Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Mol Cell ; 83(20): 3622-3641, 2023 10 19.
Article in English | MEDLINE | ID: mdl-37863029

ABSTRACT

Around 3% of the genome consists of simple DNA repeats that are prone to forming alternative (non-B) DNA structures, such as hairpins, cruciforms, triplexes (H-DNA), four-stranded guanine quadruplexes (G4-DNA), and others, as well as composite RNA:DNA structures (e.g., R-loops, G-loops, and H-loops). These DNA structures are dynamic and favored by the unwinding of duplex DNA. For many years, the association of alternative DNA structures with genome function was limited by the lack of methods to detect them in vivo. Here, we review the recent advancements in the field and present state-of-the-art technologies and methods to study alternative DNA structures. We discuss the limitations of these methods as well as how they are beginning to provide insights into causal relationships between alternative DNA structures, genome function and stability, and human disease.


Subject(s)
DNA , G-Quadruplexes , Humans , DNA/genetics , DNA/chemistry , RNA/genetics , RNA/chemistry
2.
Mol Cell ; 82(19): 3538-3552.e5, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36075220

ABSTRACT

DNA becomes single stranded (ssDNA) during replication, transcription, and repair. Transiently formed ssDNA segments can adopt alternative conformations, including cruciforms, triplexes, and quadruplexes. To determine whether there are stable regions of ssDNA in the human genome, we utilized S1-END-seq to convert ssDNA regions to DNA double-strand breaks, which were then processed for high-throughput sequencing. This approach revealed two predominant non-B DNA structures: cruciform DNA formed by expanded (TA)n repeats that accumulate in microsatellite unstable human cancer cell lines and DNA triplexes (H-DNA) formed by homopurine/homopyrimidine mirror repeats common across a variety of cell lines. We show that H-DNA is enriched during replication, that its genomic location is highly conserved, and that H-DNA formed by (GAA)n repeats can be disrupted by treatment with a (GAA)n-binding polyamide. Finally, we show that triplex-forming repeats are hotspots for mutagenesis. Our results identify dynamic DNA secondary structures in vivo that contribute to elevated genome instability.


Subject(s)
DNA, Cruciform , Nylons , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Replication , Humans , Nucleic Acid Conformation
3.
Nucleic Acids Res ; 52(8): 4361-4374, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38381906

ABSTRACT

CANVAS is a recently characterized repeat expansion disease, most commonly caused by homozygous expansions of an intronic (A2G3)n repeat in the RFC1 gene. There are a multitude of repeat motifs found in the human population at this locus, some of which are pathogenic and others benign. In this study, we conducted structure-functional analyses of the pathogenic (A2G3)n and nonpathogenic (A4G)n repeats. We found that the pathogenic, but not the nonpathogenic, repeat presents a potent, orientation-dependent impediment to DNA polymerization in vitro. The pattern of the polymerization blockage is consistent with triplex or quadruplex formation in the presence of magnesium or potassium ions, respectively. Chemical probing of both repeats in vitro reveals triplex H-DNA formation by only the pathogenic repeat. Consistently, bioinformatic analysis of S1-END-seq data from human cell lines shows preferential H-DNA formation genome-wide by (A2G3)n motifs over (A4G)n motifs. Finally, the pathogenic, but not the nonpathogenic, repeat stalls replication fork progression in yeast and human cells. We hypothesize that the CANVAS-causing (A2G3)n repeat represents a challenge to genome stability by folding into alternative DNA structures that stall DNA replication.


Subject(s)
Cerebellar Ataxia , DNA Repeat Expansion , DNA Replication , Peripheral Nervous System Diseases , Vestibular Diseases , Humans , DNA/metabolism , DNA/chemistry , DNA/genetics , DNA Repeat Expansion/genetics , DNA Replication/genetics , Nucleic Acid Conformation , Replication Protein C/genetics , Replication Protein C/metabolism , Cerebellar Ataxia/genetics , Peripheral Nervous System Diseases/genetics , Vestibular Diseases/genetics
4.
PLoS Genet ; 19(1): e1010590, 2023 01.
Article in English | MEDLINE | ID: mdl-36701275

ABSTRACT

Although homologous recombination between transposable elements can drive genomic evolution in yeast by facilitating chromosomal rearrangements, the details of the underlying mechanisms are not fully clarified. In the genome of the yeast Saccharomyces cerevisiae, the most common class of transposon is the retrotransposon Ty1. Here, we explored how Cas9-induced double-strand breaks (DSBs) directed to Ty1 elements produce genomic alterations in this yeast species. Following Cas9 induction, we observed a significant elevation of chromosome rearrangements such as deletions, duplications and translocations. In addition, we found elevated rates of mitotic recombination, resulting in loss of heterozygosity. Using Southern analysis coupled with short- and long-read DNA sequencing, we revealed important features of recombination induced in retrotransposons. Almost all of the chromosomal rearrangements reflect the repair of DSBs at Ty1 elements by non-allelic homologous recombination; clustered Ty elements were hotspots for chromosome rearrangements. In contrast, a large proportion (about three-fourths) of the allelic mitotic recombination events have breakpoints in unique sequences. Our analysis suggests that some of the latter events reflect extensive processing of the broken ends produced in the Ty element that extend into unique sequences resulting in break-induced replication. Finally, we found that haploid and diploid strain have different preferences for the pathways used to repair double-stranded DNA breaks. Our findings demonstrate the importance of DNA lesions in retrotransposons in driving genome evolution.


Subject(s)
CRISPR-Cas Systems , Saccharomyces cerevisiae , Humans , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , CRISPR-Cas Systems/genetics , DNA Breaks, Double-Stranded , Retroelements/genetics , Chromosome Aberrations , Homologous Recombination/genetics
5.
Nucleic Acids Res ; 51(16): 8532-8549, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37216608

ABSTRACT

Friedreich's ataxia (FRDA) is caused by expansions of GAA•TTC repeats in the first intron of the human FXN gene that occur during both intergenerational transmissions and in somatic cells. Here we describe an experimental system to analyze large-scale repeat expansions in cultured human cells. It employs a shuttle plasmid that can replicate from the SV40 origin in human cells or be stably maintained in S. cerevisiae utilizing ARS4-CEN6. It also contains a selectable cassette allowing us to detect repeat expansions that accumulated in human cells upon plasmid transformation into yeast. We indeed observed massive expansions of GAA•TTC repeats, making it the first genetically tractable experimental system to study large-scale repeat expansions in human cells. Further, GAA•TTC repeats stall replication fork progression, while the frequency of repeat expansions appears to depend on proteins implicated in replication fork stalling, reversal, and restart. Locked nucleic acid (LNA)-DNA mixmer oligonucleotides and peptide nucleic acid (PNA) oligomers, which interfere with triplex formation at GAA•TTC repeats in vitro, prevented the expansion of these repeats in human cells. We hypothesize, therefore, that triplex formation by GAA•TTC repeats stall replication fork progression, ultimately leading to repeat expansions during replication fork restart.


Subject(s)
Friedreich Ataxia , Oligonucleotides , Peptide Nucleic Acids , Trinucleotide Repeat Expansion , Humans , DNA , DNA Replication/drug effects , Friedreich Ataxia/genetics , Iron-Binding Proteins/genetics , Oligonucleotides/pharmacology , Peptide Nucleic Acids/pharmacology , Saccharomyces cerevisiae/genetics
6.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Article in English | MEDLINE | ID: mdl-33495349

ABSTRACT

Nearly 50 hereditary diseases result from the inheritance of abnormally long repetitive DNA microsatellites. While it was originally believed that the size of inherited repeats is the key factor in disease development, it has become clear that somatic instability of these repeats throughout an individual's lifetime strongly contributes to disease onset and progression. Importantly, somatic instability is commonly observed in terminally differentiated, postmitotic cells, such as neurons. To unravel the mechanisms of repeat instability in nondividing cells, we created an experimental system to analyze the mutability of Friedreich's ataxia (GAA)n repeats during chronological aging of quiescent Saccharomyces cerevisiae Unexpectedly, we found that the predominant repeat-mediated mutation in nondividing cells is large-scale deletions encompassing parts, or the entirety, of the repeat and adjacent regions. These deletions are caused by breakage at the repeat mediated by mismatch repair (MMR) complexes MutSß and MutLα and DNA endonuclease Rad1, followed by end-resection by Exo1 and repair of the resulting double-strand breaks (DSBs) via nonhomologous end joining. We also observed repeat-mediated gene conversions as a result of DSB repair via ectopic homologous recombination during chronological aging. Repeat expansions accrue during chronological aging as well-particularly in the absence of MMR-induced DSBs. These expansions depend on the processivity of DNA polymerase δ while being counteracted by Exo1 and MutSß, implicating nick repair. Altogether, these findings show that the mechanisms and types of (GAA)n repeat instability differ dramatically between dividing and nondividing cells, suggesting that distinct repeat-mediated mutations in terminally differentiated somatic cells might influence Friedreich's ataxia pathogenesis.


Subject(s)
Aging/genetics , DNA Replication/genetics , Friedreich Ataxia/genetics , Genomic Instability/genetics , Trinucleotide Repeat Expansion/genetics , DNA/biosynthesis , DNA Breaks, Double-Stranded , DNA End-Joining Repair/genetics , DNA Polymerase III/metabolism , DNA Repair Enzymes/metabolism , Exodeoxyribonucleases/metabolism , Gene Conversion , Humans , Models, Biological , Mutation/genetics , Protein Subunits/metabolism , Recombination, Genetic/genetics , Saccharomyces cerevisiae/genetics
7.
8.
Proc Natl Acad Sci U S A ; 117(3): 1628-1637, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31911468

ABSTRACT

Friedreich's ataxia (FRDA) is a human hereditary disease caused by the presence of expanded (GAA)n repeats in the first intron of the FXN gene [V. Campuzano et al., Science 271, 1423-1427 (1996)]. In somatic tissues of FRDA patients, (GAA)n repeat tracts are highly unstable, with contractions more common than expansions [R. Sharma et al., Hum. Mol. Genet. 11, 2175-2187 (2002)]. Here we describe an experimental system to characterize GAA repeat contractions in yeast and to conduct a genetic analysis of this process. We found that large-scale contraction is a one-step process, resulting in a median loss of ∼60 triplet repeats. Our genetic analysis revealed that contractions occur during DNA replication, rather than by various DNA repair pathways. Repeats contract in the course of lagging-strand synthesis: The processivity subunit of DNA polymerase δ, Pol32, and the catalytic domain of Rev1, a translesion polymerase, act together in the same pathway to counteract contractions. Accumulation of single-stranded DNA (ssDNA) in the lagging-strand template greatly increases the probability that (GAA)n repeats contract, which in turn promotes repeat instability in rfa1, rad27, and dna2 mutants. Finally, by comparing contraction rates for homopurine-homopyrimidine repeats differing in their mirror symmetry, we found that contractions depend on a repeat's triplex-forming ability. We propose that accumulation of ssDNA in the lagging-strand template fosters the formation of a triplex between the nascent and fold-back template strands of the repeat. Occasional jumps of DNA polymerase through this triplex hurdle, result in repeat contractions in the nascent lagging strand.


Subject(s)
DNA Replication , Friedreich Ataxia/genetics , Saccharomyces cerevisiae/genetics , Trinucleotide Repeats , DNA Polymerase III , DNA Repair , DNA, Single-Stranded , DNA-Directed DNA Polymerase , Flap Endonucleases , Humans , Mutation , Nucleotidyltransferases/genetics , Replication Protein A , Saccharomyces cerevisiae Proteins
9.
Mol Cell ; 53(1): 1-3, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24411078

ABSTRACT

Expansions of DNA repeats cause hereditary disorders in humans. Gerhardt et al. (2014) argue that a developmental switch in the direction of DNA replication through the (CGG)n repeat predisposes it to expansions during intergenerational transmissions leading to fragile X syndrome.


Subject(s)
DNA Replication , Embryonic Stem Cells/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/embryology , Genetic Loci , Trinucleotide Repeats , Humans
10.
J Biol Chem ; 295(13): 4134-4170, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32060097

ABSTRACT

Expansions of simple tandem repeats are responsible for almost 50 human diseases, the majority of which are severe, degenerative, and not currently treatable or preventable. In this review, we first describe the molecular mechanisms of repeat-induced toxicity, which is the connecting link between repeat expansions and pathology. We then survey alternative DNA structures that are formed by expandable repeats and review the evidence that formation of these structures is at the core of repeat instability. Next, we describe the consequences of the presence of long structure-forming repeats at the molecular level: somatic and intergenerational instability, fragility, and repeat-induced mutagenesis. We discuss the reasons for gender bias in intergenerational repeat instability and the tissue specificity of somatic repeat instability. We also review the known pathways in which DNA replication, transcription, DNA repair, and chromatin state interact and thereby promote repeat instability. We then discuss possible reasons for the persistence of disease-causing DNA repeats in the genome. We describe evidence suggesting that these repeats are a payoff for the advantages of having abundant simple-sequence repeats for eukaryotic genome function and evolvability. Finally, we discuss two unresolved fundamental questions: (i) why does repeat behavior differ between model systems and human pedigrees, and (ii) can we use current knowledge on repeat instability mechanisms to cure repeat expansion diseases?


Subject(s)
DNA Repeat Expansion/genetics , Genome, Human/genetics , Microsatellite Repeats/genetics , Nucleic Acid Conformation , Chromatin/genetics , DNA Repair/genetics , DNA Replication/genetics , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Genomic Instability/genetics , Humans
11.
Trends Genet ; 34(6): 448-465, 2018 06.
Article in English | MEDLINE | ID: mdl-29567336

ABSTRACT

Over 30 hereditary diseases are caused by the expansion of microsatellite repeats. The length of the expandable repeat is the main hereditary determinant of these disorders. They are also affected by numerous genomic variants that are either nearby (cis) or physically separated from (trans) the repetitive locus, which we review here. These genetic variants have largely been elucidated in model systems using gene knockouts, while a few have been directly observed as single-nucleotide polymorphisms (SNPs) in patients. There is a notable disconnect between these two bodies of knowledge: knockouts poorly approximate the SNP-level variation in human populations that gives rise to medically relevant cis- and trans-modifiers, while the rarity of these diseases limits the statistical power of SNP-based analysis in humans. We propose that high-throughput SNP-based screening in model systems could become a useful approach to quickly identify and characterize modifiers of clinical relevance for patients.


Subject(s)
DNA Repeat Expansion/genetics , Gene Expression Regulation/genetics , Microsatellite Repeats/genetics , Gene Knockout Techniques , Genome, Human/genetics , Genomics , Humans , Polymorphism, Single Nucleotide/genetics
12.
Nucleic Acids Res ; 47(2): 794-805, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30476303

ABSTRACT

Fork stabilization at DNA impediments is key to maintaining replication fork integrity and preventing chromosome breaks. Mrc1 and Tof1 are two known stabilizers that travel with the replication fork. In addition to a structural role, Mrc1 has a DNA damage checkpoint function. Using a yeast model system, we analyzed the role of Mrc1 and Tof1 at expanded CAG repeats of medium and long lengths, which are known to stall replication forks and cause trinucleotide expansion diseases such as Huntington's disease and myotonic dystrophy. We demonstrate that the fork stabilizer but not the checkpoint activation function of Mrc1 is key for preventing DNA breakage and death of cells containing expanded CAG tracts. In contrast, both Mrc1 functions are important in preventing repeat length instability. Mrc1 has a general fork protector role that is evident at forks traversing both repetitive and non-repetitive DNA, though it becomes crucial at long CAG repeat lengths. In contrast, the role of Tof1 in preventing fork breakage is specific to long CAG tracts of 85 or more repeats. Our results indicate that long CAG repeats have a particular need for Tof1 and highlight the importance of fork stabilizers in maintaining fork integrity during replication of structure-forming repeats.


Subject(s)
Cell Cycle Proteins/physiology , Chromosome Fragility , DNA-Binding Proteins/physiology , Saccharomyces cerevisiae Proteins/physiology , Trinucleotide Repeats , Cell Cycle Proteins/genetics , DNA Replication , DNA-Binding Proteins/genetics , Gene Deletion , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics
13.
Genome Res ; 27(12): 2072-2082, 2017 12.
Article in English | MEDLINE | ID: mdl-29113982

ABSTRACT

Improper DNA double-strand break (DSB) repair results in complex genomic rearrangements (CGRs) in many cancers and various congenital disorders in humans. Trinucleotide repeat sequences, such as (GAA)n repeats in Friedreich's ataxia, (CTG)n repeats in myotonic dystrophy, and (CGG)n repeats in fragile X syndrome, are also subject to double-strand breaks within the repetitive tract followed by DNA repair. Mapping the outcomes of CGRs is important for understanding their causes and potential phenotypic effects. However, high-resolution mapping of CGRs has traditionally been a laborious and highly skilled process. Recent advances in long-read DNA sequencing technologies, specifically Nanopore sequencing, have made possible the rapid identification of CGRs with single base pair resolution. Here, we have used whole-genome Nanopore sequencing to characterize several CGRs that originated from naturally occurring DSBs at (GAA)n microsatellites in Saccharomyces cerevisiae These data gave us important insights into the mechanisms of DSB repair leading to CGRs.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA, Fungal , Gene Rearrangement , Nanopores , Saccharomyces cerevisiae/genetics , Sequence Analysis, DNA/methods , Chromosomes, Fungal , Gene Duplication , Genome, Fungal , Retroelements , Trinucleotide Repeats
14.
Mol Cell ; 48(2): 254-65, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22959270

ABSTRACT

Triplex structure-forming GAA/TTC repeats pose a dual threat to the eukaryotic genome integrity. Their potential to expand can lead to gene inactivation, the cause of Friedreich's ataxia disease in humans. In model systems, long GAA/TTC tracts also act as chromosomal fragile sites that can trigger gross chromosomal rearrangements. The mechanisms that regulate the metabolism of GAA/TTC repeats are poorly understood. We have developed an experimental system in the yeast Saccharomyces cerevisiae that allows us to systematically identify genes crucial for maintaining the repeat stability. Two major groups of mutants defective in DNA replication or transcription initiation are found to be prone to fragility and large-scale expansions. We demonstrate that problems imposed by the repeats during DNA replication in actively dividing cells and during transcription initiation in nondividing cells can culminate in genome instability. We propose that similar mechanisms can mediate detrimental metabolism of GAA/TTC tracts in human cells.


Subject(s)
Chromosome Fragility/genetics , Friedreich Ataxia/genetics , Saccharomyces cerevisiae/genetics , Trinucleotide Repeats/genetics , DNA Replication , Genome, Fungal , Genome, Human , Genomic Instability , Humans , Microsatellite Repeats , Mutation , Nucleic Acid Conformation
15.
Nucleic Acids Res ; 46(7): 3487-3497, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29447396

ABSTRACT

Expansion of simple DNA repeats is responsible for numerous hereditary diseases in humans. The role of DNA replication, repair and transcription in the expansion process has been well documented. Here we analyzed, in a yeast experimental system, the role of RNA-DNA hybrids in genetic instability of long (GAA)n repeats, which cause Friedreich's ataxia. Knocking out both yeast RNase H enzymes, which counteract the formation of RNA-DNA hybrids, increased (GAA)n repeat expansion and contraction rates when the repetitive sequence was transcribed. Unexpectedly, we observed a similar increase in repeat instability in RNase H-deficient cells when we either changed the direction of transcription-replication collisions, or flipped the repeat sequence such that the (UUC)n run occurred in the transcript. The increase in repeat expansions in RNase H-deficient strains was dependent on Rad52 and Pol32 proteins, suggesting that break-induced replication (BIR) is responsible for this effect. We conclude that expansions of (GAA)n repeats are induced by the formation of RNA-DNA hybrids that trigger BIR. Since this stimulation is independent of which strand of the repeat (homopurine or homopyrimidine) is in the RNA transcript, we hypothesize that triplex H-DNA structures stabilized by an RNA-DNA hybrid (H-loops), rather than conventional R-loops, could be responsible.


Subject(s)
DNA-Directed DNA Polymerase/genetics , DNA/genetics , RNA/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Ribonuclease H/genetics , Saccharomyces cerevisiae Proteins/genetics , DNA Repair/genetics , DNA Replication/genetics , Friedreich Ataxia/genetics , Genomic Instability/genetics , Humans , Nucleic Acid Conformation , Recombination, Genetic , Saccharomyces cerevisiae/genetics , Trinucleotide Repeat Expansion/genetics
16.
Bioessays ; 39(9)2017 09.
Article in English | MEDLINE | ID: mdl-28703879

ABSTRACT

In this review, we discuss how two evolutionarily conserved pathways at the interface of DNA replication and repair, template switching and break-induced replication, lead to the deleterious large-scale expansion of trinucleotide DNA repeats that cause numerous hereditary diseases. We highlight that these pathways, which originated in prokaryotes, may be subsequently hijacked to maintain long DNA microsatellites in eukaryotes. We suggest that the negative mutagenic outcomes of these pathways, exemplified by repeat expansion diseases, are likely outweighed by their positive role in maintaining functional repetitive regions of the genome such as telomeres and centromeres.


Subject(s)
DNA/genetics , Eukaryota/genetics , Trinucleotide Repeats/genetics , Animals , DNA Repair/genetics , DNA Replication/genetics , Humans , Microsatellite Repeats/genetics , Prokaryotic Cells/physiology , Telomere/genetics
17.
Mol Cell ; 35(1): 82-92, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19595718

ABSTRACT

Large-scale expansions of DNA repeats are implicated in numerous hereditary disorders in humans. We describe a yeast experimental system to analyze large-scale expansions of triplet GAA repeats responsible for the human disease Friedreich's ataxia. When GAA repeats were placed into an intron of the chimeric URA3 gene, their expansions caused gene inactivation, which was detected on the selective media. We found that the rates of expansions of GAA repeats increased exponentially with their lengths. These rates were only mildly dependent on the repeat's orientation within the replicon, whereas the repeat-mediated replication fork stalling was exquisitely orientation dependent. Expansion rates were significantly elevated upon inactivation of the replication fork stabilizers, Tof1 and Csm3, but decreased in the knockouts of postreplication DNA repair proteins, Rad6 and Rad5, and the DNA helicase Sgs1. We propose a model for large-scale repeat expansions based on template switching during replication fork progression through repetitive DNA.


Subject(s)
Friedreich Ataxia/genetics , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeats/genetics , Yeasts/genetics , DNA Replication/genetics , Gene Expression Regulation, Fungal , Humans , Introns/genetics , Iron-Binding Proteins/genetics , Plasmids/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Frataxin
18.
Nucleic Acids Res ; 43(14): 6994-7004, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26101261

ABSTRACT

DNA sequences that can form unusual secondary structures are implicated in regulating gene expression and causing genomic instability. H-palindromes are an important class of such DNA sequences that can form an intramolecular triplex structure, H-DNA. Within an H-palindrome, the H-DNA and canonical B-DNA are in a dynamic equilibrium that shifts toward H-DNA with increased negative supercoiling. The interplay between H- and B-DNA and the fact that the process of transcription affects supercoiling makes it difficult to elucidate the effects of H-DNA upon transcription. We constructed a stable structural analog of H-DNA that cannot flip into B-DNA, and studied the effects of this structure on transcription by T7 RNA polymerase in vitro. We found multiple transcription blockage sites adjacent to and within sequences engaged in this triplex structure. Triplex-mediated transcription blockage varied significantly with changes in ambient conditions: it was exacerbated in the presence of Mn(2+) or by increased concentrations of K(+) and Li(+). Analysis of the detailed pattern of the blockage suggests that RNA polymerase is sterically hindered by H-DNA and has difficulties in unwinding triplex DNA. The implications of these findings for the biological roles of triple-stranded DNA structures are discussed.


Subject(s)
DNA/chemistry , Transcription, Genetic , Cations, Divalent , Cations, Monovalent , DNA-Directed RNA Polymerases/metabolism , Metals/chemistry , Nucleic Acid Conformation , Viral Proteins/metabolism
19.
Proc Natl Acad Sci U S A ; 110(49): 19866-71, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24191060

ABSTRACT

Interstitial telomeric sequences (ITSs) are present in many eukaryotic genomes and are linked to genome instabilities and disease in humans. The mechanisms responsible for ITS-mediated genome instability are not understood in molecular detail. Here, we use a model Saccharomyces cerevisiae system to characterize genome instability mediated by yeast telomeric (Ytel) repeats embedded within an intron of a reporter gene inside a yeast chromosome. We observed a very high rate of small insertions and deletions within the repeats. We also found frequent gross chromosome rearrangements, including deletions, duplications, inversions, translocations, and formation of acentric minichromosomes. The inversions are a unique class of chromosome rearrangement involving an interaction between the ITS and the true telomere of the chromosome. Because we previously found that Ytel repeats cause strong replication fork stalling, we suggest that formation of double-stranded DNA breaks within the Ytel sequences might be responsible for these gross chromosome rearrangements.


Subject(s)
Chromosome Aberrations , Chromosome Fragile Sites/genetics , Genomic Instability/genetics , Saccharomyces cerevisiae/genetics , Telomere/genetics , Blotting, Southern , DNA Breaks, Double-Stranded , Genes, Reporter/genetics , Microarray Analysis , Polymerase Chain Reaction
20.
Nucleic Acids Res ; 41(3): 1817-28, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23275544

ABSTRACT

The ability of DNA to adopt non-canonical structures can affect transcription and has broad implications for genome functioning. We have recently reported that guanine-rich (G-rich) homopurine-homopyrimidine sequences cause significant blockage of transcription in vitro in a strictly orientation-dependent manner: when the G-rich strand serves as the non-template strand [Belotserkovskii et al. (2010) Mechanisms and implications of transcription blockage by guanine-rich DNA sequences., Proc. Natl Acad. Sci. USA, 107, 12816-12821]. We have now systematically studied the effect of the sequence composition and single-stranded breaks on this blockage. Although substitution of guanine by any other base reduced the blockage, cytosine and thymine reduced the blockage more significantly than adenine substitutions, affirming the importance of both G-richness and the homopurine-homopyrimidine character of the sequence for this effect. A single-strand break in the non-template strand adjacent to the G-rich stretch dramatically increased the blockage. Breaks in the non-template strand result in much weaker blockage signals extending downstream from the break even in the absence of the G-rich stretch. Our combined data support the notion that transcription blockage at homopurine-homopyrimidine sequences is caused by R-loop formation.


Subject(s)
DNA Breaks, Single-Stranded , DNA/chemistry , Transcription, Genetic , Base Composition , Base Sequence , DNA-Directed RNA Polymerases/metabolism , Guanine/analysis , Nucleic Acid Conformation , Nucleotides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL