Vous êtes sur la page 1sur 13

rEVIEW

REVIEW

RNA Biology 8:3, 427-439; May/June 2011; 2011 Landes Bioscience

Potential in vivo roles of nucleic acid triple-helices


Fabian A. Buske, John S. Mattick* and Timothy L. Bailey*
Institute for Molecular Bioscience; The University of Queensland; Brisbane, QLD Australia

Key words: triple-helix, RNA-DNA interaction, triplex-forming oligonucleotide, sequence-specific, transcriptional regulation Abbreviations: ncRNA, non-protein-coding RNA; TTS, triplex target site; TFO, triplex-forming oligonucleotide; TF, transcription factor
The ability of double-stranded DNA to form a triple-helical structure by hydrogen bonding with a third strand is well established, but the biological functions of these structures remain largely unknown. There is considerable albeit circumstantial evidence for the existence of nucleic triplexes in vivo and their potential participation in a variety of biological processes including chromatin organization, DNA repair, transcriptional regulation and RNA processing has been investigated in a number of studies to date. There is also a range of possible mechanisms to regulate triplex formation through differential expression of triplex-forming RNAs, alteration of chromatin accessibility, sequence unwinding and nucleotide modifications. With the advent of next generation sequencing technology combined with targeted approaches to isolate triplexes, it is now possible to survey triplex formation with respect to their genomic context, abundance and dynamical changes during differentiation and development, which may open up new vistas in understanding genome biology and gene regulation.

Here, we critically review the body of evidence that suggests endogenous triple-helices form in vivo and implicates them in diverse biological processes. In the nal section, we suggest how modern sequencing technology can be coupled with computational approaches to unravel the prevalence and function of triplex structures in living cells. Triplex Formation and Pairing Rules Triplex-helices are formed by sequence-specic binding rules that are distinct although conceptually akin to the familiar WatsonCrick base-pairing rules. The triplex-formation involves a doublestranded nucleic acid such as duplex DNA and a single-stranded nucleic acid such as RNA. The triplex-forming oligonucleotide (TFO) of the single-stranded nucleic acid binds in the major groove of the targeted duplex through sequence-specic recognition of a polypurinepolypyrimidine sequence. Variations that bind in the minor groove have been reported11 but insights into these three-stranded variants are very limited. We therefore focus on major groove triple-helical structures unless stated otherwise. TFOs that bind in the major groove of the duplex form Hoogsteen hydrogen bonds with the purine-rich strand. Two congurations are possible: (1) the Hoogsteen conguration promotes binding of the third strand in a parallel orientation to the polypurine strand of the duplex, while (2) anti-parallel orientation of the TFO is established by reverse Hoogsteen bonds (Fig. 1). The stability of triple-helices depends heavily on the availability of hydrogen acceptor and donor groups from the third base, as well as its steric features. There are basically three motifs for stable triple-helix formation, all of which permit two stabilizing hydrogen bonds between the base in the third strand and the purine in the duplex. These motifs are referred to by the bases of the third strand that participate in the formation: [T,C] (pyrimidine motif), [G,A] (purine motif) and [G,T] (see Appendix). Of course, T refers to uracil when the third strand is made of RNA. In principle, there are eight possible ways to form a triplehelix when each of the three strands is either made of RNA or DNA. Thermodynamic in vitro studies have shown that the RNA-DNADNA and DNA-DNADNA triple-helical formations are most stable (where - refers to Hoogsteen/ reverse Hoogsteen and refers to Watson-Crick binding),12-16 although the nucleic acid backbones were found to have a profound effect on the formation efciency for different triplex

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .
Introduction

Nucleic acid triple-stranded structures, also called triplexes, are complexes of three oligonucleotide strands made of either RNA or DNA.1-3 Over the last decades triplexes have been implicated in a range of cellular functions such as transcriptional regulation, post-transcriptional RNA processing, modication of chromatin and DNA repair. Evidence underpinning these implications is, however, primarily based on in vitro experiments leaving the in vivo existence and function of triplexes a mystery yet to be revealed. Important pieces to the puzzle have been added by recent work on new methods for triplex detection,4 the discovery of new triplex-unwinding helicases,5 as well as the increasingly apparent biological roles played by non-coding RNAs6 that suggest the involvement of triple-helices.7-10 With the advent of high-throughput sequencing of RNA and DNA we now have the technology to map the genomic locations of RNA-DNA triplexes and to begin unraveling their functions.
*Correspondence to: John S. Mattick and Timothy L. Bailey; Email: j.mattick@imb.uq.edu.au and t.bailey@imb.uq.edu.au Submitted: 12/21/10; Revised: 01/26/11; Accepted: 01/28/11 DOI: 10.4161/rna.8.3.14999

www.landesbioscience.com

RNA Biology

427

Figure 1. Parallel and anti-parallel oriented triple-helix motifs. There are three basic triple-helix motifs: [T,C], [G,A] and [G,T]. The [G,T] motif can bind in both orientations to the duplex while [T,C] binds parallel and [G,A] binds anti-parallel to the purine tract of the duplex. The inset shows a cartoon of a triple-helical structure derived from a NMR-structure contained in the protein database (PDB-id:1BWG).

motifs, and RNA may not be able to participate in reverse Hoogsteen structures (reviewed in refs. 17 and 18). In addition, triplexes are distinguished according to whether the third strand is tethered to the duplex (intramolecular triplex) or an independent molecule (intermolecular triplex). Thus, triplex formation represents an additional mechanism for the cell to target nucleic acid sequences besides duplex hybridization and sequence-specic protein binding. Importantly, it permits the single-stranded nucleic acid (presumably mainly RNA) to bind the targeted duplex structure (RNA or DNA) without requiring it to unwind rst. This unique ability has considerable biotechnological potential and has been extensively studied for use in such applications as modulation of transcription19-23 and site-directed recombination as well as mutagen delivery.24-26 Evidence for the Existence of Triple-Helical Structures in vivo

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .

Triplex-specic antibodies and dyes bind nuclear triple-helical structures. The existence of triplex structures in the nucleus of insects, nematodes and mammals has been suggested by immunofluorescence using triple-helix specific monoclonal antibodies.27-34 The antibodies recognize distinct triplex epitopes as indicated by different staining patterns on polytene chromosomes of Drosophila melanogaster.30,34 Among the most compelling evidence for the existence of triplexes in vivo is immunodetection by triple-helix specic antibodies in human cell nuclei,31 which detected an abundance of triplex signatures (Fig. 2). Although the antibodies used in all but one of the above studies were prepared against DNA-DNADNA triplexes,27,28 it has subsequently been shown that these antibodies have an even stronger affinity to RNA-DNADNA triplexes.9,29,35 The signals observed in vivo may hence largely reflect RNA-DNADNA triplexes. Complementary evidence has recently been put forward using Thiazole Orange (TO), a dye that preferentially detects triplexes under in vivo conditions. TO treated U2OS cells reveal distinct staining patterns in cell nuclei, which are almost opposite to that of the duplex DNA binding dye DAPI,4 but may merely reflect

non-specic staining of the nucleolus by TO. However, TO staining signatures also extend to the cytoplasm. A preliminary study showed no evidence of the dye interacting with proteins promoting the idea that triplexes may also occur in the cytoplasm possible in the form of RNA structural motifs.4 Additional investigations into the binding specicity of these indicators are required to resolve all doubts concerning the origin of in vivo staining patterns from non-triplex-specic binding. Proteins specically recognize triple-helical structures. The existence of endogenous proteins that recognize and act specically on triplexes further supports their existence in the cell. Table 1 lists proteins that have been observed to bind to triplexes in vitro or that have a functional role specic to triplexes. Included among these are helicases of the RecQ family, which actively unwind triplexes in a 3'5' direction.5,36-39 In the case of SV40 large T-antigen helicase, as well as of DHX9 helicase a flanking 3' end on the triplex is required. Otherwise, the triplex inhibits even the unwinding of the duplex by the helicase, which has implications for DNA replication.40 In contrast, the triplex-melting

Figure 2. Immunofluorescent staining of cell nuclei using triplex-specific monoclonal antibodies. (Figure reproduced from ref. 31 with kind permission from the Springer Science + Business Media).

428

RNA Biology Volume 8 Issue 3

Table 1. Proteins detecting or binding nucleic triple-helices Protein (origin) Comments The GAGA factor (GAF) in Drosophila appears to perform different functional roles in distinct genomic locations (reviewed in ref. 196). In fact, its functional role seems to be subject to the structure of the targeted heterochromatin. The target motifs of GAF, (GA)n repeats, exist in several promoters and are able form triple-helical structures. Interestingly, GAF shows affinity to bind triple-helical structures in vitro with a similar specificity and affinity as duplex DNA. Thus triplex formation may account for the different functional roles GAF can adopt, which includes gene regulation but also alteration of chromatin structure.197,198 Stm1p preferentially associates with ribosomal RNA but can also interact with DNA, especially subtelomeric sequences199 and is thought to be involved in mitosis. Its first 113 amino acids comprise the minimal domain that is capable of binding purine triplexes but unable to bind duplex DNA. 200 CDP1p thought to be involved in chromosome segregation and histone displacement.201 Tn7 detect triplex DNA, which leads to specific insertion of the transposon adjacent to both intra- and intermolecular pyrimidine motif triplexes Nuclear localization Evidence Affinity to Ref.

GAGA factor (fruitfly)

Yes

EMSA analysis of interaction between triplex probe and protein.

pyrimidine-motif triplexes forming at (GA)n repeats

47

Stm1p (yeast)

Not reported

EMSA of yeast whole-cell extract using purine motif probe

purine-motif triplexes

48

CDP1p (yeast)

Tn7-encoded protein (Bacterial transposon)

IF proteins: vimentin, GFAB, desmin (mouse)

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .
Yes 49 Transposon Chemical footprinting intramolecular triplexes at mirror repeat sequences 202 Vimentin, GFAB and desmin, recognize and bind to repetitive DNA that preferably adopts intramolecular triplex H-DNA structure. Vimentin, which is localized in the cytosol as well as in the nuclear matrix,113,203 is possibly identical to a protein with a molecular mass of ~55 kDa found by earlier studies. 204,205 Loricrin is a major keratinocyte cell envelope protein. DNA-binding protein initially found by SouthWestern screening of a human keratinocyte cDNA expression library.45 Orc4 subunit 4 prefers triplex DNA to duplex or single stranded DNA suggesting that triplestranded structures are important for origin organization and activity during DNA replication. No binding to the corresponding single stranded DNA (TFO) or the duplex DNA Not reported Vector plasmid constructs containing IF-purified DNA fragments form triplexes as shown in band shift assays and prevention of nucleolytic cleavage and chemical modification. intramolecular triplexes at mirror repeats (GA)n 44 Yes South-Western screening of a human keratinocyte cDNA expression library using artificial triplex probe. EMSA analysis purine-motif triplexes and pyrimidine-motif triplexes (weaker) pyrimidine-motif triplexes 45 206

Southwestern library screening with subsequent EMSA analysis

purine-motif triplexes

Alternative loricrin reading frame (human) MBP-LOR3ARF (human)

Orc4 subunit 4 (human)

Yes

EMSA analysis using purified HsOrc4

(T-AT)n triplets

46

55 kDa (human)

Yes

Affinity purification from HeLa nuclear extracts using artificial triplexes Affinity purification from HeLa nuclear extracts using artificial triplexes (GT-motif) with subsequent EMSA and Southwestern blotting.

(T-AT)n triplets, pyrimidine and (weaker) purine triplexes purine-motif triplexes

204, 205

100 kDa, 60 kDa, 15 kDa (human)

No recognition of the corresponding duplex DNA or pyrimidine triplex by the proteins.

Yes

207

www.landesbioscience.com

RNA Biology

429

Table 1 (continued). Proteins detecting or binding nucleic triple-helices hnRNP (K, L, A2/B1, E1, I) (human) Detected proteins show affinity to triple-helical structures but not duplex or single-stranded DNA. Possibly identical to the proteins found by Musso et al. 207 The zinc-dependent protein present in nuclear extracts from different cell types was found to bind to DNA sequence prone to adopt triplehelical structure and to recognise structure-specific rather than sequence-specific features. HMG box proteins promote and stabilize triplex formation as shown by time course PAGE, though an unusual purine-motif triplex was suggested. In fact, the small tetra peptide PKRW containing the conserved sequences of the HMGB1 DNA-binding domain were found to be able to recognize triple-helical structures. DHX9 and SV40 large antigen and helicase are two specimen that can melt triplex in a 3'-5' polarity with respect to the displaced strand requiring a 3' overhang on the third strand and ATP. XPA-RPA repair complex participates in the nucleotide excision repair resolving DNA damage. XPA was found to bind psoralen-crosslinked TFO-induced triplexes only in conjunction with RPA but contributes to specificity in detection of triplex-induced helical distortions via RPA. Yes 2D-electrophoresis and mass spectrometry using artificial triplex probes. EMSA and DNase I study using the (GAA)n repeat from the first intron of frataxin alleles various triplexes 43

Zinc-dependent protein (human)

Yes

(GAA)n repeats

208

High mobility group (HMG) proteins (mouse)

Yes

Recombinant proteins incubated with triplex probe analyzed by time course PAGE and DNase I protection assays. Strand displacement assay using recombinant helicase incubated with DNA substrates and detected using PAGE and dimethyl sulfate footprinting.

purine-motif triplexes forming at (GGA)11 repeats

209 211

RecQ helicases

Yes

Melting of various forms of triplexes

5, 36, 38, 212

Nucleotide excision repair and mismatch repair proteins (human)

property of the single-strand DNA afne human replication protein A (RPA) could be due to competition for binding to the third (single) strand under equilibrium conditions.41 The fact that a comparable single-stranded DNA-binding protein poorly destabilizes triplex structures, however, suggests that RPA specically detects and processes triplexes as previously suggested.42 This has been further substantiated by immunodetection of triplex signatures in HeLa cells after siRNAs-induced suppression of RPA.41 A range of other triplex-binding proteins have been found in human,43 other mammals,44-46 flies47 and yeast48,49 (see Table 1). They belong to chromatin-associating protein families such as heterogeneous ribonucleoproteins (hnRNP), cytoplasmic type III intermediate lament (IF) proteins, transcription factors (TFs), high mobility group (HMG) box proteins as well as proteins involved in the cell cycle and DNA repair. While this suggests that cells throughout the eukaryotic kingdoms of life possess proteins able to recognize and process nucleic triple-helices it may as well simply reflect the preference of some (especially cationic) proteins for the higher negative charge densities of such multi-stranded nucleic acids structures. Abundance of putative triplex target sites in genomes. Polypurine regions spanning more than 15 nucleotides, which constitute putative triplex target sites (TTSs), are overrepresented in both prokaryotic and eukaryotic genomes.50-55 A special class of polypurine stretches that have the characteristics of mirror repeats are enriched in the yeast as well as the human genome and common in many other genomes.55,56 Mirror repeats are especially interesting because they can engage in inter- and

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .
Yes EMSA studies using proteins expressed in E. coli and ChIP. Anti-parallel motif triplexes 213 215

intramolecular triplex formation. This special type of intermolecular triplex could be formed by an RNA being transcribed from the repeat and binding back to the duplex, creating a perfectly matched triplex (Fig. 3).57 Intramolecular DNA-triplexes (H-DNA), on the other hand, consist of three strands of DNA where one strand folds back onto the duplex leaving an unpaired single strand (reviewed in ref. 58). Evidence that such tracts indeed form triple-helices was provided by in vitro assays using S1-nuclease, which preferentially targets single-stranded DNA, involving however a rather low pH.59 Using RNA-FISH, a recent study by Zheng et al. show a heterogeneous population of (GAA)n repeat containing ncRNA to localize in distinct subnuclear domains, which likely resemble genomic (GAA)(TTC) repeats, possibly forming a triplex at these repeats or targeting the single-strand of an H-DNA triplex already in place. Mirror repeats capable of forming H-DNA occur in promoters as well as coding regions of several disease-involved genes (reviewed in ref. 60). For Friedreich ataxia it has been demonstrated that the formation of such triple-helical structures are induced by the expansion of (GAA)n repeats, which causes the transcriptional silencing of the frataxin gene.61 Triple-helical structures in RNA. Intramolecular triplehelices in RNA are known to contribute to RNA folding and tertiary structure stability.62,63 These pseudoknot structures are also crucial for enzymatic function such as catalytic activity64,65 or ribosomal frameshifting during translation.66 In fact, the HIV virus shows absolute reliance on an ribosomal frameshift event during translation,67 and mutation analysis of the pseudoknot sequence found that intramolecular triple-helix formation in both

430

RNA Biology Volume 8 Issue 3

the minor as well as the major groove of mRNA is essential in -1 ribosomal frameshifting.68,69 These examples indicate that the cell employs intramolecular triplexformation in RNA, which likely extends to intermolecular RNA-triplexes and triplexes made of both RNA and DNA. Evidence for the Function of Triple-Helical Structures Triplex formation regulates gene transcription. The possibility of triplex formation in vivo raises the question of their functional roles. Computational studies show that putative triplex target sites are enriched upstream of eukaryotic genes,54,70 suggesting a possible role of triplexes in transcriptional regulation. Supporting evidence for this functional role is provided by several biological studies investigating triplex formation at specic gene loci as reviewed by Dyke.22 One of the most compelling pieces of evidence for a regulatory role for TFOinduced triplex formation in vivo is the Figure 3. Putative triple-helical structures formed by palindrome RNA or DNA mirror repeats. regulation of the dihydrofolate reductase The upper part illustrates how an RNA transcript from mirror repeat DNA can potentially result in (DHFR ) gene in U2OS cells.7 Here, a nona feedback signal via formation of a triplex at the same site. The lower part illustrates how mirror repeat DNA can form H-DNA. In both cases simple repeats easily comply with the respective protein-coding transcript (ncRNA) from constraints applied. a minor promoter interacts in a sequencespecic way to form a triplex within the downstream major promoter, which is responsible for 99% of the human breast cancer cells was shown using specically designed transcription of the DHFR gene. The transcript also interacts with polypurine hairpins that form intermolecular triplexes at polypyTFIIB and causes dissociation of the pre-initiation complex from rimidine target DNA sites.23 Aside from occluding DNA-binding the major promoter resulting in promoter-specic transcriptional sites triplexes could also affect the function of DNA-binding repression of the DHFR gene. While triplex formation was sup- proteins. There is evidence that the transcription factor GAGA ported by accompanying in vitro assays, the transcriptional inhi- adopts different functional roles depending on the conformation bition observed in vivo might also be attributed to formation of of the targeted DNA, which overlaps with putative TTSs and stable DNA-RNA duplexes12 possibly at the G-rich sequences in may hence form triplexes (see Table 1). the DHFR promoter. G-rich sequences on the sense strand have Interestingly, several TFs have been found to bind RNA as been shown to stall transcription in vitro71 and in vivo72 via interac- well as DNA.78 This suggests that an RNA-bound TF could tions of the nascent transcript with the DNA template. The means home in on its target loci guided by triplex-specic formation of stable RNADNA duplex formation involving a transcript of an between the RNA and the targeted duplex DNA. This concept upstream promoter remains to be investigated. has been exploited in biomedical applications for the site-specic If triplex-formation is indeed the principle cause of the silenc- delivery of mutagen agents by means of intermolecular triplex ing of DHFR in the above example, then a plausible mechanism is formation between a TFO and the targeted duplex DNA.24,79 It that triplex formation shields duplex DNA from duplex-targeting should be noted that in these biomedical applications the TFO proteins such as transcription factors.73-75 Such a mechanism was is often made of DNA or of an analogous molecule such as a shown to downregulate the Ets2 gene in vitro76 as well as in pros- peptide nucleic acid.26 Similarly, RNA-bound molecules could tate cancer cells.77 Here, specically designed TFOs overlap the facilitate triplex-formation as an anchor point to remain and act DNA binding site of the transcription factor Sp1 in the Ets2 pro- in the spatial proximity of a specic genomic context. Proof-ofmoter, restricting Sp1 from binding when the triplex is formed. principle was obtained using an articial TFO made of DNA Another example is the bcr/abl locus, where TFO-induced triplex that is physically linked to the SRE enhancer, which accommoformation was demonstrated by EMSA assays and its downregu- dates binding of the TFs SRF and ELK-1.80 Here, the SRE-TFO lating effect on bcr/abl transcription as well as cell growth was successfully recruits the TFs via the linked enhancer and forms shown in in vivo cell studies.19 Similarly, increased mortality of a triplex at its target locus, which otherwise shows no afnity for

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .

www.landesbioscience.com

RNA Biology

431

SRF and ELK-1. Hence, these proteins might be able to perform their function in the vicinity of the targeted locus such as modifying the epigenetic landscape or altering gene transcription. The fact that many proteins are known to interact with RNA81,82 suggests that the cell may employ such a cargo-delivery mechanism. Target specicity. Any loci specic targeting mechanism requires a unique, non-ambiguous address. Computational studies indicate that large numbers of putative TTSs occur exactly once in the human as well as in the mouse genome83 suggesting that these sequences generally full the requirement of site-specicity. Mirror repeats able to form H-DNA are also non-randomly distributed in the genome and are associated with genes involved in cell trafcking and communication.84 Indeed, formation of H-DNA on polypurinepolypyrimidine sequences was shown by S1 nuclease assays implicating such sequences as cis-acting transcriptional regulators (reviewed in ref. 59). The proposed formation of H-DNA on (GC)n repeats and a resulting change in gene expression has been reported to strongly depend on the length of the repeat and the induction of local superhelical strain.74 The applied assays do, however, neither rule out the possible formation of G-quadruplexes at these repeats, which have been linked to a similar phenotypic behavior85,86 nor the formation of RNA-DNA hybrids and R-loops.71 In fact, in vitro studies indicated that neither H-DNA nor G-quadruple formation is a pre-condition for the transcriptional arrest of T7 RNA polymerase at G-tracts.71 (GAA)n repeats, which do not comply to G-quadruple formation rules, have been shown to stall replication in vivo87 as well as cause transcriptional arrest.88,89 So-called suicidal mirrorrepeats were shown to cause permanent or temporary transcriptional arrest in vitro. Here, the non-template strand displaced by the polymerase during DNA synthesis folds back to the duplex prompting the formation of H-DNA.90 This transcriptional interference has been well studied in the genomic locus of the c-myc oncogene (reviewed in ref. 91). c-Myc participates in several pathways determining the cellular fate, and its abnormal expression has been associated with cancer.92 The H-DNA-forming sequences at this locus map to translocation breakpoints, which can activate or disrupt specic genes in its proximity bypassing or undermining cellular control mechanisms.93,94 Endogenous triplex-forming microRNAs interfere with viral replication. Triplex formation may not be limited to the cell nucleus. A recent study by Kanak et al. implicates cytosolic triplex formation in viral defence of eukaryotic cells. The authors showed that HeLa-CD4 + as well as T-lymphotic cells are able to resist HIV-1 infection when expressing specific endogenous microRNAs that are able to form stable triplexes with DNA motifs from HIV-1, whereas cells expressing control microRNAs lacking triplex-formation capability show no resistance to HIV1. Importantly, the proposed binding of the investigated microRNAs incorporates a substantial portion of triplets lacking at least one of the two triplex-stabilizing hydrogen bonds, which is contrary to models obtained from thermodynamic in vitro studies.95 More research is clearly needed to substantiate cytosolic triplex formation in vivo and reveal the cytosolic determinants involved in this process.

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .

Involvement of triple-helices in RNA processing. Triplehelical structures may also be involved in post-transcriptional processing of RNAs. Many proteins regulating translation and post-transcriptional events such as RNA splicing and editing associate with double- or single-stranded RNAs through RNA recognition motifs.81,96 The formation of pure RNA triple-helical structures97 provides another RNA motif to be considered as either the target of proteins or as a decoy. Some circumstantial evidence suggests that triplex formation prevents binding and activation of RNA-dependent protein kinase (PKR) and possibly other nucleotide-dependent proteins in vitro.98 Active PKR inhibits protein synthesis in higher cells and leads to growth inhibition in response to viral infection,99 and binding to double-stranded RNA activates PKR in vitro.100 The formation of a triple-helix between duplex RNA and a TFO can efciently inhibit the binding of PKR and, subsequently, its activation.98 In the eukaryotic cell, endogenous RNA may hence adopt triplex rather than duplex form to avoid triggering cellular responses involving proteins that monitor foreign (viral) duplex RNA. It also suggests that intermolecular triplex-formation may play a role in RNA-RNA interactions in other contexts, which may be responsible for the cytosolic staining patterns obtained immunoand fluorescence-detection studies.4,31 HnRNP proteins, of which ve members show specicity for pyrimidine-rich DNA, also bind to triple-helices43 implicating triplex formation in mRNA splicing. HnRNP A2/B1 and L have been linked to pre-mRNA processing and have been found in the splicosome.101 HnRNP I in particular has been found to bind to polypyrimidine tracts of introns.102,103 HnRNP K and E1 bind predominately polyC single-stranded DNA and may hence target the unbound strand of H-DNA or G-quadruplexes as well as be responsible for the single-strand binding observed in the study.104 Polypyrimidine tracts have been associated with alternative splicing events105 raising the question to what extent TFOs able to bind to their antisense polypurine may be involved in protein recruitment and alternative splicing mechanisms. In fact, it has been demonstrated in vitro that a polypurine stretch with alternating A and G can stimulate the splicing of the surrounding intron(s) by modulating the binding of polypyrimidine tract-binding protein.106 Furthermore, recent work indicates that self-splicing of precursor RNA in group I and II introns involves enzymatic triplexes (reviewed in ref. 64). U2 and U6 RNAs, which form a ribozyme complex that is also part of the spliceosome, and which are related to the self-splicing precursor RNA, can carry out RNA-based catalysis.107 Importantly, sequences involved in the rst splicing step seem to be recognized by the snRNAs without the aid of additional proteins.108 It remains to be shown if U2 and U6 RNAs also rely on catalytic triplex formation. Triplex-mediated chromatin structure, organization and epigenetics. Chromosome organization and architecture plays a central role in the regulation of differentiation and development, and has strong effects on DNA replication,109 transcription and splicing.110 The formation of H-DNA provides contact points that may participate in chromatin organization possibly through ncRNAs, which in turn may interact with the nuclear matrix or nuclear matrix-associated proteins.10 Repeat regions in particular

432

RNA Biology Volume 8 Issue 3

may be capable of forming links between different regions of DNA via triplex formation and in vitro studies on plasmid DNA substantiate the formation of such DNA loops.111 Proteins specically binding triple-helices may orchestrate chromatin organization. Intermediate lament (IF) proteins are an important structural feature in eukaryote cells that can bind to triplexes and also to other higher-order structures such as G-quadruplexes (reviewed in ref. 112). One IF protein, Vimentin, which is localized in the cytosol as well as in the nuclear matrix,113 is known to attach to several cytosolic organelles potentially positioning them in xed locations and may perform similar functions in the nucleus. Triplexes may serve as anchor points or be used as landmarks to recruit chromatin regions to nuclear compartments. A recent study links a ncRNA to epigenetic modications in gene promoters of non-coding genes.8 Here, a TFO-directed triplex has been suggested to mediate the recruitment of DNA methyltransferases, and in particular the de novo methyltransferase Dmnt3b, to gene promoters and hence regulate the methylation status of DNA. DNA methylation is known to alter the expression of nearby genes114 and plays an essential part in gene imprinting and cell differentiation.115 While RNase H, EMSA and psoralen-based assays demonstrate that the binding of the ncRNA to the promoter is neither via formation of RNA-DNA hybrids nor due to mediating proteins, further investigations into the proposed triple-helical formation are required. In particular, the observed deviation from the common triplex model needs to be addressed by characterising the triplex regarding its binding motif (see Appendix) or by evaluating alternative models such as R-triplexes, where the third strand binds parallel to an identical nucleotide sequence in the duplex.116 The authors also report the formation of heterochromatin in promoters targeted by the ncRNA subsequently leading to the silencing of downstream genes. The recruitment of chromatin remodelling complexes is due to a second motif in the ncRNA,8 demonstrating how a non-coding transcript can guide enzymes to act at specic genomic loci. The timed availability of a TFO-containing transcript together with the site-specicity of TFO-induced triplex formation may indeed provide the very means for the dynamic regulation of the epigenome. Similarly, (GAA) n repeats and other polypurinepolypyrimidine tracts prone to form H-DNA were found to cause regional epigenetic changes in cell models117,118 especially hypoacetylation and hypermethylation.119 Triplex-helices also influence chromatin condensation in a more direct way. The accommodation of the third strand in the major groove changes the rigidity of duplex DNA. Triplexed DNA is less flexible and cannot as easily wrap around a histone complex, which subsequently affects nucleosome reconstitution.117,120,121 Conversely, studies based on DNaseI cleavage patterns showed that TFOs cannot bind to DNA that is tightly associated with the nucleosome core (reviewed in ref. 122). Taken together, triple-helix formation at targeted doublestranded DNA may participate in retaining open chromatin, assist in accurate nucleosome positioning as well as modication of histone complexes and DNA itself. Triple-helices induce mutagenesis, recombination and DNA repair. Triplex formation causes duplex DNA to adopt non-B

conformations, which promotes genetic instability, mutation and recombination leading to repeat extension or genomic rearrangement.123 Both inter- and intramolecular triplexes are mutagenic and induce recombination as well as DNA repair (reviewed in ref. 124 and 125). Transgenic mice models indicate that H-DNA can induce genetic instability in the mammalian genome.126 Similarly, a purine-motif TFO induces mutagenesis in mammalian cells vefold above that of a control oligonucleotide with scrambled sequence.127 TFO-directed mutations were introduced in somatic cells of mice, validating triplex formation in vivo as well as its mutagenic character in several tissues.128 It appears that the TFO-induced helical alteration of the duplex provokes the mutagenesis129 and possibly contributes to common chromosomal translocation in cancer (reviewed in ref. 79). A more exotic triplex, where the third strand binds parallel to an identical nucleotide sequence in the duplex, was proposed to function as a primer inducing DNA replication and promoting DNA rearrangements.130 Phage and bacterial DNA polymerases could accommodate three DNA strands in their enzymatic centre and are particularly prone to elongate G-rich primers. Regulation of the Regulator?

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .
To be useful to the cell as a biological signaling mechanism, triplex formation must be reversible in a controllable way. As indicated above such a negative control system exists in the form of helicases, which actively unwind triplexes. Since triplex structures can interfere with polymerase activity it is crucial for the cell to melt such obstacles efciently before or during DNA replication. Helicases may also interact with DNA-binding proteins such as TFs to resolve triplexes in a locus-specic manner although this is purely speculative. Another potential mechanism for regulating triplex formation is sequence modication. RNA editing converts cytosine into uracil or adenosine into inosine in double-stranded RNA131 and occurs in transcripts of many coding and non-coding sequences.132 It has been shown that replacement of certain nucleotides by inosine in a triple-helix alters its stability.133,134 Methylation of cytosine to 5'-methyl-cytosine is another indigenous nucleotide modication that occurs in DNA and RNA (reviewed in refs. 135 and 136), which is particularly benecial for the formation and stability of pyrimidine-motif triplexes due to its elevated pKa value.137,138 Only non-CpG methylations, like CpC, CpT and TpC, are relevant to pyrimidine-motif triplexes. Such modications have been reported in plants,139 flies140 and mammals141,142 and seem to be mediated by the protein Dnmt2,140,142 and Dnmt3b.143 Dnmt2 appears in fact to be an RNA methyltransferase144,145 that plays an important role in developmental processes146 while Dmnt3b has been associated with hypermethylation of non-CpG cytosines at the PCG-1alpha promoter in human diabetic patients143 and has recently been suggested to associate with TFO-induced triplexes.8 Furthermore, methylation of CpT has been linked to mammalian embryonic stem cells but not to somatic cells, indicating that this reversible modication may be involved in cell differentiation.147,148 Non-CpG-methylation was also found to be reinstated at each

www.landesbioscience.com

RNA Biology

433

generation in spite of the fact that the majority of the sperm genomes contained next to no methylated non-CpG sites.149 The full extent of such modications in higher organism is yet to be revealed. Besides these natural modications, several articial modications to TFOs have also been shown to increase triplex stability (reviewed in ref. 150). Future Prospects Given the conceptual simplicity of nucleic triple-helix formation and the availability of all required components, it would be surprising if triplex formation were not utilized functionally by the cell. Challenging aspects. While there is now considerable circumstantial evidence for triplex formation and function in vivo, categorical proof is still missing. Incisive new experimental approaches are needed that can unambiguously identify the location, composition and function of naturally occurring triplexes. New triplex-capture approaches and further characterization of triplex formation rules are required in order to determine the true range of roles triplexes may play in vivo. Below, we discuss how to proceed to gain such a denitive picture. The vast majority of the genome in higher organisms is transcribed into ncRNA in complex, developmentally controlled and tissue-dependent patterns, which strongly suggests that ncRNAs play a central role in the regulation of development and cellular processes where TFO-induced triplex formation may play an important part. Consistent with this, it is now clear that ncRNA is involved in the regulation of many genetic and epigenetic processes including transcription, translation and chromatin modication, and numerous mechanisms have been identied (reviewed in ref. 6 and 151). The challenges are to determine the extent of the involvement of triplex formation in gene regulation, and to detail the specic regulatory mechanisms and the sources and targets of the triplex-forming ncRNA transcripts involved. One component of this task comprises genome-wide mapping of TFOs and TTSs, along with identifying their dynamic complex formation, in a time- and tissue-dependent manner. This may be achievable using a combination of existing technologies (such as nextgeneration sequencing) and novel approaches, several of which we describe below. Important facets include developing more detailed TFO-TTS binding rules, creating tissue-specic catalogues of expressed putative TFOs, and occupancy measurement of TTSs and associating TFOs with their target TTSs to build models of regulatory (sub-)networks. Rening triplex formation rules. While the abundance of previous biochemical experiments provides a good overview on how triplexes form, their different setups and protocols make it infeasible to infer an accurate thermodynamic binding model. Instead, the screening of the vast sequence space with high throughput methods is required. Furthermore, it would facilitate the development of more accurate in silico methods for TFO and TTS prediction by lling in gaps in our knowledge about the minimum requirements on length and purine-content as well as tolerable mismatches for triplex formation.

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .

Identication of triplex-forming sequences in genomic DNA. Empirical determination of the precise sites of triplex formation in vivo, and the dynamic changes in their position during cell differentiation, should be possible by deep sequencing of both the DNA and RNA content of triplexes isolated by chromatin immunoprecipitation with specic anti-triplex antibodies or by nuclease digestion targeted either at triplexes (analogous to that used to determine DNaseI hypersensitivity sites,152 e.g., using nuclease S1) or designed to remove duplex DNA (e.g., by DNaseI digestion). Indeed a signicant gap in the large-scale analyses to dene genome architecture and biology, such as the ENCODE project,153 is the lack of information on the dynamical occurrence of non-canonical structures in the genome, such as triplexes, Z-DNA tracts and G-quartets, all of which are known to occur and would be expected to play important roles in gene regulation and genome biology, and which should be amenable to targeted high-throughput sequence analysis. The data obtained will also enable comparison with the known or expected composition of such sequences (see below), with an expected validation of both, as well as the identity of the sequences forming the third (Hoogsteen) strand, with a concomitant renement of the rules of engagement and the precise identity of the sequences involved. Mining deep sequencing data for putative triple-helices. To identify expressed TFOs, short ncRNA sequences from deepsequencing experiments can also be ltered computationally for their propensity to function as TFOs using the general triplex formation rules reviewed here, or applying more detailed rules derived from empirical deep sequencing data, possibly in combination with RNA-folding predictions. Applying this ltering to sequenced ncRNA from a particular cell or tissue under given conditions would result in a tissue-specic catalogue of putative, expressed TFOs. Repeating this experiment under multiple conditions and intersecting the sets of predicted, expressed TFOs can uncover sets of differentially expressed TFOs. Further in silico ltering could be applied by mapping the TFOs to potential target sites in the genome, again using triplex formation rules and comparison to empirical data. Given that most promoters contain a predicted, often unique TTS,83 gene expression data can be leveraged by correlating changes in the expression of genes containing a (putative) TTS with changes in expression of a matching (putative) TFO. Rening gene regulatory networks. Determining the existence and extent of triplex-based gene regulatory networks will enable the construction and validation of network models. The critical steps in building such a model are identifying TFOs and their cognate TTSs. This can be accomplished using the approaches described above for mapping TTSs, cataloguing TFOs and associating TFO with cognate TTS using binding rules and inference based on coordinate expression of the TFO and genes associated with its predicted TTS targets, perhaps incorporating adjacency lters such as suggested by the example of the transcriptional regulation of DHFR .7 Such inferred network models will of course need to be veried to establish the direct effect of triplex formation on the epigenetic status and/or transcription of target genes, which can procede using RNA-based approaches

434

RNA Biology Volume 8 Issue 3

analogous to those currently used for testing protein-based regulation models. Appendix Triplex motifs. In the [T,C]-motif, also referred to as the pyrimidine motif, thymine binds adenosine (T-AT) and cytosine binds guanine (C-GC) in the polypurine strand of the duplex. Due to the Hoogsteen conguration, the third strand is oriented parallel to the polypurine strand. The cytosine in the third strand must be protonated to be able to form the second Hoogsteen hydrogen bond, which makes cytosine-containing triple-helices pH-dependent, and hence limits their stability at physiological pH.154 The positive charge provided by the protonated cytosine, however, compensates for unfavorable charge repulsions of the polyanionic oligonucleotide backbones. Contiguous protonated cytosines, on the other hand, result in unfavorable charge repulsion between them.155 Such a cytosine-tract can be replaced by guanine in the TFO in some circumstances.156 In the [G,A]-motif, also referred to as the purine motif, (G-GC) and (A-AT) triplets are formed in reverse Hoogsteen conguration, thus resulting in anti-parallel triple-helices.157 While guanine-rich TFOs show good DNA afnity under physiological pH,158 triplex formation by long tracts of guanine competes with quadruplex formation,159 which may limit their in vivo effectiveness.160 [G,A]-TFOs can tolerate a small fraction of thymines, which form (T-AT) triplets.161,162 The steric properties of guanine and thymine allow for the remaining [G,T] motif, where (G-GC) and (T-AT) triplets are formed, which prefers one orientation over the other depending on its underlying sequence.163 By contrast, in the parallel (recombination) R-triplex, the third nucleotide strand orientates in parallel to the identical nucleotide sequence in the duplex.116 The R-triplex is considerably less stable compared to above-mentioned triplex congurations especially for intermolecular formation.164 Triple-helix formation and sequence specicity. Triplehelical formation comes in two flavors: intramolecular and intermolecular. While in the rst case the third strand is physically tethered to the duplex molecule providing the target,165 in the second case the TFO is an independent molecule. Intramolecular DNA triple-helices, often also referred to as H-DNA, were rst observed in plasmid DNA at homopurinehomopyrimidine mirror repeats (reviewed in ref. 166). There are four possible congurations in which three of the four DNA strands form H-DNA in the pyrimidine and the purine motif, respectively (see ref. 59 and 167 for detailed reviews on the biological relevance of H-DNA). Intramolecular triple-helices also form in RNA where they may contribute to RNA folding and tertiary structure stability.62,168

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .

Sequence specicity and binding afnity of the third strand to its target duplex is a crucial factor for functional triplex formation in vivo. For a given overall base composition, the actual sequence of the participating strands has a strong impact on stability of the triple-helix (reviewed in ref. 95). Long A-tract duplex DNA, for example, seems to be unfavorable for triplex formation due to its higher propeller twist and rigidity.169,170 Long TFOs generally tend to form more stable complexes than shorter ones, although some high-afnity structures have been demonstrated with TFOs as short as nine nucleotides.171 However, TFOs of length greater than 17 nt may exhibit substantial afnity for a secondary (shorter) target site by tolerating mismatches, loops or other structures172-174 or by alternating the target strand in the duplex (as described in the next paragraph).163 Triplexes in principle tolerate mismatches between the strands,175,176 however these have a strong destabilizing effect that increases with the number of contiguous mismatches.177 The destabilizing effect depends furthermore on the nature of the mismatch and its positional location, i.e., a terminal mismatch causes less disruption to the triplex than mismatch in the centre of the triple-helix.178 A single TFO can bind to a target duplex in which the polypurine tract switches strands.163 In this situation, different segments of the TFO bind to different strands in the duplex. The alternating target strand preference is accompanied by a switch in the binding conguration (Hoogsteen/reverse Hoogsteen) of the corresponding binding segment in the TFO.163 All six possible junctions for combining two short TFO-segments have shown to be functional in vitro.179-181 The cellular environment likely imposes specic constraints on the potential for triple-helix formation. High concentrations of multivalent cations promote triplex stability by compensating for the unfavorable electrostatic repulsion of the three negatively charged oligonucleotide backbones.158,182,183 It is well established that metal cations are naturally present in DNA in vivo.184 However, the stabilizing effect of any multivalent cation is limited by its competition with available monovalent counterfeits.185,186 Physiological concentrations of potassium, for example, have been shown to reduce the efciency of triplex formation,185 most likely in favor of quadruplex formation.187 Polyamines also promote triplex formation under physiological pH,188 as do some charge-neutralizing basic polypeptides.189 In addition to the effects of cations and pH,190,191 triplex formation is also highly dependent on temperature, organic solvents2,73,192-194 and chromatin accessibility,122,195 all of which complicate in silico assessments of the potential of a given TFO and target sequence to form triplexes in vivo. So far, little is known about how local microenvironments and the involvement of other factors such as proteins might favor the formation and/or increase the stability of particular types of triplexes in vivo.

www.landesbioscience.com

RNA Biology

435

References
1. Felsenfeld G, Davies DR, Rich A. Formation of a three-stranded polynucleotide molecule. J Am Chem Soc 1957; 79:2023-4. 2. Moser HE, Dervan PB. Sequence-specific cleavage of double helical DNA by triple helix formation. Science 1987; 238:645-50. 3. Doan TL, Perrouault L, Chassignol M, Nguyen TT, Hlne C. Sequence-targeted chemical modifications of nucleic acids by complementary oligonucleotides covalently linked to porphyrins. Nucleic Acids Res 1987; 15:8643-59. 4. Lubitz I, Zikich D, Kotlyar A. Specific high-affinity binding of thiazole orange to triplex and G-quadruplex DNA. Biochemistry 2010; 49:3567-74. 5. Jain A, Bacolla A, Chakraborty P, Grosse F, Vasquez KM. Human DHX9 helicase unwinds triple-helical DNA structures. Biochemistry 2010; 49:6992-9. 6. Mattick JS, Taft RJ, Faulkner GJ. A global view of genomic informationmoving beyond the gene and the master regulator. Trends Genet 2010; 26:21-8. 7. Martianov I, Ramadass A, Barros AS, Chow N, Akoulitchev A. Repression of the human dihydrofolate reductase gene by a non-coding interfering transcript. Nature 2007; 445:666-70. 8. Schmitz KM, Mayer C, Postepska A, Grummt I. Interaction of noncoding RNA with the rDNA promoter mediates recruitment of DNMT3b and silencing of rRNA genes. Genes Dev 2010; 24:2264-9. 9. Kanak M, Alseiari M, Balasubramanian P, Addanki K, Aggarwal M, Noorali S, et al. Triplex-forming MicroRNAs form stable complexes with HIV-1 provirus and inhibit its replication. Appl Immunohistochem Mol Morphol 2010; 18:532-45; DOI: 10.1097/ PAI.0b013e3181e1ef6a. 10. Zheng R, Shen Z, Tripathi V, Xuan Z, Freier SM, Bennett CF, et al. Polypurine-repeat-containing RNAs: a novel class of long non-coding RNA in mammalian cells. J Cell Sci 2010; 123:3734-44. 11. Szewczak AA, Ortoleva-Donnelly L, Ryder SP, Moncoeur E, Strobel SA. A minor groove RNA triple helix within the catalytic core of a group I intron. Nat Struct Biol 1998; 5:1037-42. 12. Roberts RW, Crothers DM. Stability and properties of double and triple helices: dramatic effects of RNA or DNA backbone composition. Science 1992; 258:1463-6. 13. Han H, Dervan PB. Sequence-specific recognition of double helical RNA and RNA.DNA by triple helix formation. Proc Natl Acad Sci USA 1993; 90:3806-10. 14. Asensio JL, Carr R, Brown T, Lane AN. Conformational and Thermodynamic Properties of Parallel Intramolecular Triple Helices Containing a DNA RNA or 2'-OMeDNA Third Strand. J Am Chem Soc 1999; 121:11063-70. 15. Escud C, Franois JC, Sun JS, Ott G, Sprinzl M, Garestier T, et al. Stability of triple helices containing RNA and DNA strands: experimental and molecular modeling studies. Nucleic Acids Res 1993; 21:5547-53. 16. Bernal-Mndez E, Leumann CJ. Conformational diversity versus nucleic acid triplex stability, a combinatorial study. J Biol Chem 2001; 276:35320-7. 17. McDonald CD, Maher LJ. Recognition of duplex DNA by RNA polynucleotides. Nucleic Acids Res 1995; 23:500-6. 18. Soukup GA, Maher JJ. Selection and characterization of RNAs that relieve transcriptional interference in Escherichia coli. Nucleic Acids Res 1998; 26:2715-22. 19. Rapozzi V, Cogoi S, Spessotto P, Risso A, Bonora GM, Quadrifoglio F, et al. Antigene effect in K562 cells of a PEG-conjugated triplex-forming oligonucleotide targeted to the bcr/abl oncogene. Biochemistry 2002; 41:502-10. 20. Hewett PW, Daft EL, Laughton CA, Ahmad S, Ahmed A, Murray JC. Selective inhibition of the human tie-1 promoter with triplex-forming oligonucleotides targeted to Ets binding sites. Mol Med 2006; 12:8-16.

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .

21. Carbone GM, Napoli S, Valentini A, Cavalli F, Watson DK, Catapano CV. Triplex DNA-mediated downregulation of Ets2 expression results in growth inhibition and apoptosis in human prostate cancer cells. Nucleic Acids Res 2004; 32:4358-67. 22. Dyke MWV. Do DNA Triple Helices or Quadruplexes Have a Role in Transcription? DNA Conformation and Transcription: Springer US 2005; 105-26. 23. de Almagro MC, Coma S, No V, Ciudad CJ. Polypurine hairpins directed against the template strand of DNA knock down the expression of mammalian genes. J Biol Chem 2009; 284:11579-89. 24. Chin JY, Schleifman EB, Glazer PM. Repair and recombination induced by triple helix DNA. Front Biosci 2007; 12:4288-97. 25. Schleifman EB, Chin JY, Glazer PM. Triplex-mediated gene modification. Methods Mol Biol 2008; 435:175-90. 26. Simon P, Cannata F, Concordet JP, Giovannangeli C. Targeting DNA with triplex-forming oligonucleotides to modify gene sequence. Biochimie 2008; 90:1109-16. 27. Lee JS, Burkholder GD, Latimer LJ, Haug BL, Braun RP. A monoclonal antibody to triplex DNA binds to eucaryotic chromosomes. Nucleic Acids Res 1987; 15:1047-61. 28. Agazie YM, Lee JS, Burkholder GD. Characterization of a new monoclonal antibody to triplex DNA and immunofluorescent staining of mammalian chromosomes. J Biol Chem 1994; 269:7019-23. 29. Agazie YM, Burkholder GD, Lee JS. Triplex DNA in the nucleus: direct binding of triplex-specific antibodies and their effect on transcription, replication and cell growth. Biochem J 1996; 316:461-6. 30. Burkholder GD, Latimer LJ, Lee JS. Immunofluorescent localization of triplex DNA in polytene chromosomes of Chironomus and Drosophila. Chromosoma 1991; 101:11-8. 31. Ohno M, Fukagawa T, Lee JS, Ikemura T. Triplexforming DNAs in the human interphase nucleus visualized in situ by polypurine/polypyrimidine DNA probes and antitriplex antibodies. Chromosoma 2002; 111:201-13. 32. Piergentili R, Mencarelli C. Drosophila melanogaster kl-3 and kl-5 Y-loops harbor triple-stranded nucleic acids. J Cell Sci 2008; 121:1605-12. 33. Stollar BD, Raso VIC. Antibodies recognise specific structures of triple-helical polynucleotides built on poly(A) or poly(dA). Nature 1974; 250:231-4. 34. Gorab E, Amabis JM, Stocker AJ, Drummond L, Stollar BD. Potential sites of triple-helical nucleic acid formation in chromosomes of Rhynchosciara (Diptera: Sciaridae) and Drosophila melanogaster. Chromosome Res 2009; 17:821-32. 35. Bagasra O, Stir AE, Pirisi-Creek L, Creek KE, Bagasra AU, Glenn N, et al. Role of micro-RNAs in regulation of lentiviral latency and persistence. Appl Immunohistochem Mol Morphol 2006; 14:276-90. 36. Maine IP, Kodadek T. Efficient unwinding of triplex DNA by a DNA helicase. Biochem Biophys Res Commun 1994; 204:1119-24. 37. Dixon BP, Lu L, Chu A, Bissler JJ. RecQ and RecG helicases have distinct roles in maintaining the stability of polypurine.polypyrimidine sequences. Mutat Res 2008; 643:20-8. 38. Kopel V, Pozner A, Baran N, Manor H. Unwinding of the third strand of a DNA triple helix, a novel activity of the SV40 large T-antigen helicase. Nucleic Acids Res 1996; 24:330-5. 39. Brosh RM, Majumdar A, Desai S, Hickson ID, Bohr VA, Seidman MM. Unwinding of a DNA triple helix by the Werner and Bloom syndrome helicases. J Biol Chem 2001; 276:3024-30. 40. Peleg M, Kopel V, Borowiec JA, Manor H. Formation of DNA triple helices inhibits DNA unwinding by the SV40 large T-antigen helicase. Nucleic Acids Res 1995; 23:1292-9. 41. Wu Y, Rawtani N, Thazhathveetil AK, Kenny MK, Seidman MM, Brosh RM. Human replication protein A melts a DNA triple helix structure in a potent and specific manner. Biochemistry 2008; 47:5068-77. 42. Vasquez KM, Christensen J, Li L, Finch RA, Glazer PM. Human XPA and RPA DNA repair proteins participate in specific recognition of triplex-induced helical distortions. Proc Natl Acad Sci USA 2002; 99:5848-53.

43. Guillonneau F, Guieysse AL, Caer JPL, Rossier J, Praseuth D. Selection and identification of proteins bound to DNA triple-helical structures by combination of 2D-electrophoresis and MALDI-TOF mass spectrometry. Nucleic Acids Res 2001; 29:2427-36. 44. Li G, Tolstonog GV, Traub P. Interaction in vitro of type III intermediate filament proteins with triplex DNA. DNA Cell Biol 2002; 21:163-88. 45. Ciotti P, Dyke MWV, Bianchi-Scarr G, Musso M. Characterization of a triplex DNA-binding protein encoded by an alternative reading frame of loricrin. Eur J Biochem 2001; 268:225-34. 46. Kusic J, Tomic B, Divac A, Kojic S. Human initiation protein Orc4 prefers triple stranded DNA. Mol Biol Rep 2010; 37:2317-22. 47. Jimnez-Garca E, Vaquero A, Espins ML, Soliva R, Orozco M, Bernus J, et al. The GAGA factor of Drosophila binds triple-stranded DNA. J Biol Chem 1998; 273:24640-8. 48. Nelson LD, Musso M, Dyke MWV. The yeast STM1 gene encodes a purine motif triple helical DNAbinding protein. J Biol Chem 2000; 275:5573-81. 49. Musso M, Bianchi-Scarr G, Dyke MWV. The yeast CDP1 gene encodes a triple-helical DNA-binding protein. Nucleic Acids Res 2000; 28:4090-6. 50. Ussery D, Soumpasis DM, Brunak S, Staerfeldt HH, Worning P, Krogh A. Bias of purine stretches in sequenced chromosomes. Comput Chem 2002; 26:531-41. 51. Hoyne PR, Edwards LM, Viari A, Maher LJ. Searching genomes for sequences with the potential to form intrastrand triple helices. J Mol Biol 2000; 302:797-809. 52. Kohwi Y, Malkhosyan SR, Kohwi-Shigematsu T. Intramolecular dG.dG.dC triplex detected in Escherichia coli cells. J Mol Biol 1992; 223:817-22. 53. Behe MJ. An overabundance of long oligopurine tracts occurs in the genome of simple and complex eukaryotes. Nucleic Acids Res 1995; 23:689-95. 54. Goi JR, de la Cruz X, Orozco M. Triplex-forming oligonucleotide target sequences in the human genome. Nucleic Acids Res 2004; 32:354-60. 55. Schroth GP, Ho PS. Occurrence of potential cruciform and H-DNA forming sequences in genomic DNA. Nucleic Acids Res 1995; 23:1977-83. 56. Clark RM, Bhaskar SS, Miyahara M, Dalgliesh GL, Bidichandani SI. Expansion of GAA trinucleotide repeats in mammals. Genomics 2006; 87:57-67. 57. Dey I, Rath PC. A novel rat genomic simple repeat DNA with RNA-homology shows triplex (H-DNA)like structure and tissue-specific RNA expression. Biochem Biophys Res Commun 2005; 327:276-86. 58. Mirkin SM. Expandable DNA repeats and human disease. Nature 2007; 447:932-40. 59. Jain A, Wang G, Vasquez KM. DNA triple helices: biological consequences and therapeutic potential. Biochimie 2008; 90:1117-30. 60. Bissler JJ. Triplex DNA and human disease. Front Biosci 2007; 12:4536-46. 61. Wells RD. DNA triplexes and Friedreich ataxia. FASEB J 2008; 22:1625-34. 62. Holland JA, Hoffman DW. Structural features and stability of an RNA triple helix in solution. Nucleic Acids Res 1996; 24:2841-8. 63. Mitton-Fry RM, DeGregorio SJ, Wang J, Steitz TA, Steitz JA. Poly(A) tail recognition by a viral RNA element through assembly of a triple helix. Science 2010; 330:1244-7. 64. Toor N, Keating KS, Pyle AM. Structural insights into RNA splicing. Curr Opin Struct Biol 2009; 19:260-6. 65. Qiao F, Cech TR. Triple-helix structure in telomerase RNA contributes to catalysis. Nat Struct Mol Biol 2008; 15:634-40. 66. Brierley I, Gilbert RJC, Pennell S. RNA pseudoknots and the regulation of protein synthesis. Biochem Soc Trans 2008; 36:684-9. 67. Gareiss PC, Miller BL. Ribosomal frameshifting: an emerging drug target for HIV. Curr Opin Investig Drugs 2009; 10:121-8.

436

RNA Biology Volume 8 Issue 3

68. Chen G, Chang KY, Chou MY, Bustamante C, Tinoco I. Triplex structures in an RNA pseudoknot enhance mechanical stability and increase efficiency of -1 ribosomal frameshifting. Proc Natl Acad Sci USA 2009; 106:12706-11. 69. Chou MY, Chang KY. An intermolecular RNA triplex provides insight into structural determinants for the pseudoknot stimulator of -1 ribosomal frameshifting. Nucleic Acids Res 2010; 38:1676-85. 70. Goi JR, Vaquerizas JM, Dopazo J, Orozco M. Exploring the reasons for the large density of triplexforming oligonucleotide target sequences in the human regulatory regions. BMC Genomics 2006; 7:63. 71. Belotserkovskii BP, Liu R, Tornaletti S, Krasilnikova MM, Mirkin SM, Hanawalt PC. Mechanisms and implications of transcription blockage by guaninerich DNA sequences. Proc Natl Acad Sci USA 2010; 107:12816-21. 72. Krasilnikova MM, Samadashwily GM, Krasilnikov AS, Mirkin SM. Transcription through a simple DNA repeat blocks replication elongation. EMBO J 1998; 17:5095-102. 73. Maher LJ, Wold B, Dervan PB. Inhibition of DNA binding proteins by oligonucleotide-directed triple helix formation. Science 1989; 245:725-30. 74. Kohwi Y, Kohwi-Shigematsu T. Altered gene expression correlates with DNA structure. Genes Dev 1991; 5:2547-54. 75. Cooney M, Czernuszewicz G, Postel EH, Flint SJ, Hogan ME. Site-specific oligonucleotide binding represses transcription of the human c-myc gene in vitro. Science 1988; 241:456-9. 76. Mavrothalassitis GJ, Watson DK, Papas TS. Molecular and functional characterization of the promoter of ETS2, the human c-ets-2 gene. Proc Natl Acad Sci USA 1990; 87:1047-51. 77. Carbone GM, McGuffie EM, Collier A, Catapano CV. Selective inhibition of transcription of the Ets2 gene in prostate cancer cells by a triplex-forming oligonucleotide. Nucleic Acids Res 2003; 31:833-43. 78. Cassiday LA, Maher LJ. Having it both ways: transcription factors that bind DNA and RNA. Nucleic Acids Res 2002; 30:4118-26. 79. Vasquez KM. Targeting and processing of site-specific DNA interstrand crosslinks. Environ Mol Mutagen 2010; 51:527-39. 80. Svinarchuk F, Nagibneva I, Cherny D, Si-Ali SA, Pritchard LL, Robin P, et al. Recruitment of transcription factors to the target site by triplex-forming oligonucleotides. Nucleic Acids Res 1997; 25:3459-64. 81. Collins LJ, Kurland CG, Biggs P, Penny D. The modern RNP world of eukaryotes. J Hered 2009; 100:597-604. 82. Babitzke P, Baker CS, Romeo T. Regulation of translation initiation by RNA binding proteins. Annu Rev Microbiol 2009; 63:27-44. 83. Wu Q, Gaddis SS, MacLeod MC, Walborg EF, Thames HD, DiGiovanni J, et al. High-affinity triplex-forming oligonucleotide target sequences in mammalian genomes. Mol Carcinog 2007; 46:15-23. 84. Bacolla A, Collins JR, Gold B, Chuzhanova N, Yi M, Stephens RM, et al. Long homopurine*homopyrimidine sequences are characteristic of genes expressed in brain and the pseudoautosomal region. Nucleic Acids Res 2006; 34:2663-75. 85. Johnson JE, Smith JS, Kozak ML, Johnson FB. In vivo veritas: using yeast to probe the biological functions of G-quadruplexes. Biochimie 2008; 90:1250-63. 86. Siddiqui-Jain A, Grand CL, Bearss DJ, Hurley LH. Direct evidence for a G-quadruplex in a promoter region and its targeting with a small molecule to repress c-MYC transcription. Proc Natl Acad Sci USA 2002; 99:11593-8. 87. Krasilnikova MM, Mirkin SM. Replication stalling at Friedreichs ataxia (GAA)n repeats in vivo. Mol Cell Biol 2004; 24:2286-95.

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .

88. Son LS, Bacolla A, Wells RD. Sticky DNA: in vivo formation in E. coli and in vitro association of long GAA*TTC tracts to generate two independent supercoiled domains. J Mol Biol 2006; 360:267-84. 89. Sarkar PS, Brahmachari SK. Intramolecular triplex potential sequence within a gene downregulates its expression in vivo. Nucleic Acids Res 1992; 20:5713-8. 90. Samadashwily GM, Dayn A, Mirkin SM. Suicidal nucleotide sequences for DNA polymerization. EMBO J 1993; 12:4975-83. 91. Ponzielli R, Katz S, Barsyte-Lovejoy D, Penn LZ. Cancer therapeutics: targeting the dark side of Myc. Eur J Cancer 2005; 41:2485-501. 92. Hoffman B, Liebermann DA. Apoptotic signaling by c-MYC. Oncogene 2008; 27:6462-72. 93. Wang G, Vasquez KM. Naturally occurring H-DNAforming sequences are mutagenic in mammalian cells. Proc Natl Acad Sci USA 2004; 101:13448-53. 94. Belotserkovskii BP, Silva ED, Tornaletti S, Wang G, Vasquez KM, Hanawalt PC. A triplex-forming sequence from the human c-MYC promoter interferes with DNA transcription. J Biol Chem 2007; 282:32433-41. 95. Plum GE. Thermodynamics of oligonucleotide triple helices. Biopolymers 1998; 44:241-56. 96. Lunde BM, Moore C, Varani G. RNA-binding proteins: modular design for efficient function. Nat Rev Mol Cell Biol 2007; 8:479-90. 97. Toor N, Keating KS, Taylor SD, Pyle AM. Crystal structure of a self-spliced group II intron. Science 2008; 320:77-82. 98. Vuyisich M, Beal PA. Regulation of the RNAdependent protein kinase by triple helix formation. Nucleic Acids Res 2000; 28:2369-74. 99. Jaramillo ML, Abraham N, Bell JC. The interferon system: a review with emphasis on the role of PKR in growth control. Cancer Invest 1995; 13:327-38. 100. Manche L, Green SR, Schmedt C, Mathews MB. Interactions between double-stranded RNA regulators and the protein kinase DAI. Mol Cell Biol 1992; 12:5238-48. 101. Jurica MS, Licklider LJ, Gygi SR, Grigorieff N, Moore MJ. Purification and characterization of native spliceosomes suitable for three-dimensional structural analysis. RNA 2002; 8:426-39. 102. Bothwell AL, Ballard DW, Philbrick WM, Lindwall G, Maher SE, Bridgett MM, et al. Murine polypyrimidine tract binding protein. Purification, cloning and mapping of the RNA binding domain. J Biol Chem 1991; 266:24657-63. 103. Ghetti A, Piol-Roma S, Michael WM, Morandi C, Dreyfuss G. hnRNP I, the polypyrimidine tract-binding protein: distinct nuclear localization and association with hnRNAs. Nucleic Acids Res 1992; 20:3671-8. 104. Matunis MJ, Michael WM, Dreyfuss G. Characterization and primary structure of the poly(C)binding heterogeneous nuclear ribonucleoprotein complex K protein. Mol Cell Biol 1992; 12:164-71. 105. Raponi M, Buratti E, Llorian M, Stuani C, Smith CWJ, Baralle D. Polypyrimidine tract binding protein regulates alternative splicing of an aberrant pseudoexon in NF1. FEBS J 2008; 275:6101-8. 106. Tanaka K, Watakabe A, Shimura Y. Polypurine sequences within a downstream exon function as a splicing enhancer. Mol Cell Biol 1994; 14:1347-54. 107. Valadkhan S, Mohammadi A, Jaladat Y, Geisler S. Protein-free small nuclear RNAs catalyze a two-step splicing reaction. Proc Natl Acad Sci USA 2009; 106:11901-6. 108. Valadkhan S, Mohammadi A, Wachtel C, Manley JL. Protein-free spliceosomal snRNAs catalyze a reaction that resembles the first step of splicing. RNA 2007; 13:2300-11. 109. Schneider R, Grosschedl R. Dynamics and interplay of nuclear architecture, genome organization and gene expression. Genes Dev 2007; 21:3027-43.

110. Luco RF, Pan Q, Tominaga K, Blencowe BJ, PereiraSmith OM, Misteli T. Regulation of alternative splicing by histone modifications. Science 2010; 327:996-1000. 111. Hampel KJ, Burkholder GD, Lee JS. Plasmid dimerization mediated by triplex formation between polypyrimidine-polypurine repeats. Biochemistry 1993; 32:1072-7. 112. Tolstonog GV, Li G, Shoeman RL, Traub P. Interaction in vitro of type III intermediate filament proteins with higher order structures of single-stranded DNA, particularly with G-quadruplex DNA. DNA Cell Biol 2005; 24:85-110. 113. Tolstonog GV, Sabasch M, Traub P. Cytoplasmic intermediate filaments are stably associated with nuclear matrices and potentially modulate their DNA-binding function. DNA Cell Biol 2002; 21:213-39. 114. Vaillant I, Paszkowski J. Role of histone and DNA methylation in gene regulation. Curr Opin Plant Biol 2007; 10:528-33. 115. Geiman TM, Muegge K. DNA methylation in early development. Mol Reprod Dev 2010; 77:105-13. 116. Zhurkin VB, Raghunathan G, Ulyanov NB, CameriniOtero RD, Jernigan RL. A parallel DNA triplex as a model for the intermediate in homologous recombination. J Mol Biol 1994; 239:181-200. 117. Ruan H, Wang YH. Friedreichs ataxia GAA.TTC duplex and GAA.GAA.TTC triplex structures exclude nucleosome assembly. J Mol Biol 2008; 383:292-300. 118. Han YJ, de Lanerolle P. Naturally extended CT.AG repeats increase H-DNA structures and promoter activity in the smooth muscle myosin light chain kinase gene. Mol Cell Biol 2008; 28:863-72. 119. Soragni E, Herman D, Dent SYR, Gottesfeld JM, Wells RD, Napierala M. Long intronic GAA*TTC repeats induce epigenetic changes and reporter gene silencing in a molecular model of Friedreich ataxia. Nucleic Acids Res 2008; 36:6056-65. 120. Westin L, Blomquist P, Milligan JF, Wrange O. Triple helix DNA alters nucleosomal histone-DNA interactions and acts as a nucleosome barrier. Nucleic Acids Res 1995; 23:2184-91. 121. Goobes R, Cohen O, Minsky A. Unique condensation patterns of triplex DNA: physical aspects and physiological implications. Nucleic Acids Res 2002; 30:2154-61. 122. Brown PM, Fox KR. DNA triple-helix formation on nucleosome core particles. Effect of length of the oligopurine tract. Eur J Biochem 1999; 261:301-10. 123. Bacolla A, Wells RD. Non-B DNA conformations as determinants of mutagenesis and human disease. Mol Carcinog 2009; 48:273-85. 124. Chin JY, Glazer PM. Repair of DNA lesions associated with triplex-forming oligonucleotides. Mol Carcinog 2009; 48:389-99. 125. Wang G, Vasquez KM. Models for chromosomal replication-independent non-B DNA structure-induced genetic instability. Mol Carcinog 2009; 48:286-98. 126. Wang G, Carbajal S, Vijg J, DiGiovanni J, Vasquez KM. DNA structure-induced genomic instability in vivo. J Natl Cancer Inst 2008; 100:1815-7. 127. Wang G, Seidman MM, Glazer PM. Mutagenesis in mammalian cells induced by triple helix formation and transcription-coupled repair. Science 1996; 271:802-5. 128. Vasquez KM, Narayanan L, Glazer PM. Specific mutations induced by triplex-forming oligonucleotides in mice. Science 2000; 290:530-3. 129. Kalish JM, Seidman MM, Weeks DL, Glazer PM. Triplex-induced recombination and repair in the pyrimidine motif. Nucleic Acids Res 2005; 33:3492-502. 130. Lestienne PP. Are there three polynucleotide strands in the catalytic centre of DNA polymerases? Biochimie 2009; 91:1523-30. 131. Farajollahi S, Maas S. Molecular diversity through RNA editing: a balancing act. Trends Genet 2010; 26:221-30. 132. Mattick JS, Amaral PP, Dinger ME, Mercer TR, Mehler MF. RNA regulation of epigenetic processes. Bioessays 2009; 31:51-9.

www.landesbioscience.com

RNA Biology

437

133. Mills M, Vlker J, Klump HH. Triple helical structures involving inosine: there is a penalty for promiscuity. Biochemistry 1996; 35:13338-44. 134. Ueno Y, Shibata A, Matsuda A, Kitade Y. Thermal stability of triple helical DNAs containing 2'-deoxyinosine and 2'-deoxyxanthosine. Bioorg Med Chem 2004; 12:6581-6. 135. Lan J, Hua S, He X, Zhang Y. DNA methyltransferases and methyl-binding proteins of mammals. Acta Biochim Biophys Sin (Shanghai) 2010; 42:243-52. 136. Motorin Y, Lyko F, Helm M. 5-methylcytosine in RNA: detection, enzymatic formation and biological functions. Nucleic Acids Res 2010; 38:1415-30. 137. Lee JS, Woodsworth ML, Latimer LJ, Morgan AR. Poly(pyrimidine). poly(purine) synthetic DNAs containing 5-methylcytosine form stable triplexes at neutral pH. Nucleic Acids Res 1984; 12:6603-14. 138. Xodo LE, Manzini G, Quadrifoglio F, van der Marel GA, van Boom JH. Effect of 5-methylcytosine on the stability of triple-stranded DNAa thermodynamic study. Nucleic Acids Res 1991; 19:5625-31. 139. Cao X, Jacobsen SE. Locus-specific control of asymmetric and CpNpG methylation by the DRM and CMT3 methyltransferase genes. Proc Natl Acad Sci USA 2002; 99:16491-8. 140. Kunert N, Marhold J, Stanke J, Stach D, Lyko F. A Dnmt2-like protein mediates DNA methylation in Drosophila. Development 2003; 130:5083-90. 141. White GP, Watt PM, Holt BJ, Holt PG. Differential patterns of methylation of the IFNgamma promoter at CpG and non-CpG sites underlie differences in IFN gamma gene expression between human neonatal and adult CD45RO-T cells. J Immunol 2002; 168:2820-7. 142. Mund C, Musch T, Strdicke M, Assmann B, Li E, Lyko F. Comparative analysis of DNA methylation patterns in transgenic Drosophila overexpressing mouse DNA methyltransferases. Biochem J 2004; 378:763-8. 143. Barrs R, Osler ME, Yan J, Rune A, Fritz T, Caidahl K, et al. Non-CpG methylation of the PGC-1alpha promoter through DNMT3B controls mitochondrial density. Cell Metab 2009; 10:189-98. 144. Jurkowski TP, Meusburger M, Phalke S, Helm M, Nellen W, Reuter G, et al. Human DNMT2 methylates tRNA(Asp) molecules using a DNA methyltransferaselike catalytic mechanism. RNA 2008; 14:1663-70. 145. Schaefer M, Hagemann S, Hanna K, Lyko F. Azacytidine inhibits RNA methylation at DNMT2 target sites in human cancer cell lines. Cancer Res 2009; 69:8127-32. 146. Rai K, Chidester S, Zavala CV, Manos EJ, James SR, Karpf AR, et al. Dnmt2 functions in the cytoplasm to promote liver, brain and retina development in zebrafish. Genes Dev 2007; 21:261-6. 147. Ramsahoye BH, Biniszkiewicz D, Lyko F, Clark V, Bird AP, Jaenisch R. Non-CpG methylation is prevalent in embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc Natl Acad Sci USA 2000; 97:5237-42. 148. Lister R, Pelizzola M, Dowen RH, Hawkins RD, Hon G, Tonti-Filippini J, et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 2009; 462:315-22. 149. Grandjean V, Yaman R, Cuzin F, Rassoulzadegan M. Inheritance of an epigenetic mark: the CpG DNA methyltransferase 1 is required for de novo establishment of a complex pattern of non-CpG methylation. PLoS One 2007; 2:1136. 150. Duca M, Vekhoff P, Oussedik K, Halby L, Arimondo PB. The triple helix: 50 years later, the outcome. Nucleic Acids Res 2008; 36:5123-38. 151. Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet 2009; 10:155-9. Lian H, Thompson WA, Thurman R, 152. Stamatoyannopoulos JA, Noble WS, Lawrence CE. Automated mapping of large-scale chromatin structure in ENCODE. Bioinformatics 2008; 24:1911-6.

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .

153. Consortium ENCODEP. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature 2007; 447:799-816. 154. Sugimoto N, Wu P, Hara H, Kawamoto Y. pH and cation effects on the properties of parallel pyrimidine motif DNA triplexes. Biochemistry 2001; 40:9396-405. 155. Vlker J, Klump HH. Electrostatic effects in DNA triple helices. Biochemistry 1994; 33:13502-8. 156. Giovannangli C, Rouge M, Garestier T, Thuong NT, Hlne C. Triple-helix formation by oligonucleotides containing the three bases thymine, cytosine and guanine. Proc Natl Acad Sci USA 1992; 89:8631-5. 157. Beal PA, Dervan PB. Second structural motif for recognition of DNA by oligonucleotide-directed triple-helix formation. Science 1991; 251:1360-3. 158. Floris R, Scaggiante B, Manzini G, Quadrifoglio F, Xodo LE. Effect of cations on purine.purine.pyrimidine triple helix formation in mixed-valence salt solutions. Eur J Biochem 1999; 260:801-9. 159. Olivas WM, Maher LJ. Competitive triplex/quadruplex equilibria involving guanine-rich oligonucleotides. Biochemistry 1995; 34:278-84. 160. Alunni-Fabbroni M, Manzini G, Quadrifoglio F, Xodo LE. Guanine-rich oligonucleotides targeted to a critical R.Y site located in the Ki-ras promoter. The effect of competing self-structures on triplex formation. Eur J Biochem 1996; 238:143-51. 161. Mills M, Klump HH. Systematic mutations in the third strand of a purine motif DNA triple helix: a story of a molecule which hides its tail. Nucleosides, Nucleotides Nucleic Acids 1998; 17:1919-36. 162. Mills M, Arimondo PB, Lacroix L, Garestier T, Klump H, Mergny JL. Chemical modification of the third strand: differential effects on purine and pyrimidine triple helix formation. Biochemistry 2002; 41:357-66. 163. de Bizemont T, Duval-Valentin G, Sun JS, Bisagni E, Garestier T, Hlne C. Alternate strand recognition of double-helical DNA by (T,G)-containing oligonucleotides in the presence of a triple helix-specific ligand. Nucleic Acids Res 1996; 24:1136-43. 164. Shchyolkina AK, Borisova OF, Minyat EE, Timofeev EN, Ilicheva IA, Khomyakova EB, et al. Parallel purine-pyrimidine-purine triplex: experimental evidence for existence. FEBS Lett 1995; 367:81-4. 165. Sklenr V, Feigon J. Formation of a stable triplex from a single DNA strand. Nature 1990; 345:836-8. 166. Mirkin SM, Frank-Kamenetskii MD. H-DNA and related structures. Annu Rev Biophys Biomol Struct 1994; 23:541-76. 167. Zain R, Sun JS. Do natural DNA triple-helical structures occur and function in vivo? Cell Mol Life Sci 2003; 60:862-70. 168. Klaff P, Riesner D, Steger G. RNA structure and the regulation of gene expression. Plant Mol Biol 1996; 32:89-106. 169. Sandstrm K, Wrmlnder S, Grslund A, Leijon M. A-tract DNA disfavours triplex formation. J Mol Biol 2002; 315:737-48. 170. James PL, Brown T, Fox KR. Thermodynamic and kinetic stability of intermolecular triple helices containing different proportions of C+*GC and T*AT triplets. Nucleic Acids Res 2003; 31:5598-606. 171. Svinarchuk F, Paoletti J, Malvy C. An unusually stable purine(purine-pyrimidine) short triplex. The third strand stabilizes double-stranded DNA. J Biol Chem 1995; 270:14068-71. 172. Roberts RW, Crothers DM. Specificity and stringency in DNA triplex formation. Proc Natl Acad Sci USA 1991; 88:9397-401. 173. Wang Y, Patel DJ. Bulge defects in intramolecular pyrimidine.purine.pyrimidine DNA triplexes in solution. Biochemistry 1995; 34:5696-704. 174. Fox KR, Flashman E, Gowers D. Secondary binding sites for triplex-forming oligonucleotides containing bulges, loops and mismatches in the third strand. Biochemistry 2000; 39:6714-25. 175. Sun JS, Bizemont TD, Duval-Valentin G, MontenayGarestier T, Hlne C. Extension of the range of recognition sequences for triple helix formation by oligonucleotides containing guanines and thymines. C R Acad Sci III 1991; 313:585-90.

176. Rusling DA, Brown T, Fox KR. DNA triple-helix formation at target sites containing duplex mismatches. Biophys Chem 2006; 123:134-40. 177. Gowers DM, Fox KR. DNA triple helix formation at oligopurine sites containing multiple contiguous pyrimidines. Nucleic Acids Res 1997; 25:3787-94. 178. Mergny JL, Sun JS, Rouge M, Montenay-Garestier T, Barcelo F, Chomilier J, et al. Sequence specificity in triple-helix formation: experimental and theoretical studies of the effect of mismatches on triplex stability. Biochemistry 1991; 30:9791-8. 179. Brodin P, Sun JS, Mouscadet JF, Auclair C. Optimization of alternate-strand triple helix formation at the 5"-TpA-3" and 5"-ApT-3" junctions. Nucleic Acids Res 1999; 27:3029-34. 180. de Bizemont T, Sun JS, Garestier T, Hlne C. New junction models for alternate-strand triple-helix formation. Chem Biol 1998; 5:755-62. 181. Marchand C, Sun JS, Bailly C, Waring MJ, Garestier T, Hlne C. Optimization of alternate-strand triple helix formation at the 5CpG3 and 5GpC3 junction steps. Biochemistry 1998; 37:13322-9. 182. Malkov VA, Voloshin ON, Soyfer VN, FrankKamenetskii MD. Cation and sequence effects on stability of intermolecular pyrimidine-purine-purine triplex. Nucleic Acids Res 1993; 21:585-91. Paris C, Geinguenaud F, Gouyette C, Liquier J, 183. Lacoste J. Mechanism of copper mediated triple helix formation at neutral pH in Drosophila satellite repeats. Biophys J 2007; 92:2498-506. 184. Sagripanti JL, Goering PL, Lamanna A. Interaction of copper with DNA and antagonism by other metals. Toxicol Appl Pharmacol 1991; 110:477-85. Olivas WM, Maher LJ. Overcoming potassium185. mediated triplex inhibition. Nucleic Acids Res 1995; 23:1936-41. 186. Wu P, Hara H, Kawamoto Y, Sugimoto N. Effect of cytosine protonation and cation on thermodynamic properties of parallel DNA triplex family. Nucleic Acids Res Suppl 2001; 1:39-40. 187. Sen D, Gilbert W. A sodium-potassium switch in the formation of four-stranded G4-DNA. Nature 1990; 344:410-4. 188. Hampel KJ, Crosson P, Lee JS. Polyamines favor DNA triplex formation at neutral pH. Biochemistry 1991; 30:4455-9. 189. Potaman VN, Sinden RR. Stabilization of intramolecular triple/single-strand structure by cationic peptides. Biochemistry 1998; 37:12952-61. 190. Lee HT, Arciniegas S, Marky LA. Unfolding thermodynamics of DNA pyrimidine triplexes with different molecularities. J Phys Chem B 2008; 112:4833-40. 191. Lee HT, Khutsishvili I, Marky LA. DNA complexes containing joined triplex and duplex motifs: melting behavior of intramolecular and bimolecular complexes with similar sequences. J Phys Chem B 2010; 114:541-8. 192. Lyamichev VI, Mirkin SM, Frank-Kamenetskii MD, Cantor CR. A stable complex between homopyrimidine oligomers and the homologous regions of duplex DNAs. Nucleic Acids Res 1988; 16:2165-78. 193. Povsic TJ, Dervan PB. Triple helix formation by oligonucleotides on DNA extended to the physiological pH range. J Am Chem Soc 1989; 111:59-3061. 194. Jaumot J, Avia A, Eritja R, Tauler R, Gargallo R. Resolution of parallel and antiparallel oligonucleotide triple helices formation and melting processes by multivariate curve resolution. J Biomol Struct Dyn 2003; 21:267-78. 195. Brown PM, Fox KR. Nucleosome core particles inhibit DNA triple helix formation. Biochem J 1996; 319:607-11. 196. Adkins NL, Hagerman TA, Georgel P. GAGA protein: a multi-faceted transcription factor. Biochem Cell Biol 2006; 84:559-67. 197. Granok H, Leibovitch BA, Shaffer CD, Elgin SC. Chromatin. Ga-ga over GAGA factor. Curr Biol 1995; 5:238-41. 198. Lehmann M. Anything else but GAGA: a nonhistone protein complex reshapes chromatin structure. Trends Genet 2004; 20:15-22.

438

RNA Biology Volume 8 Issue 3

199. Dyke MWV, Nelson LD, Weilbaecher RG, Mehta DV. Stm1p, a G4 quadruplex and purine motif triplex nucleic acid-binding protein, interacts with ribosomes and subtelomeric Y DNA in Saccharomyces cerevisiae. J Biol Chem 2004; 279:24323-33. 200. Katayama T, Inoue N, Torigoe H. Location of the triplex DNA-binding domain of Saccharomyces cerevisiae Stm1 protein. Nucleic Acids Symp Ser 2007; 53:123-4. 201. Marton HA, Desiderio S. The Paf1 complex promotes displacement of histones upon rapid induction of transcription by RNA polymerase II. BMC Mol Biol 2008; 9:4. 202. Rao JE, Craig NL. Selective recognition of pyrimidine motif triplexes by a protein encoded by the bacterial transposon Tn7. J Mol Biol 2001; 307:1161-70. 203. Tolstonog GV, Mothes E, Shoeman RL, Traub P. Isolation of SDS-stable complexes of the intermediate filament protein vimentin with repetitive, mobile, nuclear matrix attachment region and mitochondrial DNA sequence elements from cultured mouse and human fibroblasts. DNA Cell Biol 2001; 20:531-54. 204. Kiyama R, Camerini-Otero RD. A triplex DNAbinding protein from human cells: purification and characterization. Proc Natl Acad Sci USA 1991; 88:10450-4.

205. Guieysse AL, Praseuth D, Hlne C. Identification of a triplex DNA-binding protein from human cells. J Mol Biol 1997; 267:289-98. 206. Shibata A, Ueno Y, Shinbo K, Nakanishi M, Matsuda A, Kitade Y. Synthesis of linked triple helical DNAs possessing high affinity to triple helical DNA binding protein. Bioorg Med Chem Lett 2006; 16:1410-3. 207. Musso M, Nelson LD, Dyke MWV. Characterization of purine-motif triplex DNA-binding proteins in HeLa extracts. Biochemistry 1998; 37:3086-95. 208. Mueler W, Bassili G, Epplen C, Keyl HG, Epplen JT. Protein binding to simple repetitive sequences depends on DNA secondary structure(s). Chromosome Res 1999; 7:163-6. 209. Suda T, Mishima Y, Takayanagi K, Asakura H, Odani S, Kominami R. A novel activity of HMG domains: promotion of the triple-stranded complex formation between DNA containing (GGA/TCC)11 and d(GGA)11 oligonucleotides. Nucleic Acids Res 1996; 24:4733-40. 210. Mishima Y, Suda T, Kominami R. Formation of a triple-stranded DNA between d(GGA:TCC) repeats and d(GGA) repeat oligonucleotides. J Biochem 1996; 119:805-10.

211. Jain A, Akanchha S, Rajeswari MR. Stabilization of purine motif DNA triplex by a tetrapeptide from the binding domain of HMGBI protein. Biochimie 2005; 87:781-90. 212. Plyler J, Jasheway K, Tuesuwan B, Karr J, Brennan JS, Kerwin SM, et al. Real-time investigation of SV40 large T-antigen helicase activity using surface plasmon resonance. Cell Biochem Biophys 2009; 53:43-52. 213. Lange SS, Reddy MC, Vasquez KM. Human HMGB1 directly facilitates interactions between nucleotide excision repair proteins on triplex-directed psoralen interstrand crosslinks. DNA Repair (Amst) 2009; 8:865-72. 214. Thoma BS, Wakasugi M, Christensen J, Reddy MC, Vasquez KM. Human XPC-hHR23B interacts with XPA-RPA in the recognition of triplex-directed psoralen DNA interstrand crosslinks. Nucleic Acids Res 2005; 33:2993-3001. 215. Zhao J, Jain A, Iyer RR, Modrich PL, Vasquez KM. Mismatch repair and nucleotide excision repair proteins cooperate in the recognition of DNA interstrand crosslinks. Nucleic Acids Res 2009; 37:4420-9.

2 0 1 1 L a n d e s B i o s c i e n c e . D o n o t d i s t r i b u t e .

www.landesbioscience.com

RNA Biology

439

Vous aimerez peut-être aussi