Glen Report 33-21: L-DNA: Applications and the Recognition of Mirror Image Nucleic Acids

Author: Jonathan Sczepanski
Department of Chemistry
Texas A&M University
College Station, TX

 

When you think about DNA, do you consider its stereochemistry? You should! DNA is a chiral molecule, and consequently, has non-superimposable mirror images (enantiomers). The source of chirality in DNA is the deoxyribose sugar, which contains three chirality centers (Figure 1).  On one “hand”, native DNA found in all living organisms consists exclusively of D-deoxyribose sugars (D-DNA) and forms right-handed helices. On the other hand, its enantiomer, L-DNA, consists of L-deoxyribose sugars and forms left-handed helices. L-DNA is not found in nature, but can be prepared synthetically in the laboratory, with the earliest syntheses of L-oligonucleotides dating back to the 1970s.1 As enantiomers, D-DNA and L-DNA are physically indistinguishable, except for their opposed optical activity, making them identical from a design perspective (i.e. same hybridization rates, duplex thermal stability, etc.). Importantly, L-DNA is orthogonal to the stereospecific environment of native biology, which has evolved to recognize D-nucleic acids. As a result, L-DNA is highly resistant to nuclease degradation and off-target interactions with other cellular macromolecules.2-3 These favorable properties, along with the recent commercialization of L-DNA phosphoramidite building blocks, has catalyzed the development of many promising L-DNA based biotechnologies.4Figure 1

 

Figure 1. The two enantiomers of DNA. Chirality centers are indicated with an asterisk.

One of the first5-6, and still common, uses of L-DNA is in the form of aptamers. To date, L-DNA aptamers (also called Spiegelmers) have been evolved to bind a variety of targets, including small molecules, peptides, and proteins.7 Our laboratory has recently pioneered the use of L-DNA aptamers to bind native D-RNA structures.8 When applied in vivo, Spiegelmers retain a high affinity for their targets, while being nontoxic and have very low immunogenic potential.9 Given these desirable properties, it is not surprising that several Speigelmer therapeutics are in various stages of clinical development.10

Another powerful application of L-DNA is in the construction of bio-orthogonal molecular sensors. For example, L-DNA-based molecular beacons, which are not substrates for native polymerases, can be used for optical monitoring of PCR reactions, a method referred to as “adaptive PCR”.11 L-DNA molecular beacon probes have also been employed for measuring intracellular temperatures.12 DNA-based sensors for achiral analytes can be mirrored without a loss in activity. This property was exploited to generate nuclease-resistant L-DNAzyme sensors for monitoring metal ion concentrations in living cells.13-14 In the case of chiral analytes, use of both enantiomers of the corresponding nucleic acid sensor enables rapid screening of enantiomeric purity.15

Watson–Crick base pairing between complementary strands is stereospecific, and thus, L-DNA is incapable of hybridizing to native DNA and RNA.3, 16 While this property excludes the direct use of L-DNA as antisense agents, it ensures that sensors and other biotechnologies constructed from L-DNA have minimal off-target hybridization, an important consideration for analytical applications. Indeed, one of the earliest applications of L-DNA was the development of a universal microarray platform that employed L-DNA capture strands as a means to reduce cross-hybridization of nucleic acid samples with different regions of the array.3 The stereospecificity of hybridization can also be exploited to create internal controls during sample analysis.17 

Although D-DNA and L-DNA do not interact directly, sequence information can still be transferred between the two enantiomers via strand-displacement strategies.18-20 For example, our laboratory showed that DNA enantiomers can be sequence-specifically interfaced via toehold-mediated strand-displacement from peptide nucleic acid (PNA), a process referred to as “heterochiral” strand displacement (Figure 2).18 Because PNA is achiral, and hybridizes equally well to both D-DNA and L-DNA, it functions as the intermediary, allowing a strand of D-DNA to displace a strand of L-DNA (and vice versa) in a sequence-specific manner. In principle, heterochiral strand displacement allows for any D-nucleic acid target (DNA or RNA) to be interfaced with sensors and nanodevices composed of bioorthogonal L-DNA. For example, we used this approach to interface microRNAs with L-DNA-based logic circuits and catalytic amplifiers in vitro18, 21-22, and with an L-RNA-based fluorescent biosensor in living cells.23 Chimeric strands of both D-DNA and L-DNA also enable native nucleic acids to be interfaced with L-DNA, and have been employed for various biosensing purposes.24-25

 

Figure 2Figure 2. Schematic illustration of the heterochiral toehold-mediated strand displacement reaction. Nucleic acids are depicted as lines with half arrows denoting the 3 end and an asterisk indicating complementarity between sequence domains. The single-stranded toehold domain (t*) resides on the achiral PNA strand in the L-DNA/PNA heteroduplex, allowing a D-Input strand bind (via t and t*) and displace the incumbent L-Output strand.

 

It is clear from the examples discussed herein (and elsewhere4) that L-DNA provides a powerful opportunity to develop nucleic acid-based technologies having capabilities not possible using only the native stereoisomer. As more researchers step “Through the Looking-Glass”, and use of mirror image DNA becomes more routine, completely new and exciting applications will become possible.

References

  1. Tazawa, I.; Tazawa, S.; Stempel, L. M.; Ts'o, P. O. P., Conformation and interaction of dinucleoside mono- and diphosphates. 3. L-adenylyl-(3'-5')-L-adenosine and L-adenylyl-(2'-5')-L-adenosine. Biochemistry 1970, 9 (18), 3499-3514.
  2. Urata, H.; Ogura, E.; Shinohara, K.; Ueda, Y.; Akagi, M., Synthesis and properties of mirror-image DNA. Nucleic Acids Research 1992, 20 (13), 3325-3332.
  3. Hauser, N. C.; Martinez, R.; Jacob, A.; Rupp, S.; Hoheisel, J. D.; Matysiak, S., Utilising the left-helical conformation of L-DNA for analysing different marker types on a single universal microarray platform. Nucleic Acids Research 2006, 34 (18), 5101-5111.
  4. Young, B. E.; Kundu, N.; Sczepanski, J. T., Mirror-Image Oligonucleotides: History and Emerging Applications. Chemistry – A European Journal 2019, 25 (34), 7981-7990.
  5. Klussmann, S.; Nolte, A.; Bald, R.; Erdmann, V. A.; Fürste, J. P., Mirror-image RNA that binds D-adenosine. Nat. Biotechnol. 1996, 14, 1112-1115.
  6. Nolte, A.; Klussmann, S.; Bald, R.; Erdmann, V. A.; Furste, J. P., Mirror-design of L-oligonucleotide ligands binding to L-arginine. Nat. Biotechnol. 1996, 14, 1116-1119.
  7. Vater, A.; Klussmann, S., Turning mirror-image oligonucleotides into drugs: the evolution of Spiegelmer therapeutics. Drug Discov. Today 2015, 20, 147-155.
  8. Dey, S.; Sczepanski, J. T., In vitro selection of l-DNA aptamers that bind a structured d-RNA molecule. Nucleic Acids Research 2020, 48 (4), 1669-1680.
  9. Wlotzka, B.; Leva, S.; Eschgfaller, B.; Burmeister, J.; Kleinjung, F.; Kaduk, C.; Muhn, P.; Hess-Stumpp, H.; Klussmann, S., In vivo properties of an anti-GnRH spiegelmer: an example of an oligonucleotide-based therapeutic substance class. Proc. Natl. Acad. Sci. USA 2002, 99, 8898-8902.
  10. NOXXON Pharma, https://www.noxxon.com (Visited 9/9/2021).
  11. Adams, N. M.; Gabella, W. E.; Hardcastle, A. N.; Haselton, F. R., Adaptive PCR Based on Hybridization Sensing of Mirror-Image l-DNA. Analytical Chemistry 2017, 89 (1), 728-735.
  12. Ke, G.; Wang, C.; Ge, Y.; Zheng, N.; Zhu, Z.; Yang, C. J., l-DNA Molecular Beacon: A Safe, Stable, and Accurate Intracellular Nano-thermometer for Temperature Sensing in Living Cells. Journal of the American Chemical Society 2012, 134 (46), 18908-18911.
  13. Cui, L.; Peng, R.; Fu, T.; Zhang, X.; Wu, C.; Chen, H.; Liang, H.; Yang, C. J.; Tan, W., Biostable L-DNAzyme for sensing of metal ions in biological systems. Anal. Chem. 2016, 88, 1850-1855.
  14. Liang, H.; Xie, S.; Cui, L.; Wu, C.; Zhang, X., Designing a biostable L-DNAzyme for lead(ii) ion detection in practical samples. Analytical Methods 2016, 8 (39), 7260-7264.
  15. Feagin, T. A.; Olsen, D. P. V.; Headman, Z. C.; Heemstra, J. M., High-Throughput Enantiopurity Analysis Using Enantiomeric DNA-Based Sensors. Journal of the American Chemical Society 2015, 137 (12), 4198-4206.
  16. Hoehlig, K.; Bethge, L.; Klussmann, S., Stereospecificity of Oligonucleotide Interactions Revisited: No Evidence for Heterochiral Hybridization and Ribozyme/DNAzyme Activity. PLOS ONE 2015, 10 (2), e0115328.
  17. Zimmers, Z. A.; Adams, N. M.; Haselton, F. R., Addition of mirror-image L-DNA elements to DNA amplification circuits to distinguish leakage from target signal. Biosensors and Bioelectronics 2021, 188, 113354.
  18. Kabza, A. M.; Young, B. E.; Sczepanski, J. T., Heterochiral DNA Strand-Displacement Circuits. Journal of the American Chemical Society 2017, 139 (49), 17715-17718.
  19. Young, B. E.; Sczepanski, J. T., Heterochiral DNA Strand-Displacement Based on Chimeric d/l-Oligonucleotides. ACS Synthetic Biology 2019, 8 (12), 2756-2759.
  20. Mallette, T. L.; Stojanovic, M. N.; Stefanovic, D.; Lakin, M. R., Robust Heterochiral Strand Displacement Using Leakless Translators. ACS Synthetic Biology 2020, 9 (7), 1907-1910.
  21. Zhong, W.; Sczepanski, J. T., Direct Comparison of d-DNA and l-DNA Strand-Displacement Reactions in Living Mammalian Cells. ACS Synthetic Biology 2021, 10 (1), 209-212.
  22. Kabza, A. M.; Sczepanski, J. T., l-DNA-Based Catalytic Hairpin Assembly Circuit. Molecules 2020, 25, 947.
  23. Zhong, W.; Sczepanski, J. T., A Mirror Image Fluorogenic Aptamer Sensor for Live-Cell Imaging of MicroRNAs. ACS Sensors 2019, 4 (3), 566-570.
  24. Kim, Y.; Yang, C. J.; Tan, W., Superior structure stability and selectivity of hairpin nucleic acid probes with an l -DNA stem. Nucleic Acids Research 2007, 35 (21), 7279-7287.
  25. Ogata, M.; Hayashi, G.; Ichiu, A.; Okamoto, A., l-DNA-tagged fluorescence in situ hybridization for highly sensitive imaging of RNAs in single cells. Organic & Biomolecular Chemistry 2020, 18 (40), 8084-8088.

Product Information

beta-L-DNA Phosphoramidites