa K, Khoo KJ, Liu Y, Knijnenburg TA, and Maizels N
- Subjects
- Computer Simulation, DNA Breaks, Double-Stranded, DNA End-Joining Repair genetics, DNA Repair genetics, Gene Amplification genetics, Gene Expression Regulation, Neoplastic, Humans, Mutagenesis, Neoplasms pathology, Recombinational DNA Repair genetics, Signal Transduction genetics, Genomic Instability genetics, Neoplasms genetics, Rad51 Recombinase genetics, RecQ Helicases genetics
- Abstract
RECQ5 (RECQL5) is one of several human helicases that dissociates RAD51-DNA filaments. The gene that encodes RECQ5 is frequently amplified in human tumors, but it is not known whether amplification correlates with increased gene expression, or how increased RECQ5 levels affect DNA repair at nicks and double-strand breaks. Here, we address these questions. We show that RECQ5 gene amplification correlates with increased gene expression in human tumors, by in silico analysis of over 9000 individual tumors representing 32 tumor types in the TCGA dataset. We demonstrate that, at double-strand breaks, increased RECQ5 levels inhibited canonical homology-directed repair (HDR) by double-stranded DNA donors, phenocopying the effect of BRCA deficiency. Conversely, at nicks, increased RECQ5 levels stimulated 'alternative' HDR by single-stranded DNA donors, which is normally suppressed by RAD51; this was accompanied by stimulation of mutagenic end-joining. Even modest changes (2-fold) in RECQ5 levels caused significant dysregulation of repair, especially HDR. These results suggest that in some tumors, RECQ5 gene amplification may have profound consequences for genomic instability.
- Published
- 2018
- Full Text
- View/download PDF
14. Initiation of homologous recombination at DNA nicks.
- Author
-
Maizels N and Davis L
- Subjects
- Antigenic Variation, DNA Breaks, Single-Stranded, DNA Replication, Escherichia coli genetics, F Factor genetics, Fimbriae Proteins genetics, G-Quadruplexes, Gene Conversion, Long Interspersed Nucleotide Elements, Saccharomyces cerevisiae genetics, DNA Damage, Recombinational DNA Repair
- Abstract
Discontinuities in only a single strand of the DNA duplex occur frequently, as a result of DNA damage or as intermediates in essential nuclear processes and DNA repair. Nicks are the simplest of these lesions: they carry clean ends bearing 3'-hydroxyl groups that can undergo ligation or prime new DNA synthesis. In contrast, single-strand breaks also interrupt only one DNA strand, but they carry damaged ends that require clean-up before subsequent steps in repair. Despite their apparent simplicity, nicks can have significant consequences for genome stability. The availability of enzymes that can introduce a nick almost anywhere in a large genome now makes it possible to systematically analyze repair of nicks. Recent experiments demonstrate that nicks can initiate recombination via pathways distinct from those active at double-strand breaks (DSBs). Recombination at targeted DNA nicks can be very efficient, and because nicks are intrinsically less mutagenic than DSBs, nick-initiated gene correction is useful for genome engineering and gene therapy. This review revisits some physiological examples of recombination at nicks, and outlines experiments that have demonstrated that nicks initiate homology-directed repair by distinctive pathways, emphasizing research that has contributed to our current mechanistic understanding of recombination at nicks in mammalian cells.
- Published
- 2018
- Full Text
- View/download PDF
15. Assaying Repair at DNA Nicks.
- Author
-
Davis L, Zhang Y, and Maizels N
- Subjects
- DNA metabolism, DNA End-Joining Repair, Genetic Techniques, Humans, Recombinational DNA Repair, DNA Breaks, DNA Repair
- Abstract
Nicks are the most common form of DNA damage, but they have only recently been shown to initiate damage that requires repair. Analysis of the pathways of nick repair in human cells has benefited from the development of enzymes that target nicks to specific sites in the genome and of reporters that enable rapid analysis of homology-directed repair and mutagenic end joining. Nicks undergo efficient repair by single-stranded oligonucleotide donors complementary to either the nicked or intact DNA strand, via pathways that are normally suppressed by RAD51. Here we discuss the details of reporter assays that take advantage of the convenience and sensitivity of flow cytometry to analyze pathways of repair at targeted DNA nicks. These assays are readily carried out in 96-well format cell culture plates, enabling mechanistic questions to be addressed by determining the contributions of specific factors by depletion and/or ectopic expression., (© 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
16. Two Distinct Pathways Support Gene Correction by Single-Stranded Donors at DNA Nicks.
- Author
-
Davis L and Maizels N
- Subjects
- Biomarkers metabolism, DNA Breaks, Double-Stranded, DNA Breaks, Single-Stranded, HEK293 Cells, Humans, Rad51 Recombinase metabolism, Recombinational DNA Repair, Replication Protein A metabolism, DNA Damage, DNA, Single-Stranded metabolism, Gene Editing
- Abstract
Nicks are the most common form of DNA damage. The mechanisms of their repair are fundamental to genomic stability and of practical importance for genome engineering. We define two pathways that support homology-directed repair by single-stranded DNA donors. One depends upon annealing-driven strand synthesis and acts at both nicks and double-strand breaks. The other depends upon annealing-driven heteroduplex correction and acts at nicks. Homology-directed repair via these pathways, as well as mutagenic end joining, are inhibited by RAD51 at nicks but largely independent of RAD51 at double-strand breaks. Guidelines for coordinated design of targets and donors for gene correction emerge from definition of these pathways. This analysis further suggests that naturally occurring nicks may have significant recombinogenic and mutagenic potential that is normally inhibited by RAD51 loading onto DNA, thereby identifying a function for RAD51 in maintenance of genomic stability., (Copyright © 2016 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
17. The Werner syndrome RECQ helicase targets G4 DNA in human cells to modulate transcription.
- Author
-
Tang W, Robles AI, Beyer RP, Gray LT, Nguyen GH, Oshima J, Maizels N, Harris CC, and Monnat RJ Jr
- Subjects
- Carcinogenesis genetics, DNA-Binding Proteins metabolism, Fibroblasts, G-Quadruplexes, Gene Expression Regulation, Genome, Human, Humans, MicroRNAs, Neoplasms pathology, Nucleotide Motifs, RecQ Helicases metabolism, DNA-Binding Proteins genetics, Genomic Instability genetics, Neoplasms genetics, RecQ Helicases genetics, Werner Syndrome genetics
- Abstract
The Werner syndrome (WS) is a prototypic adult Mendelian progeroid syndrome in which signs of premature aging are associated with genomic instability and an elevated risk of cancer. The WRN RECQ helicase protein binds and unwinds G-quadruplex (G4) DNA substrates in vitro, and we identified significant enrichment in G4 sequence motifs at the transcription start site and 5' ends of first introns (false discovery rate < 0.001) of genes down-regulated in WS patient fibroblasts. This finding provides strong evidence that WRN binds G4 DNA structures at many chromosomal sites to modulate gene expression. WRN appears to bind a distinct subpopulation of G4 motifs in human cells, when compared with the related Bloom syndrome RECQ helicase protein. Functional annotation of the genes and miRNAs altered in WS provided new insight into WS disease pathogenesis. WS patient fibroblasts displayed altered expression of multiple, mechanistically distinct, senescence-associated gene expression programs, with altered expression of disease-associated miRNAs, and dysregulation of canonical pathways that regulate cell signaling, genome stability and tumorigenesis. WS fibroblasts also displayed a highly statistically significant and distinct gene expression signature, with coordinate overexpression of nearly all of the cytoplasmic tRNA synthetases and associated ARS-interacting multifunctional protein genes. The 'non-canonical' functions of many of these upregulated tRNA charging proteins may together promote WS disease pathogenesis. Our results identify the human WRN RECQ protein as a G4 helicase that modulates gene expression in G4-dependent fashion at many chromosomal sites and provide several new and unexpected mechanistic insights into WS disease pathogenesis., (© The Author 2016. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
18. Cell Cycle Regulates Nuclear Stability of AID and Determines the Cellular Response to AID.
- Author
-
Le Q and Maizels N
- Subjects
- Biocatalysis, Cell Line, Enzyme Stability, Humans, Phosphorylation, Proteolysis, Ubiquitin metabolism, Cell Cycle, Cell Nucleus enzymology, Cytidine Deaminase metabolism
- Abstract
AID (Activation Induced Deaminase) deaminates cytosines in DNA to initiate immunoglobulin gene diversification and to reprogram CpG methylation in early development. AID is potentially highly mutagenic, and it causes genomic instability evident as translocations in B cell malignancies. Here we show that AID is cell cycle regulated. By high content screening microscopy, we demonstrate that AID undergoes nuclear degradation more slowly in G1 phase than in S or G2-M phase, and that mutations that affect regulatory phosphorylation or catalytic activity can alter AID stability and abundance. We directly test the role of cell cycle regulation by fusing AID to tags that destabilize nuclear protein outside of G1 or S-G2/M phases. We show that enforced nuclear localization of AID in G1 phase accelerates somatic hypermutation and class switch recombination, and is well-tolerated; while nuclear AID compromises viability in S-G2/M phase cells. We identify AID derivatives that accelerate somatic hypermutation with minimal impact on viability, which will be useful tools for engineering genes and proteins by iterative mutagenesis and selection. Our results further suggest that use of cell cycle tags to regulate nuclear stability may be generally applicable to studying DNA repair and to engineering the genome.
- Published
- 2015
- Full Text
- View/download PDF
19. G4-associated human diseases.
- Author
-
Maizels N
- Subjects
- Animals, DNA genetics, Genomic Instability, Humans, Neoplasms genetics, Disease genetics, G-Quadruplexes, Genome, Human, Nervous System Diseases genetics
- Abstract
Recent research has established clear connections between G-quadruplexes and human disease. Features of quadruplex structures that promote genomic instability have been determined. Quadruplexes have been identified as transcriptional, translational and epigenetic regulatory targets of factors associated with human genetic disease. An expandable GGGGCC motif that can adopt a G4 structure, located in the previously obscure C9ORF72 locus, has been shown to contribute to two well-recognized neurodegenerative diseases, amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). This review focuses on these advances, which further dispel the view that genomic biology is limited to the confines of the canonical B-form DNA duplex, and show how quadruplexes contribute spatial and temporal dimensionalities to linear sequence information. This recent progress also has clear practical ramifications, as prevention, diagnosis, and treatment of disease depend on understanding the underlying mechanisms., (© 2015 The Author.)
- Published
- 2015
- Full Text
- View/download PDF
20. MRE11-deficiency associated with improved long-term disease free survival and overall survival in a subset of stage III colon cancer patients in randomized CALGB 89803 trial.
- Author
-
Pavelitz T, Renfro L, Foster NR, Caracol A, Welsch P, Lao VV, Grady WB, Niedzwiecki D, Saltz LB, Bertagnolli MM, Goldberg RM, Rabinovitch PS, Emond M, Monnat RJ Jr, and Maizels N
- Subjects
- Adult, Aged, Aged, 80 and over, Camptothecin administration & dosage, Camptothecin analogs & derivatives, Camptothecin therapeutic use, Colonic Neoplasms drug therapy, DNA-Binding Proteins metabolism, Disease-Free Survival, Female, Fluorouracil administration & dosage, Fluorouracil therapeutic use, Follow-Up Studies, Genetic Association Studies, Humans, Irinotecan, Leucovorin administration & dosage, Leucovorin therapeutic use, MRE11 Homologue Protein, Male, Middle Aged, Polymorphism, Single Nucleotide, Prognosis, Treatment Outcome, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colonic Neoplasms genetics, Colonic Neoplasms mortality, DNA-Binding Proteins genetics
- Abstract
Purpose: Colon cancers deficient in mismatch repair (MMR) may exhibit diminished expression of the DNA repair gene, MRE11, as a consequence of contraction of a T11 mononucleotide tract. This study investigated MRE11 status and its association with prognosis, survival and drug response in patients with stage III colon cancer., Patients and Methods: Cancer and Leukemia Group B 89803 (Alliance) randomly assigned 1,264 patients with stage III colon cancer to postoperative weekly adjuvant bolus 5-fluorouracil/leucovorin (FU/LV) or irinotecan+FU/LV (IFL), with 8 year follow-up. Tumors from these patients were analyzed to determine stability of a T11 tract in the MRE11 gene. The primary endpoint was overall survival (OS), and a secondary endpoint was disease-free survival (DFS). Non-proportional hazards were addressed using time-dependent covariates in Cox analyses., Results: Of 625 tumor cases examined, 70 (11.2%) exhibited contraction at the T11 tract in one or both MRE11 alleles and were thus predicted to be deficient in MRE11 (dMRE11). In pooled treatment analyses, dMRE11 patients showed initially reduced DFS and OS but improved long-term DFS and OS compared with patients with an intact MRE11 T11 tract. In the subgroup of dMRE11 patients treated with IFL, an unexplained early increase in mortality but better long-term DFS than IFL-treated pMRE11 patients was observed., Conclusions: Analysis of this relatively small number of patients and events showed that the dMRE11 marker predicts better prognosis independent of treatment in the long-term. In subgroup analyses, dMRE11 patients treated with irinotecan exhibited unexplained short-term mortality. MRE11 status is readily assayed and may therefore prove to be a useful prognostic marker, provided that the results reported here for a relatively small number of patients can be generalized in independent analyses of larger numbers of samples., Trial Registration: ClinicalTrials.gov NCT00003835.
- Published
- 2014
- Full Text
- View/download PDF
21. CpG island methylator phenotype is associated with response to adjuvant irinotecan-based therapy for stage III colon cancer.
- Author
-
Shiovitz S, Bertagnolli MM, Renfro LA, Nam E, Foster NR, Dzieciatkowski S, Luo Y, Lao VV, Monnat RJ Jr, Emond MJ, Maizels N, Niedzwiecki D, Goldberg RM, Saltz LB, Venook A, Warren RS, and Grady WM
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma mortality, Adenocarcinoma pathology, Adult, Aged, Aged, 80 and over, Camptothecin administration & dosage, Camptothecin analogs & derivatives, Chemotherapy, Adjuvant, Colectomy, Colonic Neoplasms genetics, Colonic Neoplasms mortality, Colonic Neoplasms pathology, DNA Mismatch Repair, Disease-Free Survival, Female, Fluorouracil administration & dosage, Humans, Irinotecan, Kaplan-Meier Estimate, Leucovorin administration & dosage, Male, Middle Aged, Neoplasm Staging, Phenotype, Proportional Hazards Models, Risk Assessment, Risk Factors, Time Factors, Treatment Outcome, Young Adult, Adenocarcinoma drug therapy, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colonic Neoplasms drug therapy, CpG Islands, DNA Methylation
- Abstract
Background & Aims: The CpG island methylator phenotype (CIMP), defined by a high frequency of aberrantly methylated genes, is a characteristic of a subclass of colon tumors with distinct clinical and molecular features. Cohort studies have produced conflicting results on responses of CIMP-positive tumors to chemotherapy. We assessed the association between tumor CIMP status and survival of patients receiving adjuvant fluorouracil and leucovorin alone or with irinotecan (IFL)., Methods: We analyzed data from patients with stage III colon adenocarcinoma randomly assigned to groups given fluorouracil and leucovorin or IFL after surgery, from April 1999 through April 2001. The primary end point of the trial was overall survival and the secondary end point was disease-free survival. DNA isolated from available tumor samples (n = 615) was used to determine CIMP status based on methylation patterns at the CACNA1G, IGF2, NEUROG1, RUNX3, and SOCS1 loci. The effects of CIMP on survival were modeled using Kaplan-Meier and Cox proportional hazards; interactions with treatment and BRAF, KRAS, and mismatch repair (MMR) status were also investigated., Results: Of the tumor samples characterized for CIMP status, 145 were CIMP positive (23%). Patients with CIMP-positive tumors had shorter overall survival times than patients with CIMP-negative tumors (hazard ratio = 1.36; 95% confidence interval: 1.01-1.84). Treatment with IFL showed a trend toward increased overall survival for patients with CIMP-positive tumors, compared with treatment with fluorouracil and leucovorin (hazard ratio = 0.62; 95% CI: 0.37-1.05; P = .07), but not for patients with CIMP-negative tumors (hazard ratio = 1.38; 95% CI: 1.00-1.89; P = .049). In a 3-way interaction analysis, patients with CIMP-positive, MMR-intact tumors benefited most from the addition of irinotecan to fluorouracil and leucovorin therapy (for the interaction, P = .01). CIMP was more strongly associated with response to IFL than MMR status. Results for disease-free survival times were comparable among all analyses., Conclusions: Patients with stage III, CIMP-positive, MMR-intact colon tumors have longer survival times when irinotecan is added to combination therapy with fluorouracil and leucovorin., (Copyright © 2014 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
22. Regulation of gene expression by the BLM helicase correlates with the presence of G-quadruplex DNA motifs.
- Author
-
Nguyen GH, Tang W, Robles AI, Beyer RP, Gray LT, Welsh JA, Schetter AJ, Kumamoto K, Wang XW, Hickson ID, Maizels N, Monnat RJ Jr, and Harris CC
- Subjects
- Cells, Cultured, Gene Expression Profiling, Humans, RNA, Messenger genetics, Bloom Syndrome genetics, DNA chemistry, G-Quadruplexes, Gene Expression Regulation, Enzymologic, RecQ Helicases genetics
- Abstract
Bloom syndrome is a rare autosomal recessive disorder characterized by genetic instability and cancer predisposition, and caused by mutations in the gene encoding the Bloom syndrome, RecQ helicase-like (BLM) protein. To determine whether altered gene expression might be responsible for pathological features of Bloom syndrome, we analyzed mRNA and microRNA (miRNA) expression in fibroblasts from individuals with Bloom syndrome and in BLM-depleted control fibroblasts. We identified mRNA and miRNA expression differences in Bloom syndrome patient and BLM-depleted cells. Differentially expressed mRNAs are connected with cell proliferation, survival, and molecular mechanisms of cancer, and differentially expressed miRNAs target genes involved in cancer and in immune function. These and additional altered functions or pathways may contribute to the proportional dwarfism, elevated cancer risk, immune dysfunction, and other features observed in Bloom syndrome individuals. BLM binds to G-quadruplex (G4) DNA, and G4 motifs were enriched at transcription start sites (TSS) and especially within first introns (false discovery rate ≤ 0.001) of differentially expressed mRNAs in Bloom syndrome compared with normal cells, suggesting that G-quadruplex structures formed at these motifs are physiologic targets for BLM. These results identify a network of mRNAs and miRNAs that may drive the pathogenesis of Bloom syndrome.
- Published
- 2014
- Full Text
- View/download PDF
23. Ultrasensitive isolation, identification and quantification of DNA-protein adducts by ELISA-based RADAR assay.
- Author
-
Kiianitsa K and Maizels N
- Subjects
- Antigens, Neoplasm analysis, Cell Line, Cell Line, Tumor, DNA Adducts isolation & purification, DNA Gyrase analysis, DNA Repair, DNA Topoisomerases, Type I analysis, DNA Topoisomerases, Type II analysis, DNA-Binding Proteins isolation & purification, Escherichia coli enzymology, Escherichia coli genetics, Humans, Poly-ADP-Ribose Binding Proteins, DNA Adducts analysis, DNA-Binding Proteins analysis, Enzyme-Linked Immunosorbent Assay methods
- Abstract
Enzymes that form transient DNA-protein covalent complexes are targets for several potent classes of drugs used to treat infectious disease and cancer, making it important to establish robust and rapid procedures for analysis of these complexes. We report a method for isolation of DNA-protein adducts and their identification and quantification, using techniques compatible with high-throughput screening. This method is based on the RADAR assay for DNA adducts that we previously developed (Kiianitsa and Maizels (2013) A rapid and sensitive assay for DNA-protein covalent complexes in living cells. Nucleic Acids Res., 41:e104), but incorporates three key new steps of broad applicability. (i) Silica-assisted ethanol/isopropanol precipitation ensures reproducible and efficient recovery of DNA and DNA-protein adducts at low centrifugal forces, enabling cell culture and DNA precipitation to be carried out in a single microtiter plate. (ii) Rigorous purification of DNA-protein adducts by a procedure that eliminates free proteins and free nucleic acids, generating samples suitable for detection of novel protein adducts (e.g. by mass spectroscopy). (iii) Identification and quantification of DNA-protein adducts by direct ELISA assay. The ELISA-based RADAR assay can detect Top1-DNA and Top2a-DNA adducts in human cells, and gyrase-DNA adducts in Escherichia coli. This approach will be useful for discovery and characterization of new drugs to treat infectious disease and cancer, and for development of companion diagnostics assays for individualized medicine., (© The Author(s) 2014. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2014
- Full Text
- View/download PDF
24. G quadruplexes are genomewide targets of transcriptional helicases XPB and XPD.
- Author
-
Gray LT, Vallur AC, Eddy J, and Maizels N
- Subjects
- Capillary Electrochromatography, Cell Line, DNA genetics, DNA Helicases metabolism, Genetic Vectors, Humans, Microarray Analysis, Point Mutation genetics, Point Mutation physiology, RNA biosynthesis, RNA genetics, Sequence Alignment, Sequence Analysis, DNA, Signal Transduction genetics, DNA Helicases genetics, DNA-Binding Proteins genetics, G-Quadruplexes, Gene Expression Regulation genetics, Genome genetics, Xeroderma Pigmentosum Group D Protein genetics
- Abstract
G4 motifs are greatly enriched near promoters, suggesting that quadruplex structures may be targets of transcriptional regulation. Here we show, by ChIP-Seq analysis of human cells, that 40% of the binding sites of the transcription-associated helicases, XPB and XPD, overlap with G4 motifs. The highly significant overlap of XPB and XPD binding sites with G4 motifs cannot be explained by GC richness or parameters of the genomewide analysis, but instead suggests that these proteins are recruited to quadruplex structures that form in genomic DNA (G4 DNA). Biochemical analysis demonstrates that XPD is a robust G4 DNA helicase and that XPB binds G4 DNA. XPB and XPD are enriched near the transcription start site at 20% of genes, especially highly transcribed genes. XPB and XPD enrichment at G4 motifs characterizes specific signaling pathways and regulatory pathways associated with specific cancers. These results identify new candidate pathways for therapies targeted to quadruplexes.
- Published
- 2014
- Full Text
- View/download PDF
25. Homology-directed repair of DNA nicks via pathways distinct from canonical double-strand break repair.
- Author
-
Davis L and Maizels N
- Subjects
- BRCA2 Protein genetics, Cell Line, Flow Cytometry, Gene Expression Regulation physiology, Genetic Engineering methods, Humans, RNA, Small Interfering genetics, Rad51 Recombinase genetics, Recombinational DNA Repair genetics, Reverse Transcriptase Polymerase Chain Reaction, DNA Breaks, Single-Stranded, Models, Genetic, Recombinational DNA Repair physiology
- Abstract
DNA nicks are the most common form of DNA damage, and if unrepaired can give rise to genomic instability. In human cells, nicks are efficiently repaired via the single-strand break repair pathway, but relatively little is known about the fate of nicks not processed by that pathway. Here we show that homology-directed repair (HDR) at nicks occurs via a mechanism distinct from HDR at double-strand breaks (DSBs). HDR at nicks, but not DSBs, is associated with transcription and is eightfold more efficient at a nick on the transcribed strand than at a nick on the nontranscribed strand. HDR at nicks can proceed by a pathway dependent upon canonical HDR factors RAD51 and BRCA2; or by an efficient alternative pathway that uses either ssDNA or nicked dsDNA donors and that is strongly inhibited by RAD51 and BRCA2. Nicks generated by either I-AniI or the CRISPR/Cas9(D10A) nickase are repaired by the alternative HDR pathway with little accompanying mutagenic end-joining, so this pathway may be usefully applied to genome engineering. These results suggest that alternative HDR at nicks may be stimulated in physiological contexts in which canonical RAD51/BRCA2-dependent HDR is compromised or down-regulated, which occurs frequently in tumors.
- Published
- 2014
- Full Text
- View/download PDF
26. Novel fluorescent genome editing reporters for monitoring DNA repair pathway utilization at endonuclease-induced breaks.
- Author
-
Kuhar R, Gwiazda KS, Humbert O, Mandt T, Pangallo J, Brault M, Khan I, Maizels N, Rawlings DJ, Scharenberg AM, and Certo MT
- Subjects
- Flow Cytometry, Fluorescence, Gene Silencing, Genes, Genetic Loci, Genome, Genomics methods, HEK293 Cells, Humans, Luminescent Proteins genetics, Transcription, Genetic, DNA Breaks, Double-Stranded, DNA Repair, Endodeoxyribonucleases, Genes, Reporter
- Abstract
The creation of a DNA break at a specific locus by a designer endonuclease can be harnessed to edit a genome. However, DNA breaks may engage one of several competing repair pathways that lead to distinct types of genomic alterations. Therefore, understanding the contribution of different repair pathways following the introduction of a targeted DNA break is essential to further advance the safety and efficiency of nuclease-induced genome modification. To gain insight into the role of different DNA repair pathways in resolving nuclease-induced DNA breaks into genome editing outcomes, we previously developed a fluorescent-based reporter system, designated the Traffic Light Reporter, which provides a readout of gene targeting and gene disruption downstream of a targeted DNA double-strand break. Here we describe two related but novel reporters that extend this technology: one that allows monitoring of the transcriptional activity at the reporter locus, and thus can be applied to interrogate break resolution at active and repressed loci; and a second that reads out single-strand annealing in addition to gene targeting and gene disruption. Application of these reporters to assess repair pathway usage in several common gene editing contexts confirms the importance that chromatin status and initiation of end resection have on the resolution of nuclease-induced breaks.
- Published
- 2014
- Full Text
- View/download PDF
27. Genome engineering with Cre-loxP.
- Author
-
Maizels N
- Subjects
- Animals, Humans, Gene Targeting methods, Genes, Switch, Immunoglobulin Class Switching genetics, Integrases genetics, Recombination, Genetic
- Published
- 2013
- Full Text
- View/download PDF
28. Molecular and genetic bases of disease.
- Author
-
Maizels N and Lupski JR
- Subjects
- Base Sequence, Disease etiology, Humans, Chromosomes genetics, Disease genetics, Genome, Human
- Published
- 2013
- Full Text
- View/download PDF
29. A rapid and sensitive assay for DNA-protein covalent complexes in living cells.
- Author
-
Kiianitsa K and Maizels N
- Subjects
- Azacitidine analogs & derivatives, Azacitidine chemistry, Camptothecin toxicity, Cell Line, DNA (Cytosine-5-)-Methyltransferase 1, DNA (Cytosine-5-)-Methyltransferases analysis, DNA (Cytosine-5-)-Methyltransferases immunology, DNA Adducts chemistry, DNA Topoisomerases, Type I analysis, DNA Topoisomerases, Type I immunology, DNA-Binding Proteins chemistry, Decitabine, Humans, Nuclear Proteins antagonists & inhibitors, Topoisomerase I Inhibitors toxicity, DNA Adducts analysis, DNA-Binding Proteins analysis, Immunoassay methods
- Abstract
A number of proteins form covalent bonds with DNA as obligatory transient intermediates in normal nuclear transactions. Drugs that trap these complexes have proven to be potent therapeutics in both cancer and infectious disease. Nonetheless, current assays for DNA-protein adducts are cumbersome, limiting both mechanistic studies and translational applications. We have developed a rapid and sensitive assay that enables quantitative immunodetection of protein-DNA adducts. This new 'RADAR' (rapid approach to DNA adduct recovery) assay accelerates processing time 4-fold, increases sample throughput 20-fold and requires 50-fold less starting material than the current standard. It can be used to detect topoisomerase 1-DNA adducts in as little as 60 ng of DNA, corresponding to 10 000 human cells. We apply the RADAR assay to demonstrate that expression of SLFN11 does not increase camptothecin sensitivity by promoting accumulation of topoisomerase 1-DNA adducts. The RADAR assay will be useful for analysis of the mechanisms of formation and resolution of DNA-protein adducts in living cells, and identification and characterization of reactions in which covalent DNA adducts are transient intermediates. The assay also has potential application to drug discovery and individualized medicine.
- Published
- 2013
- Full Text
- View/download PDF
30. The G4 genome.
- Author
-
Maizels N and Gray LT
- Subjects
- DNA chemistry, DNA Helicases genetics, Humans, DNA Replication, G-Quadruplexes
- Abstract
Recent experiments provide fascinating examples of how G4 DNA and G4 RNA structures--aka quadruplexes--may contribute to normal biology and to genomic pathologies. Quadruplexes are transient and therefore difficult to identify directly in living cells, which initially caused skepticism regarding not only their biological relevance but even their existence. There is now compelling evidence for functions of some G4 motifs and the corresponding quadruplexes in essential processes, including initiation of DNA replication, telomere maintenance, regulated recombination in immune evasion and the immune response, control of gene expression, and genetic and epigenetic instability. Recognition and resolution of quadruplex structures is therefore an essential component of genome biology. We propose that G4 motifs and structures that participate in key processes compose the G4 genome, analogous to the transcriptome, proteome, or metabolome. This is a new view of the genome, which sees DNA as not only a simple alphabet but also a more complex geography. The challenge for the future is to systematically identify the G4 motifs that form quadruplexes in living cells and the features that confer on specific G4 motifs the ability to function as structural elements., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2013
- Full Text
- View/download PDF
31. Epigenetic modification of the repair donor regulates targeted gene correction.
- Author
-
Humbert O and Maizels N
- Abstract
Optimizing design of vectors is critical to effective gene therapy. In targeted gene correction (TGC), cleavage of chromosomal DNA near a mutation stimulates homology-directed repair of a target gene using a donor provided in trans. We have systematically addressed epigenetic parameters of donor design, using a flow-based assay to quantify correction frequencies and expression levels of a green fluorescent protein (GFP) reporter gene in a human cell line. We show that active transcription of the donor increased correction frequency by threefold, establishing that a proximal promoter enhances donor use. Conversely, CpG methylation of the donor diminished correction frequency and reduced expression of the repaired gene. However, bisulfite sequencing of the target revealed no transfer of methylation marks during repair with a methylated donor. Treatment with histone deacetylase (HDAC) inhibitors can partially compensate for epigenetic inactivation, suggesting a role for class I and II HDACs in regulation of donor use. These results establish that epigenetic status of a trans-donor determines both the efficiency and outcome of gene correction, and identify and clarify parameters that should guide donor design for targeted gene therapy.Molecular Therapy - Nucleic Acids (2012) 1, e49; doi:10.1038/mtna.2012.42; published online 23 October 2012.
- Published
- 2012
- Full Text
- View/download PDF
32. G4 motifs in human genes.
- Author
-
Maizels N
- Subjects
- Base Sequence, DNA Replication, DNA, Single-Stranded, Humans, Molecular Sequence Data, Mutagenesis, Mutation, Nucleic Acid Conformation, Polymorphism, Genetic, Regulatory Sequences, Nucleic Acid, Transcription, Genetic, Genome, Human, Genomic Instability
- Abstract
The G4 motif, G(≥3) N(x) G(≥3) N(x) G(≥3) N(x) G(≥3) , is enriched in some genomic regions and depleted in others. This motif confers the ability to form an unusual four-stranded DNA structure, G4 DNA. G4 DNA is associated with genomic instability, which may explain depletion of G4 motifs from some genes and genomic regions. Conversely, G4 motifs are enriched downstream of transcription start sites, where they correlate with pausing. The uneven distribution of G4 motifs in the genome strongly suggests that mechanisms of selection act not only on one-dimensional genomic sequence, but also on structures formed by genomic DNA. The biological roles of G4 structures illustrate that, to understand genome function, it is important to consider the dynamic structural potential implicit in the G4 motif., (© 2012 New York Academy of Sciences.)
- Published
- 2012
- Full Text
- View/download PDF
33. Targeted gene therapies: tools, applications, optimization.
- Author
-
Humbert O, Davis L, and Maizels N
- Subjects
- Chromosomes, Human genetics, DNA Breaks, DNA Cleavage, DNA End-Joining Repair, Endonucleases therapeutic use, Genetic Diseases, Inborn genetics, Genome, Human, Humans, Mutation, Protein Engineering, Recombinational DNA Repair, Targeted Gene Repair standards, Transgenes, Genetic Diseases, Inborn therapy, Targeted Gene Repair methods
- Abstract
Many devastating human diseases are caused by mutations in a single gene that prevent a somatic cell from carrying out its essential functions, or by genetic changes acquired as a result of infectious disease or in the course of cell transformation. Targeted gene therapies have emerged as potential strategies for treatment of such diseases. These therapies depend upon rare-cutting endonucleases to cleave at specific sites in or near disease genes. Targeted gene correction provides a template for homology-directed repair, enabling the cell's own repair pathways to erase the mutation and replace it with the correct sequence. Targeted gene disruption ablates the disease gene, disabling its function. Gene targeting can also promote other kinds of genome engineering, including mutation, insertion, or gene deletion. Targeted gene therapies present significant advantages compared to approaches to gene therapy that depend upon delivery of stably expressing transgenes. Recent progress has been fueled by advances in nuclease discovery and design, and by new strategies that maximize efficiency of targeting and minimize off-target damage. Future progress will build on deeper mechanistic understanding of critical factors and pathways.
- Published
- 2012
- Full Text
- View/download PDF
34. Antibody discovery ex vivo accelerated by the LacO/LacI regulatory network.
- Author
-
Yabuki M, Cummings WJ, Leppard JB, Immormino RM, Wood CL, Allison DS, Gray PW, Tjoelker LW, and Maizels N
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal, Humanized chemistry, Antibodies, Monoclonal, Humanized immunology, Antibody Affinity immunology, Cell Line, Chickens, Clone Cells, Complementarity Determining Regions genetics, Conserved Sequence genetics, Genetic Engineering, Humans, Immunoglobulin Heavy Chains immunology, Immunoglobulin Variable Region chemistry, Immunoglobulin Variable Region immunology, Molecular Sequence Data, Mutation genetics, Receptors, Cell Surface immunology, Streptavidin immunology, Antibodies, Monoclonal immunology, Gene Regulatory Networks genetics, Lac Operon genetics, Lac Repressors genetics
- Abstract
Monoclonal antibodies (mAbs) can be potent and highly specific therapeutics, diagnostics and research reagents. Nonetheless, mAb discovery using current in vivo or in vitro approaches can be costly and time-consuming, with no guarantee of success. We have established a platform for rapid discovery and optimization of mAbs ex vivo. This DTLacO platform derives from a chicken B cell line that has been engineered to enable rapid selection and seamless maturation of high affinity mAbs. We have validated the DTLacO platform by generation of high affinity and specific mAbs to five cell surface targets, the receptor tyrosine kinases VEGFR2 and TIE2, the glycoprotein TROP2, the small TNF receptor family member FN14, and the G protein-coupled receptor FZD10. mAb discovery is rapid and humanization is straightforward, establishing the utility of the DTLacO platform for identification of mAbs for therapeutic and other applications.
- Published
- 2012
- Full Text
- View/download PDF
35. DNA repair factor MRE11/RAD50 cleaves 3'-phosphotyrosyl bonds and resects DNA to repair damage caused by topoisomerase 1 poisons.
- Author
-
Sacho EJ and Maizels N
- Subjects
- Archaeal Proteins antagonists & inhibitors, Archaeal Proteins metabolism, DNA Breaks, Double-Stranded, DNA-Binding Proteins metabolism, Pyrococcus furiosus metabolism, Archaeal Proteins chemistry, Camptothecin chemistry, DNA Repair, DNA Topoisomerases, Type I chemistry, DNA-Binding Proteins chemistry, Pyrococcus furiosus chemistry, Topoisomerase I Inhibitors chemistry
- Abstract
MRE11-RAD50 is a highly conserved multifunctional DNA repair factor. Here, we show that MRE11-RAD50 cleaves the covalent 3'-phosphotyrosyl-DNA bonds that join topoisomerase 1 (Top1) to the DNA backbone and that are the hallmark of damage caused by Top1 poisons such as camptothecin. Cleavage generates a 3'-phosphate DNA end that MRE11-RAD50 can resect in an ATP-regulated reaction, to produce a 3'-hydroxyl that can prime repair synthesis. The 3'-phosphotyrosyl cleavage activity maps to the MRE11 active site. These results define a new activity of MRE11 and distinguish MRE11-RAD50 functions in repair of Top1-DNA complexes and double-strand breaks.
- Published
- 2011
- Full Text
- View/download PDF
36. G4 DNA: at risk in the genome.
- Author
-
Davis L and Maizels N
- Subjects
- DNA Replication, G-Quadruplexes
- Published
- 2011
- Full Text
- View/download PDF
37. G4 motifs correlate with promoter-proximal transcriptional pausing in human genes.
- Author
-
Eddy J, Vallur AC, Varma S, Liu H, Reinhold WC, Pommier Y, and Maizels N
- Subjects
- Binding Sites, Cell Line, Tumor, CpG Islands, Heterogeneous-Nuclear Ribonucleoproteins metabolism, Humans, Introns, RNA Polymerase II metabolism, Transcription Factors metabolism, Transcription Initiation Site, Guanine analysis, Promoter Regions, Genetic, Regulatory Elements, Transcriptional, Transcription, Genetic
- Abstract
The RNA Pol II transcription complex pauses just downstream of the promoter in a significant fraction of human genes. The local features of genomic structure that contribute to pausing have not been defined. Here, we show that genes that pause are more G-rich within the region flanking the transcription start site (TSS) than RefSeq genes or non-paused genes. We show that enrichment of binding motifs for common transcription factors, such as SP1, may account for G-richness upstream but not downstream of the TSS. We further show that pausing correlates with the presence of a GrIn1 element, an element bearing one or more G4 motifs at the 5'-end of the first intron, on the non-template DNA strand. These results suggest potential roles for dynamic G4 DNA and G4 RNA structures in cis-regulation of pausing, and thus genome-wide regulation of gene expression, in human cells.
- Published
- 2011
- Full Text
- View/download PDF
38. DNA nicks promote efficient and safe targeted gene correction.
- Author
-
Davis L and Maizels N
- Subjects
- DNA Breaks, Double-Stranded, DNA End-Joining Repair genetics, Endodeoxyribonucleases genetics, Endodeoxyribonucleases metabolism, HEK293 Cells, Humans, DNA Breaks, Single-Stranded, Targeted Gene Repair methods
- Abstract
Targeted gene correction employs a site-specific DNA lesion to promote homologous recombination that eliminates mutation in a disease gene of interest. The double-strand break typically used to initiate correction can also result in genomic instability if deleterious repair occurs rather than gene correction, possibly compromising the safety of targeted gene correction. Here we show that single-strand breaks (nicks) and double-strand breaks both promote efficient gene correction. However, breaks promote high levels of inadvertent but heritable genomic alterations both locally and elsewhere in the genome, while nicks are accompanied by essentially no collateral local mutagenesis, and thus provide a safer approach to gene correction. Defining efficacy as the ratio of gene correction to local deletion, nicks initiate gene correction with 70-fold greater efficacy than do double-strand breaks (29.0±6.0% and 0.42±0.03%, respectively). Thus nicks initiate efficient gene correction, with limited local mutagenesis. These results have clear therapeutic implications, and should inform future design of meganucleases for targeted gene correction.
- Published
- 2011
- Full Text
- View/download PDF
39. MRE11 function in response to topoisomerase poisons is independent of its function in double-strand break repair in Saccharomyces cerevisiae.
- Author
-
Hamilton NK and Maizels N
- Subjects
- DNA Damage, DNA Repair, DNA Topoisomerases, Type I metabolism, Endodeoxyribonucleases genetics, Exodeoxyribonucleases genetics, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics
- Abstract
Camptothecin (CPT) and etoposide (ETP) trap topoisomerase-DNA covalent intermediates, resulting in formation of DNA damage that can be cytotoxic if unrepaired. CPT and ETP are prototypes for molecules widely used in chemotherapy of cancer, so defining the mechanisms for repair of damage induced by treatment with these compounds is of great interest. In S. cerevisiae, deficiency in MRE11, which encodes a highly conserved factor, greatly enhances sensitivity to treatment with CPT or ETP. This has been thought to reflect the importance of double-strand break (DSB) repair pathways in the response to these to agents. Here we report that an S. cerevisiae strain expressing the mre11-H59A allele, mutant at a conserved active site histidine, is sensitive to hydroxyurea and also to ionizing radiation, which induces DSBs, but not to CPT or ETP. We show that TDP1, which encodes a tyrosyl-DNA phosphodiesterase activity able to release both 5'- and 3'-covalent topoisomerase-DNA complexes in vitro, contributes to ETP-resistance but not CPT-resistance in the mre11-H59A background. We further show that CPT- and ETP-resistance mediated by MRE11 is independent of SAE2, and thus independent of the coordinated functions of MRE11 and SAE2 in homology-directed repair and removal of Spo11 from DNA ends in meiosis. These results identify a function for MRE11 in the response to topoisomerase poisons that is distinct from its functions in DSB repair or meiotic DNA processing. They also establish that cellular proficiency in repair of DSBs may not correlate with resistance to topoisomerase poisons, a finding with potential implications for stratification of tumors with specific DNA repair deficiencies for treatment with these compounds.
- Published
- 2010
- Full Text
- View/download PDF
40. Complementary roles for exonuclease 1 and Flap endonuclease 1 in maintenance of triplet repeats.
- Author
-
Vallur AC and Maizels N
- Subjects
- Animals, DNA Footprinting methods, DNA Repair Enzymes chemistry, DNA Repair Enzymes genetics, Exodeoxyribonucleases chemistry, Exodeoxyribonucleases genetics, Flap Endonucleases chemistry, Flap Endonucleases genetics, Humans, Protein Binding physiology, DNA Repair Enzymes metabolism, DNA Replication physiology, Exodeoxyribonucleases metabolism, Flap Endonucleases metabolism, Trinucleotide Repeats physiology
- Abstract
Trinucleotide repeats can form stable secondary structures that promote genomic instability. To determine how such structures are resolved, we have defined biochemical activities of the related RAD2 family nucleases, FEN1 (Flap endonuclease 1) and EXO1 (exonuclease 1), on substrates that recapitulate intermediates in DNA replication. Here, we show that, consistent with its function in lagging strand replication, human (h) FEN1 could cleave 5'-flaps bearing structures formed by CTG or CGG repeats, although less efficiently than unstructured flaps. hEXO1 did not exhibit endonuclease activity on 5'-flaps bearing structures formed by CTG or CGG repeats, although it could excise these substrates. Neither hFEN1 nor hEXO1 was affected by the stem-loops formed by CTG repeats interrupting duplex regions adjacent to 5'-flaps, but both enzymes were inhibited by G4 structures formed by CGG repeats in analogous positions. Hydroxyl radical footprinting showed that hFEN1 binding caused hypersensitivity near the flap/duplex junction, whereas hEXO1 binding caused hypersensitivity very close to the 5'-end, correlating with the predominance of hFEN1 endonucleolytic activity versus hEXO1 exonucleolytic activity on 5'-flap substrates. These results show that FEN1 and EXO1 can eliminate structures formed by trinucleotide repeats in the course of replication, relying on endonucleolytic and exonucleolytic activities, respectively. These results also suggest that unresolved G4 DNA may prevent key steps in normal post-replicative DNA processing.
- Published
- 2010
- Full Text
- View/download PDF
41. Distinct activities of exonuclease 1 and flap endonuclease 1 at telomeric g4 DNA.
- Author
-
Vallur AC and Maizels N
- Subjects
- Base Sequence, DNA Primers, DNA Replication, Humans, Hydrolysis, Recombination, Genetic, Substrate Specificity, DNA metabolism, Exodeoxyribonucleases metabolism, Flap Endonucleases metabolism, Telomere
- Abstract
Background: Exonuclease 1 (EXO1) and Flap endonuclease 1 (FEN1) are members of the RAD2 family of structure-specific nucleases. Genetic analysis has identified roles for EXO1 and FEN1 in replication, recombination, DNA repair and maintenance of telomeres. Telomeres are composed of G-rich repeats that readily form G4 DNA. We recently showed that human EXO1 and FEN1 exhibit distinct activities on G4 DNA substrates representative of intermediates in immunoglobulin class switch recombination., Methodology/principal Findings: We have now compared activities of these enzymes on telomeric substrates bearing G4 DNA, identifying non-overlapping functions that provide mechanistic insight into the distinct telomeric phenotypes caused by their deficiencies. We show that hFEN1 but not hEXO1 cleaves substrates bearing telomeric G4 DNA 5'-flaps, consistent with the requirement for FEN1 in telomeric lagging strand replication. Both hEXO1 and hFEN1 are active on substrates bearing telomeric G4 DNA tails, resembling uncapped telomeres. Notably, hEXO1 but not hFEN1 is active on transcribed telomeric G-loops., Conclusion/significance: Our results suggest that EXO1 may act at transcription-induced telomeric structures to promote telomere recombination while FEN1 has a dominant role in lagging strand replication at telomeres. Both enzymes can create ssDNA at uncapped telomere ends thereby contributing to recombination.
- Published
- 2010
- Full Text
- View/download PDF
42. RAD51 paralogs promote homology-directed repair at diversifying immunoglobulin V regions.
- Author
-
Ordinario EC, Yabuki M, Handa P, Cummings WJ, and Maizels N
- Subjects
- Alleles, Animals, B-Lymphocytes metabolism, Cell Line, Cell Nucleus metabolism, Chickens, Cytidine Deaminase metabolism, DNA Damage, DNA-Binding Proteins metabolism, Gene Conversion, Green Fluorescent Proteins, Humans, Immunoglobulin Light Chains genetics, Recombinant Fusion Proteins metabolism, DNA Repair, Genetic Variation, Immunoglobulin Variable Region genetics, Rad51 Recombinase chemistry, Sequence Homology, Amino Acid
- Abstract
Background: Gene conversion depends upon the same factors that carry out more general process of homologous recombination, including homologous gene targeting and recombinational repair. Among these are the RAD51 paralogs, conserved factors related to the key recombination factor, RAD51. In chicken and other fowl, gene conversion (templated mutation) diversifies immunoglobulin variable region sequences. This allows gene conversion and recombinational repair to be studied using the chicken DT40 B cell line, which carries out constitutive gene conversion and provides a robust and physiological model for homology-directed repair in vertebrate cells., Results: We show that DT40 contains constitutive nuclear foci of the repair factors RAD51D and XRCC2, consistent with activated homologous recombination. Single-cell imaging of a DT40 derivative in which the rearranged and diversifying immunoglobulin lambdaR light chain gene is tagged with polymerized lactose operator, DT40 PolyLacO-lambdaR, showed that RAD51D and XRCC2 localize to the diversifying lambdaR gene. Colocalizations correlate both functionally and physically with active immunoglobulin gene conversion. Ectopic expression of either RAD51D or XRCC2 accelerated the clonal rate of gene conversion, and conversion tracts were significantly longer in RAD51D than XRCC2 transfectants., Conclusion: These results demonstrate direct functions of RAD51D and XRCC2 in immunoglobulin gene conversion, and also suggest that modulation of levels of repair factors may be a useful strategy to promote gene correction in other cell types.
- Published
- 2009
- Full Text
- View/download PDF
43. Temporal regulation of Ig gene diversification revealed by single-cell imaging.
- Author
-
Ordinario EC, Yabuki M, Larson RP, and Maizels N
- Subjects
- Animals, Bacterial Proteins biosynthesis, Bacterial Proteins genetics, Bacterial Proteins metabolism, Cell Line, Tumor, Cell Nucleus enzymology, Cell Nucleus genetics, Cell Nucleus immunology, Chickens, Clone Cells, Cytidine Deaminase biosynthesis, Cytidine Deaminase genetics, Cytidine Deaminase metabolism, G1 Phase genetics, G1 Phase immunology, Gene Rearrangement, B-Lymphocyte, Light Chain immunology, Luminescent Proteins biosynthesis, Luminescent Proteins genetics, Luminescent Proteins metabolism, Lymphoma enzymology, Lymphoma genetics, Lymphoma immunology, Time Factors, Antibody Diversity genetics, Cell Cycle genetics, Cell Cycle immunology, Genes, Immunoglobulin immunology
- Abstract
Rearranged Ig V regions undergo activation-induced cytidine deaminase (AID)-initiated diversification in sequence to produce either nontemplated or templated mutations, in the related pathways of somatic hypermutation and gene conversion. In chicken DT40 B cells, gene conversion normally predominates, producing mutations templated by adjacent pseudo-V regions, but impairment of gene conversion switches mutagenesis to a nontemplated pathway. We recently showed that the activator, E2A, functions in cis to promote diversification, and that G(1) phase of cell cycle is the critical window for E2A action. By single-cell imaging of stable AID-yellow fluorescent protein transfectants, we now demonstrate that AID-yellow fluorescent protein can stably localize to the nucleus in G(1) phase, but undergoes ubiquitin-dependent proteolysis later in cell cycle. By imaging of DT40 polymerized lactose operator-lambda(R) cells, in which polymerized lactose operator tags the rearranged lambda(R) gene, we show that both the repair polymerase Poleta and the multifunctional factor MRE11/RAD50/NBS1 localize to lambda(R), and that lambda(R)/Poleta colocalizations occur predominately in G(1) phase, when they reflect repair of AID-initiated damage. We find no evidence of induction of gamma-H2AX, the phosphorylated variant histone that is a marker of double-strand breaks, and Ig gene conversion may therefore proceed by a pathway involving templated repair at DNA nicks rather than double-strand breaks. These results lead to a model in which Ig gene conversion initiates and is completed or nearly completed in G(1) phase. AID deaminates ssDNA, and restriction of mutagenesis to G(1) phase would contribute to protecting the genome from off-target attack by AID when DNA replication occurs in S phase.
- Published
- 2009
- Full Text
- View/download PDF
44. Selection for the G4 DNA motif at the 5' end of human genes.
- Author
-
Eddy J and Maizels N
- Subjects
- DNA chemistry, Exons genetics, Humans, Regulatory Sequences, Nucleic Acid, 5' Untranslated Regions genetics, DNA analysis, G-Quadruplexes, Genes, Tumor Suppressor, Genome, Human, Proto-Oncogenes genetics
- Abstract
Formation of G4 DNA may occur in the course of replication and transcription, and contribute to genomic instability. We have quantitated abundance of G4 motifs and potential for G4 DNA formation of the nontemplate strand of 5' exons and introns of transcripts of human genes. We find that, for all human genes, G4 motifs are enriched in 5' regions of transcripts relative to downstream regions; and in 5' regulatory regions relative to coding regions. Notably, although tumor suppressor genes are depleted and proto-oncogenes enriched in G4 motifs, abundance of G4 motifs in the 5' regions of transcripts of genes in these categories does not differ. These results support the hypothesis that G4 motifs are under selection in the human genome. They further show that for tumor suppressor genes and proto-oncogenes, independent selection determines potential for G4 DNA formation of 5' regulatory regions of transcripts and downstream coding regions., ((c) 2009 Wiley-Liss, Inc.)
- Published
- 2009
- Full Text
- View/download PDF
45. Generation of a nicking enzyme that stimulates site-specific gene conversion from the I-AniI LAGLIDADG homing endonuclease.
- Author
-
McConnell Smith A, Takeuchi R, Pellenz S, Davis L, Maizels N, Monnat RJ Jr, and Stoddard BL
- Subjects
- Base Sequence, Catalytic Domain, DNA Cleavage, DNA Repair, Humans, Mutagenesis, Mutation, Substrate Specificity, Deoxyribonuclease I genetics, Endonucleases genetics, Protein Engineering methods, RNA-Directed DNA Polymerase genetics
- Abstract
Homing endonucleases stimulate gene conversion by generating site-specific DNA double-strand breaks that are repaired by homologous recombination. These enzymes are potentially valuable tools for targeted gene correction and genome engineering. We have engineered a variant of the I-AniI homing endonuclease that nicks its cognate target site. This variant contains a mutation of a basic residue essential for proton transfer and solvent activation in one active site. The cleavage mechanism, DNA-binding affinity, and substrate specificity profile of the nickase are similar to the wild-type enzyme. I-AniI nickase stimulates targeted gene correction in human cells, in cis and in trans, at approximately 1/4 the efficiency of the wild-type enzyme. The development of sequence-specific nicking enzymes like the I-AniI nickase will facilitate comparative analyses of DNA repair and mutagenesis induced by single- or double-strand breaks.
- Published
- 2009
- Full Text
- View/download PDF
46. E2A acts in cis in G1 phase of cell cycle to promote Ig gene diversification.
- Author
-
Yabuki M, Ordinario EC, Cummings WJ, Fujii MM, and Maizels N
- Subjects
- Animals, B-Lymphocytes immunology, B-Lymphocytes metabolism, Basic Helix-Loop-Helix Transcription Factors antagonists & inhibitors, Cell Line, Tumor, Chickens, Immunoglobulin lambda-Chains biosynthesis, Immunoglobulin lambda-Chains genetics, Inhibitor of Differentiation Proteins biosynthesis, Inhibitor of Differentiation Proteins genetics, Inhibitor of Differentiation Proteins physiology, Isopropyl Thiogalactoside analogs & derivatives, Isopropyl Thiogalactoside physiology, TCF Transcription Factors biosynthesis, TCF Transcription Factors genetics, Transcription Factor 7-Like 1 Protein, Antibody Diversity genetics, Basic Helix-Loop-Helix Transcription Factors physiology, E-Box Elements genetics, G1 Phase genetics, Gene Rearrangement, B-Lymphocyte, Light Chain, Genes, Immunoglobulin
- Abstract
Rearranged Ig genes undergo diversification in sequence and structure initiated by the DNA deaminase, activation-induced deaminase. Ig genes must be transcribed for diversification to occur, but whether there are additional requirements for cis activation has not been established. Here we show, by chromatin immunoprecipitation, that the regulatory factor E2A associates with the rearranged Ig lambda(R) gene in the chicken DT40 B cell line, which performs constitutive Ig gene diversification. By analysis of a DT40 derivative in which polymerized lactose operator tags the rearranged lambda(R) gene, we show that E2A must function in cis to promote diversification and that stimulation of diversification in cis depends on the E2A activation domains. By direct imaging, we show that lambda(R)/E2A colocalizations are most prominent in G(1). We further show that expression of the E2A antagonist Id1 prevents lambda(R)/E2A colocalizations in G(1) and impairs diversification but not transcription of lambda(R). Thus, E2A acts in cis to promote Ig gene diversification, and G(1) phase is the critical window for E2A action.
- Published
- 2009
- Full Text
- View/download PDF
47. Activities of human exonuclease 1 that promote cleavage of transcribed immunoglobulin switch regions.
- Author
-
Vallur AC and Maizels N
- Subjects
- Animals, DNA Repair Enzymes physiology, Exodeoxyribonucleases physiology, Humans, Hydrolysis, Immunoglobulin Class Switching genetics, Mice, Mice, Knockout, Transcription, Genetic, Cytidine Deaminase metabolism, DNA Repair Enzymes metabolism, Exodeoxyribonucleases metabolism, Immunoglobulin Switch Region genetics
- Abstract
Eukaryotic exonuclease 1 functions in replication, recombination, mismatch repair, telomere maintenance, immunoglobulin (Ig) gene class switch recombination, and somatic hypermutation. The enzyme has 5'-3' exonuclease, flap endonuclease, and weak RNaseH activity in vitro, but it has been difficult to reconcile these activities with its diverse biological functions. We report robust cleavage by human exonuclease 1 of transcribed G-rich DNA sequences with potential to form G loops and G4 DNA. Predicted Ig switch recombination intermediates are substrates for both exonucleolytic and 5' flap endonucleolytic cleavage. Excision is nick-dependent and structure-dependent. These results lead to a model for exonuclease 1 function in class switch recombination in which cleavage at activation-induced deaminase (AID)-initiated nicks produces gaps that become substrates for further attack by AID and subsequent repair.
- Published
- 2008
- Full Text
- View/download PDF
48. High-fidelity correction of genomic uracil by human mismatch repair activities.
- Author
-
Larson ED, Bednarski DW, and Maizels N
- Subjects
- Adenine, B-Lymphocytes metabolism, Cell Extracts, Cell Line, Cell Nucleus enzymology, DNA, Circular metabolism, DNA-Binding Proteins metabolism, Guanine, Humans, Mutation genetics, Nucleic Acid Heteroduplexes metabolism, Substrate Specificity, Uracil-DNA Glycosidase metabolism, DNA Mismatch Repair, Genome, Human genetics, Uracil metabolism
- Abstract
Background: Deamination of cytosine to produce uracil is a common and potentially mutagenic lesion in genomic DNA. U*G mismatches occur spontaneously throughout the genome, where they are repaired by factors associated with the base excision repair pathway. U*G mismatches are also the initiating lesion in immunoglobulin gene diversification, where they undergo mutagenic processing by redundant pathways, one dependent upon uracil excision and the other upon mismatch recognition by MutS alpha. While UNG is well known to initiate repair of uracil in DNA, the ability of MutS alpha to direct correction of this base has not been directly demonstrated., Results: Using a biochemical assay for mismatch repair, we show that MutS alpha can promote efficient and faithful repair of U*G mismatches, but does not repair U*A pairs in DNA. This contrasts with UNG, which readily excises U opposite either A or G. Repair of U*G by MutS alpha depends upon DNA polymerase delta (pol delta), ATP, and proliferating cell nuclear antigen (PCNA), all properties of canonical mismatch repair., Conclusion: These results show that faithful repair of U*G can be carried out by either the mismatch repair or base excision repair pathways. Thus, the redundant functions of these pathways in immunoglobulin gene diversification reflect their redundant functions in faithful repair. Faithful repair by either pathway is comparably efficient, suggesting that mismatch repair and base excision repair share the task of faithful repair of genomic uracil.
- Published
- 2008
- Full Text
- View/download PDF
49. Genomic stability: FANCJ-dependent G4 DNA repair.
- Author
-
Maizels N
- Subjects
- Animals, Caenorhabditis elegans genetics, Caenorhabditis elegans metabolism, Caenorhabditis elegans Proteins genetics, Caenorhabditis elegans Proteins metabolism, DNA chemistry, DNA Helicases genetics, DNA Helicases metabolism, DNA, Helminth chemistry, DNA, Helminth genetics, DNA, Helminth metabolism, Genes, Helminth, Humans, Models, Biological, Mutation, Nucleic Acid Conformation, Basic-Leucine Zipper Transcription Factors metabolism, DNA genetics, DNA metabolism, DNA Repair, Fanconi Anemia Complementation Group Proteins metabolism, Genomic Instability
- Abstract
G-rich regions have the potential to form G4 DNA upon replication, which can lead to genomic instability. FANCJ, a G4 DNA helicase, has been shown to be critical for the stability of regions that match the G4 signature motif by experiments analyzing its nematode homolog.
- Published
- 2008
- Full Text
- View/download PDF
50. Conserved elements with potential to form polymorphic G-quadruplex structures in the first intron of human genes.
- Author
-
Eddy J and Maizels N
- Subjects
- Base Sequence, Binding Sites, Chromosome Mapping, Conserved Sequence, CpG Islands, Genomics, Heterogeneous-Nuclear Ribonucleoproteins metabolism, Humans, Promoter Regions, Genetic, Regulatory Elements, Transcriptional, Sp1 Transcription Factor metabolism, Transcription Factors metabolism, Transcription Initiation Site, G-Quadruplexes, Introns, Regulatory Sequences, Nucleic Acid
- Abstract
To understand how potential for G-quadruplex formation might influence regulation of gene expression, we examined the 2 kb spanning the transcription start sites (TSS) of the 18 217 human RefSeq genes, distinguishing contributions of template and nontemplate strands. Regions both upstream and downstream of the TSS are G-rich, but the downstream region displays a clear bias toward G-richness on the nontemplate strand. Upstream of the TSS, much of the G-richness and potential for G-quadruplex formation derives from the presence of well-defined canonical regulatory motifs in duplex DNA, including CpG dinucleotides which are sites of regulatory methylation, and motifs recognized by the transcription factor SP1. This challenges the notion that quadruplex formation upstream of the TSS contributes to regulation of gene expression. Downstream of the TSS, G-richness is concentrated in the first intron, and on the nontemplate strand, where polymorphic sequence elements with potential to form G-quadruplex structures and which cannot be accounted for by known regulatory motifs are found in almost 3000 (16%) of the human RefSeq genes, and are conserved through frogs. These elements could in principle be recognized either as DNA or as RNA, providing structural targets for regulation at the level of transcription or RNA processing.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.