Next Article in Journal
Differential Modulation of Catecholamine and Adipokine Secretion by the Short Chain Fatty Acid Receptor FFAR3 and α2-Adrenergic Receptors in PC12 Cells
Previous Article in Journal
Puerarin—A Promising Flavonoid: Biosynthesis, Extraction Methods, Analytical Techniques, and Biological Effects
Previous Article in Special Issue
Differential Gene Expression following DHX36/G4R1 Knockout Is Associated with G-Quadruplex Content and Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Special Issue “Bioinformatics of Unusual DNA and RNA Structures”

1
Department of Biology and Ecology, Faculty of Science, University of Ostrava, 710 00 Ostrava, Czech Republic
2
Institute of Biophysics, Czech Academy of Sciences, Královopolská 135, 612 00 Brno, Czech Republic
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(10), 5226; https://doi.org/10.3390/ijms25105226
Submission received: 18 April 2024 / Revised: 29 April 2024 / Accepted: 6 May 2024 / Published: 10 May 2024
(This article belongs to the Special Issue Bioinformatics of Unusual DNA and RNA Structures)

1. Introduction

Nucleic acids are not only static carriers of genetic information but also play vital roles in controlling cellular lifecycles through their fascinating structural diversity. Gone are the days when DNA was perceived as a right-handed double helix and mediator RNA was imagined mostly as a linear single-stranded molecule. We now know that various structures, including quadruplexes, triplexes, and looped and zig-zagged structures, dynamically interact with proteins and cooperate in fine-tuning molecular processes. Section 2 of this Editorial is an overview of this exciting field of science and serves as a short guide to the topic of “unusual” DNA and RNA structures. In Section 3, we briefly summarize and highlight the main messages in the eight impactful articles published in this Special Issue, each of them representing various and important aspects of current research efforts. Finally, Section 4 discusses the current and future directions and implications for applications.

2. Overview of Unusual Nucleic Acid Structures

Unusual nucleic acid structures (UNas) can be defined as noncanonical nucleic acids differing from the classical double-stranded structure of B-DNA and are mostly abbreviated to non-B DNA (a term introduced in the early 80s [1]). However, this may be quite misleading, as these structures often arise also in RNA or DNA–RNA hybrid molecules (hence our use of the novel term “unusual nucleic acid structures”, abbreviated to UNas). The division of UNas may be quite tricky; historically, the most traditional categories are duplexes [2,3], triplexes [4], and quadruplexes [5]. In addition, all these “-plexes” can be intramolecular (containing only a single nucleic acid strand) or intermolecular (wherein two or more nucleic acid strands are involved) [6,7]. Unusual duplexes are mostly represented by the A-form [8] and Z-form of nucleic acids [9]. Triplexes are characterized by a triple-helical DNA structure in the case of intramolecular DNA triplexes (sometimes called H-DNA) [10] or by noncoding RNA pairing with DNA duplexes through Hoogsteen interactions in the case of intermolecular triplexes [11,12]. Quadruplexes can be divided into G-quadruplexes and i-motifs [13]. Aside from these, there are also somewhat special UNas called R-loops (three-stranded structures consisting of a DNA–RNA hybrid and a displaced strand of DNA) [14] and cruciforms, the latter of which form four-way-junction, double-stranded-stems, and single-stranded loops [15]. All the abovementioned UNas are depicted in Table 1, and key information about them is summarized.
It is worth mentioning that the in vivo existence (and potential biological relevance) of even-higher-order “-plexes” cannot be excluded. Such structures could then be named pentaplexes, sextuplexes, and so on [43].
The formation of most UNas is dependent on primary sequence information [44]; specific UNa-forming sequential motifs have been discovered and experimentally validated (Table 1). Thanks to the rapid development of computer technology and capacities, it is now possible to predict the occurrence of UNa-forming sequences in the whole eukaryotic chromosomes relatively easily, and several user-friendly web servers have been developed for this purpose. One such example allows the prediction of G-quadruplex-forming sequences [45], cruciform-forming sequences [46], R-loop-forming sequences [47], and Z-DNA-forming sequences: the DNA analyzer web server (https://bioinformatics.ibp.cz/#/, accessed on 29 April 2024).

3. Current Research Highlights

Here, we would like to shortly highlight the main findings of works published in this Special Issue and encourage readers to read through the articles in their entirety.
Anthony Mittermaier and his team developed a very useful (and user-friendly) web server that allows users to analyze multiplicities of their provided G-quadruplex-forming sequences (https://www.mcgill.ca/mittermaierlab/greg-webserver, accessed on 29 April 2024). In addition, their article presents a detailed bioinformatic survey of the G-quadruplex polymorphism in human gene promoter regions, linking G-quadruplex polymorphisms to biological functions and providing new criteria with which to identify biologically relevant G-quadruplex-forming regions [48].
Another freely accessible service was developed by Jiří Šťastný’s group from Mendel University. Existing computer programs cannot easily predict where R-loops might occur in DNA. To address this, a new tool called R-loop tracker was developed (https://bioinformatics.ibp.cz/#/analyse/rloopr, accessed on 29 April 2024). This free web-based tool can predict R-loops in genomic DNA and allows researchers to compare these predictions to other DNA analyses [47].
It was previously found that G-quadruplexes can arise in long noncoding RNAs (lncRNAs) [33]. The study by Singh et al. identified lncRNA clusters with G4-forming sequences in cervical cancer patients, confirmed the formation of G-quadruplexes in specific lncRNAs, and discussed their roles as potential prognostic biomarkers for cervical cancer [49].
A recent article by Nicoletto et al. discusses the presence, conservation, and localization of putative G-quadruplex-forming sequences in human arboviruses [50]. Arboviruses are transmitted by arthropod vectors (arthropod-borne viruses, i.e., arboviruses) and comprise many important human pathogens, including Dengue virus, West Nile virus, Zika virus, or Tick-borne encephalitis virus [51]. Their study reveals the predominant locations of G-quadruplex-forming sequences in coding sequences and three-prime untranslated regions (3′UTRs). It also highlights their regulatory roles, emphasizing the potential of using G-quadruplex structures as antiviral targets [50].
The article by Gumina and Richardson et al. discusses the role of G-quadruplexes and the DHX36 helicase in gene expression regulation, particularly in cancer cells, highlighting the impact of DHX36 knockout on gene expression associated with G-quadruplex content in promoters or gene regions [52]. The findings suggest that DHX36 knockout leads to subtle but widespread changes in gene expression and provides valuable insights into the complex interplay between G-quadruplex structures, helicases like DHX36, and gene expression regulation, especially in the context of cancer [52].
The study by Feng, Luo et al. explores the effects of potassium (K+) and sodium (Na+) ions on global G-quadruplex formation in rice (Oryza sativa) [53]. The authors utilized a high-throughput method called BG4-DNA-IP-seq (DNA immunoprecipitation with anti-BG4 antibody coupled with sequencing). One of the exciting findings is that K+-specific G-quadruplexes are more associated with active histone marks and low DNA methylation levels compared to Na+-specific G-quadruplexes. This important research will facilitate the functional characterization of G-quadruplexes in rice and allow the potential use of specific G-quadruplex locations for biotechnological advancements in the future [53].
The study by Shavkunov et al. focused on tRNA fragments (tRFs) and their roles in interspecies interactions in bacterial communities [54]. Their research highlighted the emergence of novel types of RNAs and their potential significance in bacterial and eukaryotic cells [54].
The last published work in this Special Issue is a thorough review by Zulfiqar et al. dealing with Virus-Induced Gene Silencing (VIGS), mainly with respect to crop improvement [55]. VIGS represents a powerful tool for analyzing gene function and inducing heritable epigenetic modifications. This review also highlights the role of VIGS in developing crop varieties with improved agronomic traits and stress tolerance [55].

4. Future Perspectives

There is a growing number of bioinformatic tools for UNas prediction and biophysical characterization [45,56,57,58,59] as well as for determining their roles in various diseases including cancer [60,61]. Contemporary, specific antibodies against cruciforms [62,63], left-handed nucleic acids [64], G-quadruplexes [65], and i-motifs [66] have also been developed, allowing effective analyses of UNas both in vitro and in situ. Even if UNas are often difficult to sequence, current methods and their modifications [67] allow accurate sequencing and determination in genomes, leading to the finalization of telomere-to-telomere gapless assemblies [68,69,70,71]. Although there are currently many tools for the prediction and experimental validation of UNas, their structural bioinformatical characterization or modeling is somewhat lagging. In the field of protein science, there are currently many approaches for the de novo (ab initio) prediction of structures only from the amino acid sequences, e.g., AlphaFold [72] or trRosetta [73], and they are also usually accessible via a user-friendly interface [73,74]. In the case of UNas, no such straightforward method exists so far. Although some pioneering works have been published [75,76], they rely on arbitrary (user-provided) instructions and (sequential/spatial) restrictions; in addition, only structures with previously known similar (experimentally solved) templates can be modeled. Moreover, the work toward an ab initio nucleic-acid-structure-modeling tool is complicated by the fact that UNa formation is often driven by additional (but important) factors like negative/positive supercoiling (in the case of DNA) [77], chromatin epigenetic marks [78], chemical modifications of nucleobases [79], molecular crowding conditions/local microenvironment [80,81], interacting proteins [82], and other (de)stabilizers. In other words, predicting nucleic acid structure can be paradoxically even more challenging than predicting the structures of proteins.
Experimental or modeled structures of UNas can be further inspected using virtual screening/high-throughput molecular docking to determine which known chemical or natural substances are capable of specific binding, as successfully applied, e.g., in the case of bimolecular human telomeric G-quadruplexes [83]. The resulting UNas and their binders can be further characterized using molecular dynamics methods [84],constituting a complementary approach to biophysical methods of wet-lab characterization. Unfortunately, another limiting factor here is the lack of user-friendly software that would allow a wide range of scientists to carry out these analyses independently. Considering the computational complexity of these analyses, an ideal solution could be an integrative web server allowing scientists to analyze UNa .pdb structures using natural-language commands with the help of artificial intelligence [85].
So far, for the entirety of the UNa entity, only G-quadruplexes have been considered the primary target in two clinical trials with CX-3543 and CX-5461 compounds [86,87]. The main pitfall of UNa-binding compounds has been low specificity and relatively high toxicity in vivo, as, for example, in the case of the known in vitro G-quadruplex stabilizer TMPyP4 [88]. Later, more specific compounds were developed, recognizing, e.g., only parallel or antiparallel types of G-quadruplexes [89]. We believe that advances in bioinformatic methods will soon allow the implementation of the well-known concept of one drug–one target in the field of UNas; this would allow selective targeting of particular pathological UNas that arise, for example, due to nucleotide repeat expansion [90,91]. Altogether, UNas represent very promising molecular targets, and the current boom in methods of computational biology can pave the way for their future application in drug discovery.

Author Contributions

Conceptualization, M.B.; software, V.B.; resources, M.B. and V.B.; original draft preparation, M.B. and V.B.; writing—review and editing, P.P., V.B. and M.B.; visualization, M.B.; project administration, P.P. All authors have read and agreed to the published version of the manuscript.

Funding

M.B. was supported by the European Union under the LERCO project number CZ.10.03.01/00/22_003/0000003 via the Operational Programme Just Transition.

Acknowledgments

We would like to express our sincere gratitude to all contributors who decided to submit their excellent research for our Special Issue “Bioinformatics of Unusual DNA and RNA Structures”. We are very thankful to Adriana Volná and Jiří Červeň for a fruitful discussion and their proofreading services.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Zimmer, C.; Marck, C.; Guschlbauer, W. Z-DNA and Other non-B-DNA Structures Are Reversed to B-DNA by Interaction with Netropsin. FEBS Lett. 1983, 154, 156–160. [Google Scholar] [CrossRef] [PubMed]
  2. Rich, A. Right-Handed and Left-Handed DNA: Conformational Information in Genetic Material. Cold Spring Harb. Symp. Quant. Biol. 1983, 47, 1–12. [Google Scholar] [CrossRef] [PubMed]
  3. Conner, B.N.; Takano, T.; Tanaka, S.; Itakura, K.; Dickerson, R.E. The Molecular Structure of d (ICpCpGpG), a Fragment of Right-Handed Double Helical A-DNA. Nature 1982, 295, 294–299. [Google Scholar] [CrossRef]
  4. Frank-Kamenetskii, M.D.; Mirkin, S.M. Triplex DNA Structures. Annu. Rev. Biochem. 1995, 64, 65–95. [Google Scholar] [CrossRef] [PubMed]
  5. Sundquist, W.I.; Klug, A. Telomeric DNA Dimerizes by Formation of Guanine Tetrads between Hairpin Loops. Nature 1989, 342, 825–829. [Google Scholar] [CrossRef]
  6. Kim, M.-Y.; Gleason-Guzman, M.; Izbicka, E.; Nishioka, D.; Hurley, L.H. The Different Biological Effects of Telomestatin and TMPyP4 Can Be Attributed to Their Selectivity for Interaction with Intramolecular or Intermolecular G-Quadruplex Structures. Cancer Res. 2003, 63, 3247–3256. [Google Scholar] [PubMed]
  7. James, P.L.; Brown, T.; Fox, K.R. Thermodynamic and Kinetic Stability of Intermolecular Triple Helices Containing Different Proportions of C+·GC and T·AT Triplets. Nucleic Acids Res. 2003, 31, 5598–5606. [Google Scholar] [CrossRef]
  8. Waters, J.T.; Lu, X.-J.; Galindo-Murillo, R.; Gumbart, J.C.; Kim, H.D.; Cheatham, T.E.I.; Harvey, S.C. Transitions of Double-Stranded DNA between the A- and B-Forms. J. Phys. Chem. B 2016, 120, 8449–8456. [Google Scholar] [CrossRef]
  9. Tang, Q. Z-Nucleic Acids: Uncovering the Functions from Past to Present. Eur. J. Immunol. 2022, 52, 1700–1711. [Google Scholar] [CrossRef]
  10. Holder, I.T.; Wagner, S.; Xiong, P.; Sinn, M.; Frickey, T.; Meyer, A.; Hartig, J.S. Intrastrand Triplex DNA Repeats in Bacteria: A Source of Genomic Instability. Nucleic Acids Res. 2015, 43, 10126–10142. [Google Scholar] [CrossRef]
  11. Paugh, S.W.; Coss, D.R.; Bao, J.; Laudermilk, L.T.; Grace, C.R.; Ferreira, A.M.; Waddell, M.B.; Ridout, G.; Naeve, D.; Leuze, M.; et al. MicroRNAs Form Triplexes with Double Stranded DNA at Sequence-Specific Binding Sites; a Eukaryotic Mechanism via Which microRNAs Could Directly Alter Gene Expression. PLoS Comput. Biol. 2016, 12, e1004744. [Google Scholar] [CrossRef] [PubMed]
  12. Li, Y.; Syed, J.; Sugiyama, H. RNA-DNA Triplex Formation by Long Noncoding RNAs. Cell Chem. Biol. 2016, 23, 1325–1333. [Google Scholar] [CrossRef] [PubMed]
  13. Sengupta, P.; Bose, D.; Chatterjee, S. The Molecular Tête-à-Tête between G-Quadruplexes and the i-Motif in the Human Genome. ChemBioChem 2021, 22, 1517–1537. [Google Scholar] [CrossRef] [PubMed]
  14. Sollier, J.; Cimprich, K.A. Breaking Bad: R-Loops and Genome Integrity. Trends Cell Biol. 2015, 25, 514–522. [Google Scholar] [CrossRef] [PubMed]
  15. Inagaki, H.; Ohye, T.; Kogo, H.; Tsutsumi, M.; Kato, T.; Tong, M.; Emanuel, B.S.; Kurahashi, H. Two Sequential Cleavage Reactions on Cruciform DNA Structures Cause Palindrome-Mediated Chromosomal Translocations. Nat. Commun. 2013, 4, 1592. [Google Scholar] [CrossRef] [PubMed]
  16. Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Couch, G.S.; Greenblatt, D.M.; Meng, E.C.; Ferrin, T.E. UCSF Chimera—A Visualization System for Exploratory Research and Analysis. J. Comput. Chem. 2004, 25, 1605–1612. [Google Scholar] [CrossRef] [PubMed]
  17. Jovin, T.M. The Origin of Left-Handed Poly[d(G-C)]. In Z-DNA: Methods and Protocols; Springer: Berlin/Heidelberg, Germany, 2023; ISBN 978-1-07-163084-6. [Google Scholar]
  18. Herbert, A. Z-DNA and Z-RNA in Human Disease. Commun. Biol. 2019, 2, 7. [Google Scholar] [CrossRef] [PubMed]
  19. Chiang, D.C.; Li, Y.; Ng, S.K. The Role of the Z-DNA Binding Domain in Innate Immunity and Stress Granules. Front. Immunol. 2021, 11, 3779. [Google Scholar] [CrossRef]
  20. Herbert, A. Z-DNA and Z-RNA: Methods—Past and Future. In Z-DNA: Methods and Protocols; Springer: Berlin/Heidelberg, Germany, 2023; pp. 295–329. ISBN 978-1-07-163084-6. [Google Scholar]
  21. Song, Q.; Fan, Y.; Zhang, H.; Wang, N. Z-DNA Binding Protein 1 Orchestrates Innate Immunity and Inflammatory Cell Death. Cytokine Growth Factor Rev. 2024. [Google Scholar] [CrossRef]
  22. Sun, L.; Miao, Y.; Wang, Z.; Chen, H.; Dong, P.; Zhang, H.; Wu, L.; Jiang, M.; Chen, L.; Yang, W.; et al. Structural Insight into African Swine Fever Virus I73R Protein Reveals It as a Z-DNA Binding Protein. Transbound. Emerg. Dis. 2022, 69, e1923–e1935. [Google Scholar] [CrossRef]
  23. Kim, C. How Z-DNA/RNA Binding Proteins Shape Homeostasis, Inflammation, and Immunity. BMB Rep. 2020, 53, 453–457. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, J.; Fakharzadeh, A.; Pan, F.; Roland, C.; Sagui, C. Atypical Structures of GAA/TTC Trinucleotide Repeats Underlying Friedreich’s Ataxia: DNA Triplexes and RNA/DNA Hybrids. Nucleic Acids Res. 2020, 48, 9899–9917. [Google Scholar] [CrossRef] [PubMed]
  25. Poggi, L.; Richard, G.-F. Alternative DNA Structures In Vivo: Molecular Evidence and Remaining Questions. MMBR 2020, 85, e00110-20. [Google Scholar] [CrossRef] [PubMed]
  26. San Martin-Alonso, M.; Soler-Oliva, M.E.; García-Rubio, M.; García-Muse, T.; Aguilera, A. Harmful R-Loops Are Prevented via Different Cell Cycle-Specific Mechanisms. Nat. Commun. 2021, 12, 4451. [Google Scholar] [CrossRef] [PubMed]
  27. Ngo, G.H.P.; Grimstead, J.W.; Baird, D.M. UPF1 Promotes the Formation of R Loops to Stimulate DNA Double-Strand Break Repair. Nat. Commun. 2021, 12, 3849. [Google Scholar] [CrossRef] [PubMed]
  28. Mackay, R.P.; Xu, Q.; Weinberger, P.M. R-Loop Physiology and Pathology: A Brief Review. DNA Cell Biol. 2020, 39, 1914–1925. [Google Scholar] [CrossRef] [PubMed]
  29. Prorok, P.; Artufel, M.; Aze, A.; Coulombe, P.; Peiffer, I.; Lacroix, L.; Guédin, A.; Mergny, J.-L.; Damaschke, J.; Schepers, A.; et al. Involvement of G-Quadruplex Regions in Mammalian Replication Origin Activity. Nat. Commun. 2019, 10, 3274. [Google Scholar] [CrossRef] [PubMed]
  30. Esain-Garcia, I.; Kirchner, A.; Melidis, L.; Tavares, R.d.C.A.; Dhir, S.; Simeone, A.; Yu, Z.; Madden, S.K.; Hermann, R.; Tannahill, D.; et al. G-Quadruplex DNA Structure Is a Positive Regulator of MYC Transcription. Proc. Natl. Acad. Sci. USA 2024, 121, e2320240121. [Google Scholar] [CrossRef] [PubMed]
  31. Maltby, C.J.; Schofield, J.P.R.; Houghton, S.D.; O’Kelly, I.; Vargas-Caballero, M.; Deinhardt, K.; Coldwell, M.J. A 5′ UTR GGN Repeat Controls Localisation and Translation of a Potassium Leak Channel mRNA through G-Quadruplex Formation. Nucleic Acids Res. 2020, 48, 9822–9839. [Google Scholar] [CrossRef]
  32. Mei, Y.; Deng, Z.; Vladimirova, O.; Gulve, N.; Johnson, F.B.; Drosopoulos, W.C.; Schildkraut, C.L.; Lieberman, P.M. TERRA G-Quadruplex RNA Interaction with TRF2 GAR Domain Is Required for Telomere Integrity. Sci. Rep. 2021, 11, 3509. [Google Scholar] [CrossRef]
  33. Tassinari, M.; Richter, S.N.; Gandellini, P. Biological Relevance and Therapeutic Potential of G-Quadruplex Structures in the Human Noncoding Transcriptome. Nucleic Acids Res. 2021, 49, 3617–3633. [Google Scholar] [CrossRef]
  34. Kharel, P.; Balaratnam, S.; Beals, N.; Basu, S. The Role of RNA G-Quadruplexes in Human Diseases and Therapeutic Strategies. Wiley Interdiscip. Rev. RNA 2020, 11, e1568. [Google Scholar] [CrossRef] [PubMed]
  35. Zyner, K.G.; Simeone, A.; Flynn, S.M.; Doyle, C.; Marsico, G.; Adhikari, S.; Portella, G.; Tannahill, D.; Balasubramanian, S. G-Quadruplex DNA Structures in Human Stem Cells and Differentiation. Nat. Commun. 2022, 13, 142. [Google Scholar] [CrossRef] [PubMed]
  36. Zanin, I.; Ruggiero, E.; Nicoletto, G.; Lago, S.; Maurizio, I.; Gallina, I.; Richter, S.N. Genome-Wide Mapping of i-Motifs Reveals Their Association with Transcription Regulation in Live Human Cells. Nucleic Acids Res. 2023, 51, 8309–8321. [Google Scholar] [CrossRef] [PubMed]
  37. Wang, B. The RNA I-Motif in the Primordial RNA World. Orig. Life Evol. Biosph. 2019, 49, 105–109. [Google Scholar] [CrossRef] [PubMed]
  38. Brázda, V.; Laister, R.C.; Jagelská, E.B.; Arrowsmith, C. Cruciform Structures Are a Common DNA Feature Important for Regulating Biological Processes. BMC Mol. Biol. 2011, 12, 33. [Google Scholar] [CrossRef] [PubMed]
  39. Fleming, A.M.; Zhu, J.; Jara-Espejo, M.; Burrows, C.J. Cruciform DNA Sequences in Gene Promoters Can Impact Transcription upon Oxidative Modification of 2′-Deoxyguanosine. Biochemistry 2020, 59, 2616–2626. [Google Scholar] [CrossRef]
  40. Mengoli, V.; Ceppi, I.; Sanchez, A.; Cannavo, E.; Halder, S.; Scaglione, S.; Gaillard, P.; McHugh, P.J.; Riesen, N.; Pettazzoni, P.; et al. WRN Helicase and Mismatch Repair Complexes Independently and Synergistically Disrupt Cruciform DNA Structures. EMBO J. 2023, 42, e111998. [Google Scholar] [CrossRef]
  41. Goswami, P.; Bartas, M.; Lexa, M.; Bohálová, N.; Volná, A.; Červeň, J.; Červeňová, V.; Pečinka, P.; Špunda, V.; Fojta, M. SARS-CoV-2 Hot-Spot Mutations Are Significantly Enriched within Inverted Repeats and CpG Island Loci. Brief. Bioinform. 2021, 22, 1338–1345. [Google Scholar] [CrossRef]
  42. Bartas, M.; Goswami, P.; Lexa, M.; Červeň, J.; Volná, A.; Fojta, M.; Brázda, V.; Pečinka, P. Letter to the Editor: Significant Mutation Enrichment in Inverted Repeat Sites of New SARS-CoV-2 Strains. Brief. Bioinform. 2021, 22, bbab129. [Google Scholar] [CrossRef]
  43. Chaput, J.C.; Switzer, C. A DNA Pentaplex Incorporating Nucleobase Quintets. Proc. Natl. Acad. Sci. USA 1999, 96, 10614–10619. [Google Scholar] [CrossRef] [PubMed]
  44. Makova, K.D.; Weissensteiner, M.H. Noncanonical DNA Structures Are Drivers of Genome Evolution. Trends Genet. 2023, 39, 109–124. [Google Scholar] [CrossRef] [PubMed]
  45. Brázda, V.; Kolomazník, J.; Lỳsek, J.; Bartas, M.; Fojta, M.; Št’astnỳ, J.; Mergny, J.-L. G4Hunter Web Application: A Web Server for G-Quadruplex Prediction. Bioinformatics 2019, 35, 3493–3495. [Google Scholar] [CrossRef]
  46. Brázda, V.; Kolomazník, J.; Lýsek, J.; Hároníková, L.; Coufal, J.; Št’astný, J. Palindrome Analyser—A New Web-Based Server for Predicting and Evaluating Inverted Repeats in Nucleotide Sequences. Biochem. Biophys. Res. Commun. 2016, 478, 1739–1745. [Google Scholar] [CrossRef]
  47. Brázda, V.; Havlík, J.; Kolomazník, J.; Trenz, O.; Št’astnỳ, J. R-Loop Tracker: Web Access-Based Tool for R-Loop Detection and Analysis in Genomic DNA Sequences. Int. J. Mol. Sci. 2021, 22, 12857. [Google Scholar] [CrossRef] [PubMed]
  48. Hennecker, C.; Yamout, L.; Zhang, C.; Zhao, C.; Hiraki, D.; Moitessier, N.; Mittermaier, A. Structural Polymorphism of Guanine Quadruplex-Containing Regions in Human Promoters. Int. J. Mol. Sci. 2022, 23, 16020. [Google Scholar] [CrossRef]
  49. Singh, D.; Desai, N.; Shah, V.; Datta, B. In Silico Identification of Potential Quadruplex Forming Sequences in LncRNAs of Cervical Cancer. Int. J. Mol. Sci. 2023, 24, 12658. [Google Scholar] [CrossRef] [PubMed]
  50. Nicoletto, G.; Richter, S.N.; Frasson, I. Presence, Location and Conservation of Putative G-Quadruplex Forming Sequences in Arboviruses Infecting Humans. Int. J. Mol. Sci. 2023, 24, 9523. [Google Scholar] [CrossRef]
  51. Gould, E.; Pettersson, J.; Higgs, S.; Charrel, R.; de Lamballerie, X. Emerging Arboviruses: Why Today? One Health 2017, 4, 1–13. [Google Scholar] [CrossRef]
  52. Gumina, J.M.; Richardson, A.E.; Shojiv, M.H.; Chambers, A.E.; Sandwith, S.N.; Reisinger, M.A.; Karns, T.J.; Osborne, T.L.; Alashi, H.N.; Anderson, Q.T.; et al. Differential Gene Expression Following DHX36/G4R1 Knockout Is Associated with G-Quadruplex Content and Cancer. Int. J. Mol. Sci. 2024, 25, 1753. [Google Scholar] [CrossRef]
  53. Feng, Y.; Luo, Z.; Huang, R.; Yang, X.; Cheng, X.; Zhang, W. Epigenomic Features and Potential Functions of K+ and Na+ Favorable DNA G-Quadruplexes in Rice. Int. J. Mol. Sci. 2022, 23, 8404. [Google Scholar] [CrossRef] [PubMed]
  54. Shavkunov, K.S.; Markelova, N.Y.; Glazunova, O.A.; Kolzhetsov, N.P.; Panyukov, V.V.; Ozoline, O.N. The Fate and Functionality of Alien tRNA Fragments in Culturing Medium and Cells of Escherichia Coli. Int. J. Mol. Sci. 2023, 24, 12960. [Google Scholar] [CrossRef]
  55. Zulfiqar, S.; Farooq, M.A.; Zhao, T.; Wang, P.; Tabusam, J.; Wang, Y.; Xuan, S.; Zhao, J.; Chen, X.; Shen, S.; et al. Virus-Induced Gene Silencing (VIGS): A Powerful Tool for Crop Improvement and Its Advancement towards Epigenetics. Int. J. Mol. Sci. 2023, 24, 5608. [Google Scholar] [CrossRef] [PubMed]
  56. Zhong, H.-S.; Dong, M.-J.; Gao, F. G4Bank: A Database of Experimentally Identified DNA G-Quadruplex Sequences. Interdiscip. Sci. Comput. Life. Sci. 2023, 15, 515–523. [Google Scholar] [CrossRef]
  57. Elimelech-Zohar, K.; Orenstein, Y. An Overview on Nucleic-Acid G-Quadruplex Prediction: From Rule-Based Methods to Deep Neural Networks. Brief. Bioinform. 2023, 24, bbad252. [Google Scholar] [CrossRef] [PubMed]
  58. Cer, R.Z.; Donohue, D.E.; Mudunuri, U.S.; Temiz, N.A.; Loss, M.A.; Starner, N.J.; Halusa, G.N.; Volfovsky, N.; Yi, M.; Luke, B.T. Non-B DB v2. 0: A Database of Predicted Non-B DNA-Forming Motifs and Its Associated Tools. Nucleic Acids Res. 2012, 41, D94–D100. [Google Scholar] [CrossRef]
  59. Luo, Y.; Granzhan, A.; Marquevielle, J.; Cucchiarini, A.; Lacroix, L.; Amrane, S.; Verga, D.; Mergny, J.-L. Guidelines for G-Quadruplexes: I. In Vitro Characterization. Biochimie 2023, 214, 5–23. [Google Scholar] [CrossRef] [PubMed]
  60. Xu, Q.; Kowalski, J. NBBC: A Non-B DNA Burden Explorer in Cancer. Nucleic Acids Res. 2023, 51, W357–W364. [Google Scholar] [CrossRef] [PubMed]
  61. Matos-Rodrigues, G.; Hisey, J.A.; Nussenzweig, A.; Mirkin, S.M. Detection of Alternative DNA Structures and Its Implications for Human Disease. Mol. Cell 2023, 83, 3622–3641. [Google Scholar] [CrossRef]
  62. Frappier, L.; Price, G.B.; Martin, R.G.; Zannis-Hadjopoulos, M. Monoclonal Antibodies to Cruciform DNA Structures. J. Mol. Biol. 1987, 193, 751–758. [Google Scholar] [CrossRef]
  63. Feng, X.; Xie, F.-Y.; Ou, X.-H.; Ma, J.-Y. Cruciform DNA in Mouse Growing Oocytes: Its Dynamics and Its Relationship with DNA Transcription. PLoS ONE 2020, 15, e0240844. [Google Scholar] [CrossRef] [PubMed]
  64. Yin, C.; Zhang, T.; Balachandran, S. Detecting Z-RNA and Z-DNA in Mammalian Cells. In Z-DNA; Kim, K.K., Subramani, V.K., Eds.; Methods in Molecular Biology; Springer: New York, NY, USA, 2023; Volume 2651, pp. 277–284. ISBN 978-1-07-163083-9. [Google Scholar]
  65. Javadekar, S.M.; Nilavar, N.M.; Paranjape, A.; Das, K.; Raghavan, S.C. Characterization of G-Quadruplex Antibody Reveals Differential Specificity for G4 DNA Forms. DNA Res. 2020, 27, dsaa024. [Google Scholar] [CrossRef] [PubMed]
  66. Zeraati, M.; Langley, D.B.; Schofield, P.; Moye, A.L.; Rouet, R.; Hughes, W.E.; Bryan, T.M.; Dinger, M.E.; Christ, D. I-Motif DNA Structures Are Formed in the Nuclei of Human Cells. Nat. Chem. 2018, 10, 631–637. [Google Scholar] [CrossRef] [PubMed]
  67. Weissensteiner, M.H.; Cremona, M.A.; Guiblet, W.M.; Stoler, N.; Harris, R.S.; Cechova, M.; Eckert, K.A.; Chiaromonte, F.; Huang, Y.-F.; Makova, K.D. Accurate Sequencing of DNA Motifs Able to Form Alternative (Non-B) Structures. Genome Res. 2023, 33, 907–922. [Google Scholar] [CrossRef] [PubMed]
  68. Nurk, S.; Koren, S.; Rhie, A.; Rautiainen, M.; Bzikadze, A.V.; Mikheenko, A.; Vollger, M.R.; Altemose, N.; Uralsky, L.; Gershman, A.; et al. The Complete Sequence of a Human Genome. Science 2022, 376, 44–53. [Google Scholar] [CrossRef] [PubMed]
  69. Huang, H.-R.; Liu, X.; Arshad, R.; Wang, X.; Li, W.-M.; Zhou, Y.; Ge, X.-J. Telomere-to-Telomere Haplotype-Resolved Reference Genome Reveals Subgenome Divergence and Disease Resistance in Triploid Cavendish Banana. Hortic. Res. 2023, 10, uhad153. [Google Scholar] [CrossRef] [PubMed]
  70. Chen, J.; Wang, Z.; Tan, K.; Huang, W.; Shi, J.; Li, T.; Hu, J.; Wang, K.; Wang, C.; Xin, B. A Complete Telomere-to-Telomere Assembly of the Maize Genome. Nat. Genet. 2023, 55, 1221–1231. [Google Scholar] [CrossRef] [PubMed]
  71. Koren, S.; Bao, Z.; Guarracino, A.; Ou, S.; Goodwin, S.; Jenike, K.M.; Lucas, J.; McNulty, B.; Park, J.; Rautiainen, M. Gapless Assembly of Complete Human and Plant Chromosomes Using Only Nanopore Sequencing. bioRxiv 2024, 2024-03. [Google Scholar]
  72. Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Žídek, A.; Potapenko, A.; et al. Highly Accurate Protein Structure Prediction with AlphaFold. Nature 2021, 596, 583–589. [Google Scholar] [CrossRef]
  73. Du, Z.; Su, H.; Wang, W.; Ye, L.; Wei, H.; Peng, Z.; Anishchenko, I.; Baker, D.; Yang, J. The trRosetta Server for Fast and Accurate Protein Structure Prediction. Nat. Protoc. 2021, 16, 5634–5651. [Google Scholar] [CrossRef]
  74. Mirdita, M.; Schütze, K.; Moriwaki, Y.; Heo, L.; Ovchinnikov, S.; Steinegger, M. ColabFold: Making Protein Folding Accessible to All. Nat. Methods 2022, 19, 679–682. [Google Scholar] [CrossRef] [PubMed]
  75. Patro, L.P.P.; Kumar, A.; Kolimi, N.; Rathinavelan, T. 3D-NuS: A Web Server for Automated Modeling and Visualization of Non-Canonical 3-Dimensional Nucleic Acid Structures. J. Mol. Biol. 2017, 429, 2438–2448. [Google Scholar] [CrossRef]
  76. Li, S.; Olson, W.K.; Lu, X.-J. Web 3DNA 2.0 for the Analysis, Visualization, and Modeling of 3D Nucleic Acid Structures. Nucleic Acids Res. 2019, 47, W26–W34. [Google Scholar] [CrossRef] [PubMed]
  77. Mellor, C.; Perez, C.; Sale, J.E. Creation and Resolution of Non-B-DNA Structural Impediments during Replication. Crit. Rev. Biochem. Mol. 2022, 57, 412–442. [Google Scholar] [CrossRef]
  78. Herbert, A. ALU Non-B-DNA Conformations, Flipons, Binary Codes and Evolution. R. Soc. Open Sci. 2020, 7, 200222. [Google Scholar] [CrossRef]
  79. Wright, E.P.; Abdelhamid, M.A.S.; Ehiabor, M.O.; Grigg, M.C.; Irving, K.; Smith, N.M.; Waller, Z.A.E. Epigenetic Modification of Cytosines Fine Tunes the Stability of I-Motif DNA. Nucleic Acids Res. 2020, 48, 55–62. [Google Scholar] [CrossRef]
  80. Sugimoto, N.; Endoh, T.; Takahashi, S.; Tateishi-Karimata, H. Chemical Biology of Double Helical and Non-Double Helical Nucleic Acids: “To B or Not To B, That Is the Question”. Bull. Chem. Soc. Jpn. 2021, 94, 1970–1998. [Google Scholar] [CrossRef]
  81. Matsumoto, S.; Sugimoto, N. New Insights into the Functions of Nucleic Acids Controlled by Cellular Microenvironments. Top. Curr. Chem. 2021, 379, 17. [Google Scholar] [CrossRef] [PubMed]
  82. Sharma, S. Non-B DNA Secondary Structures and Their Resolution by RecQ Helicases. J. Nucleic Acids 2011, 2011, e724215. [Google Scholar] [CrossRef]
  83. Rocca, R.; Moraca, F.; Costa, G.; Nadai, M.; Scalabrin, M.; Talarico, C.; Distinto, S.; Maccioni, E.; Ortuso, F.; Artese, A.; et al. Identification of G-Quadruplex DNA/RNA Binders: Structure-Based Virtual Screening and Biophysical Characterization. Biochim. Biophys. Acta–Gen. Subj. 2017, 1861, 1329–1340. [Google Scholar] [CrossRef]
  84. Pina, A.F.; Sousa, S.F.; Azevedo, L.; Carneiro, J. Non-B DNA Conformations Analysis through Molecular Dynamics Simulations. Biochim. Biophys. Acta–Gen. Subj. 2022, 1866, 130252. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, L.; Ge, X.; Liu, L.; Hu, G. Code Interpreter for Bioinformatics: Are We There Yet? Ann. Biomed. Eng. 2024, 52, 754–756. [Google Scholar] [CrossRef] [PubMed]
  86. Hilton, J.; Gelmon, K.; Bedard, P.L.; Tu, D.; Xu, H.; Tinker, A.V.; Goodwin, R.; Laurie, S.A.; Jonker, D.; Hansen, A.R.; et al. Results of the Phase I CCTG IND.231 Trial of CX-5461 in Patients with Advanced Solid Tumors Enriched for DNA-Repair Deficiencies. Nat. Commun. 2022, 13, 3607. [Google Scholar] [CrossRef]
  87. Drygin, D.; Siddiqui-Jain, A.; O’Brien, S.; Schwaebe, M.; Lin, A.; Bliesath, J.; Ho, C.B.; Proffitt, C.; Trent, K.; Whitten, J.P.; et al. Anticancer Activity of CX-3543: A Direct Inhibitor of rRNA Biogenesis. Cancer Res. 2009, 69, 7653–7661. [Google Scholar] [CrossRef]
  88. Du, Y.; Zhou, X. Targeting Non-B-Form DNA in Living Cells. Chem. Rec. 2013, 13, 371–384. [Google Scholar] [CrossRef] [PubMed]
  89. Kreig, A.; Calvert, J.; Sanoica, J.; Cullum, E.; Tipanna, R.; Myong, S. G-Quadruplex Formation in Double Strand DNA Probed by NMM and CV Fluorescence. Nucleic Acids Res. 2015, 43, 7961–7970. [Google Scholar] [CrossRef] [PubMed]
  90. Šket, P.; Pohleven, J.; Kovanda, A.; Štalekar, M.; Župunski, V.; Zalar, M.; Plavec, J.; Rogelj, B. Characterization of DNA G-Quadruplex Species Forming from C9ORF72 G4C2-Expanded Repeats Associated with Amyotrophic Lateral Sclerosis and Frontotemporal Lobar Degeneration. Neurobiol. Aging 2015, 36, 1091–1096. [Google Scholar] [CrossRef]
  91. Grishchenko, I.V.; Purvinsh, Y.V.; Yudkin, D.V. Mystery of Expansion: DNA Metabolism and Unstable Repeats. In Mechanisms of Genome Protection and Repair; Zharkov, D.O., Ed.; Springer International Publishing: Cham, Switzerland, 2020; pp. 101–124. ISBN 978-3-030-41283-8. [Google Scholar]
Table 1. The selection of the most-researched UNas together with their basic characteristics, formation sequences, and known molecular-biological functions. Schematic structures were visualized using UCSF Chimera [16] based on experimentally solved structures in the case of A-DNA (PDB: 2RMQ), Z-DNA (PDB: 4OCB), and triplexes (PDB: 149D). For the remaining and more structurally complicated UNas, simplified diagrams were made using BioRender.
Table 1. The selection of the most-researched UNas together with their basic characteristics, formation sequences, and known molecular-biological functions. Schematic structures were visualized using UCSF Chimera [16] based on experimentally solved structures in the case of A-DNA (PDB: 2RMQ), Z-DNA (PDB: 4OCB), and triplexes (PDB: 149D). For the remaining and more structurally complicated UNas, simplified diagrams were made using BioRender.
UNasSchematic StructureBasic Characteristics and Typical UNas-Forming SequencesMolecular-Biological Function
A-DNA
(A-RNA)
Ijms 25 05226 i001
  • Short and wide helix
  • Rigid, low flexibility
  • No typical formation sequence
  • Can arise under dehydrating conditions (e.g., desiccation of bacteria)
Most of double-stranded RNA is believed to exist in the A-form [17]. In addition, simple prokaryotic organisms and viruses can utilize this conformation to withstand adverse conditions [2,3].
Z-DNA
(Z-RNA)
Ijms 25 05226 i002
  • Left-handed nucleic acid
  • Zig-zag phosphodiester backbone
  • Alternation of purine and pyrimidine, typically d(CG)n or d(TG)n
  • In vitro, it usually requires a high concentration of salts to be formed from B-DNA (4M NaCl)
Z-DNA and Z-RNA are associated with several human diseases, including various cancers and the autoimmune disease Aicardi–Goutières syndrome [18]. These UNas are also believed to play crucial roles in innate immunity and host–virus interactions [19]. Proteins preferentially recognizing left-handed nucleic acids are known [20,21,22,23].
TriplexesIjms 25 05226 i003
  • Triple-helical structure
  • Can arise in sites of long homopurine and homopyridine tracts
  • Prefers acidic or neutral pH and divalent cations
Sites of triplex-forming sequences are connected with microsatellite repeat expansion disorders, including Friedreich’s ataxia [24]. Although the in vivo formation of DNA triplexes has yet to be directly proved, it is believed to drive genomic instability [25].
R-loopsIjms 25 05226 i004
  • Three-stranded structure
  • Consists of a DNA–RNA hybrid and a displaced strand of DNA
  • Arises mainly in guanine-rich clusters
R-loops are formed during transcription when the nascent RNA strand base pairs with template DNA. They may cause genomic instability via the blocking of DNA replication [26]. R-loops are involved in the repair of double-stranded DNA breaks [27] and related to several human diseases [28].
G-quadruplexesIjms 25 05226 i005
  • Guanines form planar G-quartets; these stack on each other
  • Stabilized by monovalent cations (mostly K+ and Na+)
  • Guanine-rich sequences (tracts) are usually separated by short loop-forming sequences
G-quadruplexes play vital roles in the replication [29], transcription [30], translation [31], telomere maintenance [32], and biogenesis of noncoding RNAs [33]. They are connected to many human diseases [34] as well as important physiological developmental processes [35].
i-motifsIjms 25 05226 i006
  • Formed by intercalated cytosine base pairs in slightly acidic conditions
  • Cytosine-rich sequences, analogous to G-quadruplexes
i-motifs likely participate in transcription regulation [36]. In DNA, they can can arise in the opposite strand to the G-quadruplex [13]. There is also a hypothesis that i-motifs played an important role in the primordial RNA world [37].
CruciformsIjms 25 05226 i007
  • Structures consisting of a stem, branch point, and loop in the shape of a cruciform
  • Can arise from inverted repeat sequences
  • Stabilized by negative supercoiling
Cruciforms may play an important role in various biological processes, including replication [38], gene expression [39], recombination [38], and repair [40]. Inverted repeats that form cruciforms or hairpins are essential features of viral genomes, potentially driving their mutability [41,42].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bartas, M.; Brázda, V.; Pečinka, P. Special Issue “Bioinformatics of Unusual DNA and RNA Structures”. Int. J. Mol. Sci. 2024, 25, 5226. https://doi.org/10.3390/ijms25105226

AMA Style

Bartas M, Brázda V, Pečinka P. Special Issue “Bioinformatics of Unusual DNA and RNA Structures”. International Journal of Molecular Sciences. 2024; 25(10):5226. https://doi.org/10.3390/ijms25105226

Chicago/Turabian Style

Bartas, Martin, Václav Brázda, and Petr Pečinka. 2024. "Special Issue “Bioinformatics of Unusual DNA and RNA Structures”" International Journal of Molecular Sciences 25, no. 10: 5226. https://doi.org/10.3390/ijms25105226

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop