Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Comparing proteins and nucleic acids for next-generation biomolecular engineering

Abstract

Nanostructures built from biomolecules such as proteins, DNA and RNA are attracting attention in many areas of biological and materials sciences. Such nanoscale engineering was pioneered with proteins, yet the use of DNA is rapidly gaining traction. What are the advantages of the different biopolymers and which is best suited for a given molecular structure, function or application? In this Review, we evaluate the different structural properties of proteins and nucleic acids, as well as possible designs and synthetic routes for functional nanostructures. By comparing protein engineering and DNA nanotechnology, we highlight molecular architectures that are relevant in biotechnology, biomedicine and synthetic biology research, and identify emerging areas for research such as hybrid materials composed of protein and DNA/RNA.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Proteins, DNA and RNA.
Fig. 2: Natural and engineered proteins and DNA nanostructures used for biomolecular recognition.
Fig. 3: Engineered biocatalysts.
Fig. 4: Natural and engineered proteins that fulfil structural and cytoskeletal roles.
Fig. 5: Rationally designed DNA nanostructures.
Fig. 6: Natural and engineered hybrid systems that combine proteins, DNA and RNA.

Similar content being viewed by others

References

  1. Jeschek, M. et al. Directed evolution of artificial metalloenzymes for in vivo metathesis. Nature 537, 661–665 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Renata, H., Wang, Z. J. & Arnold, F. H. Expanding the enzyme universe: accessing non-natural reactions by mechanism-guided directed evolution. Angew. Chem. Int. Ed. 54, 3351–3367 (2015).

    Article  CAS  Google Scholar 

  3. Denard, C. A., Ren, H. & Zhao, H. Improving and repurposing biocatalysts via directed evolution. Curr. Opin. Chem. Biol. 25, 55–64 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Liszka, M. J., Clark, M. E., Schneider, E. & Clark, D. S. Nature versus nurture: developing enzymes that function under extreme conditions. Annu. Rev. Chem. Biomol. Eng. 3, 77–102 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Tiller, K. E. & Tessier, P. M. Advances in antibody design. Annu. Rev. Biomed. Eng. 17, 191–216 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Rosenbaum, L. Tragedy, perseverance, and chance — the story of CAR-T therapy. N. Engl. J. Med. 377, 1313–1315 (2017).

  7. Rodriguez, E. A. et al. The growing and glowing toolbox of fluorescent and photoactive proteins. Trends Biochem. Sci. 42, 111–129 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Martell, J. D. et al. A split horseradish peroxidase for the detection of intercellular protein-protein interactions and sensitive visualization of synapses. Nat. Biotechnol. 34, 774–780 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ellefson, J. W. et al. Synthetic evolutionary origin of a proofreading reverse transcriptase. Science 352, 1590–1593 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Seeman, N. C. Nanomaterials based on DNA. Annu. Rev. Biochem. 79, 65–87 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Rothemund, P. W. Folding DNA to create nanoscale shapes and patterns. Nature 440, 297–302 (2006).

    Article  CAS  PubMed  Google Scholar 

  12. Chen, Y. J., Groves, B., Muscat, R. A. & Seelig, G. DNA nanotechnology from the test tube to the cell. Nat. Nanotechnol. 10, 748–760 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Pinheiro, A. V., Han, D., Shih, W. M. & Yan, H. Challenges and opportunities for structural DNA nanotechnology. Nat. Nanotechnol. 6, 763–772 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Jones, M. R., Seeman, N. C. & Mirkin, C. A. Nanomaterials. Programmable materials and the nature of the DNA bond. Science 347, 1260901 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Seeman, N. C. & Sleiman, H. F. DNA nanotechnology. Nat. Rev. Mater. 3, 17068 (2017).

    Article  CAS  Google Scholar 

  16. Torring, T., Voigt, N. V., Nangreave, J., Yan, H. & Gothelf, K. V. DNA origami: a quantum leap for self-assembly of complex structures. Chem. Soc. Rev. 40, 5636–5646 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sacca, B. & Niemeyer, C. M. DNA origami: the art of folding DNA. Angew. Chem. Int. Ed. 51, 58–66 (2012).

    Article  CAS  Google Scholar 

  18. Hong, F., Zhang, F., Liu, Y. & Yan, H. DNA origami: scaffolds for creating higher order structures. Chem. Rev. 117, 12584–12640 (2017).

    Article  CAS  PubMed  Google Scholar 

  19. Ellington, A. D. & Szostak, J. W. In vitro selection of RNA molecules that bind specific ligands. Nature 346, 818–822 (1990).

    Article  CAS  PubMed  Google Scholar 

  20. Tuerk, C. & Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 249, 505–510 (1990).

    Article  CAS  PubMed  Google Scholar 

  21. Zhou, J. & Rossi, J. Aptamers as targeted therapeutics: current potential and challenges. Nat. Rev. Drug. Discov. 16, 181–202 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Mor-Vaknin, N. et al. DEK-targeting DNA aptamers as therapeutics for inflammatory arthritis. Nat. Commun. 8, 14252 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Nimjee, S. M., White, R. R., Becker, R. C. & Sullenger, B. A. Aptamers as therapeutics. Annu. Rev. Pharmacol. Toxicol. 57, 61–79 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gotrik, M. R., Feagin, T. A., Csordas, A. T., Nakamoto, M. A. & Soh, H. T. Advancements in aptamer discovery technologies. Acc. Chem. Res. 49, 1903–1910 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Attwood, T. K., Peffifer, S. R. & Thorne, D. Bioinformatics challenges at the interface of biology and computer science: Mind the gap. (John Wiley & Sons, 2016).

  26. Dobson, C. M. Protein folding and misfolding. Nature 426, 884–890 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. Kozlowski, L. P. Proteome-pI: proteome isoelectric point database. Nucleic Acids Res. 45, D1112–D1116 (2017).

    Article  CAS  PubMed  Google Scholar 

  28. Manning, G. S. The persistence length of DNA is reached from the persistence length of its null isomer through an internal electrostatic stretching force. Biophys. J. 91, 3607–3616 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lane, M. D. & Seelig, B. Advances in the directed evolution of proteins. Curr. Opin. Chem. Biol. 22, 129–136 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Turner, N. J. Directed evolution drives the next generation of biocatalysts. Nat. Chem. Biol. 5, 567–573 (2009).

    Article  CAS  PubMed  Google Scholar 

  31. Packer, M. S. & Liu, D. R. Methods for the directed evolution of proteins. Nat. Rev. Genet. 16, 379–394 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Seelig, B. & Szostak, J. W. Selection and evolution of enzymes from a partially randomized non-catalytic scaffold. Nature 448, 828–831 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wilson, D. S., Keefe, A. D. & Szostak, J. W. The use of mRNA display to select high-affinity protein-binding peptides. Proc. Natl Acad. Sci. USA 98, 3750–3755 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Boder, E. T. & Wittrup, K. D. Yeast surface display for screening combinatorial polypeptide libraries. Nat. Biotechnol. 15, 553–557 (1997).

    Article  CAS  PubMed  Google Scholar 

  35. Wang, X. X., Cho, Y. K. & Shusta, E. V. Mining a yeast library for brain endothelial cell-binding antibodies. Nat. Methods 4, 143–145 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yi, L. et al. Engineering of TEV protease variants by yeast ER sequestration screening (YESS) of combinatorial libraries. Proc. Natl Acad. Sci. USA 110, 7229–7234 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Woolfson, D. N. et al. De novo protein design: how do we expand into the universe of possible protein structures? Curr. Opin. Struct. Biol. 33, 16–26 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Huang, P. S., Boyken, S. E. & Baker, D. The coming of age of de novo protein design. Nature 537, 320–327 (2016).

    Article  CAS  PubMed  Google Scholar 

  39. Damborsky, J. & Brezovsky, J. Computational tools for designing and engineering enzymes. Curr. Opin. Chem. Biol. 19, 8–16 (2014).

    Article  CAS  PubMed  Google Scholar 

  40. Hilvert, D. Design of protein catalysts. Annu. Rev. Biochem. 82, 447–470 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Khoury, G. A., Smadbeck, J., Kieslich, C. A. & Floudas, C. A. Protein folding and de novo protein design for biotechnological applications. Trends Biotechnol. 32, 99–109 (2014).

    Article  CAS  PubMed  Google Scholar 

  42. Rocklin, G. J. et al. Global analysis of protein folding using massively parallel design, synthesis, and testing. Science 357, 168–175 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Butterfield, G. L. et al. Evolution of a designed protein assembly encapsulating its own RNA genome. Nature 552, 415–420 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Chevalier, A. et al. Massively parallel de novo protein design for targeted therapeutics. Nature 550, 74–79 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Keefe, A. D., Pai, S. & Ellington, A. Aptamers as therapeutics. Nat. Rev. Drug. Discov. 9, 537–550 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Han, D. et al. Single-stranded DNA and RNA origami. Science 358, eaao2648 (2017).

    Article  CAS  Google Scholar 

  47. Geary, C., Rothemund, P. W. & Andersen, E. S. RNA nanostructures. A single-stranded architecture for cotranscriptional folding of RNA nanostructures. Science 345, 799–804 (2014).

    Article  CAS  PubMed  Google Scholar 

  48. Wagenbauer, K. F., Sigl, C. & Dietz, H. Gigadalton-scale shape-programmable DNA assemblies. Nature 552, 78–83 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Tikhomirov, G., Petersen, P. & Qian, L. Fractal assembly of micrometre-scale DNA origami arrays with arbitrary patterns. Nature 552, 67–71 (2017).

    Article  CAS  PubMed  Google Scholar 

  50. Wickham, S. F. et al. A DNA-based molecular motor that can navigate a network of tracks. Nat. Nanotechnol. 7, 169–173 (2012).

    Article  CAS  PubMed  Google Scholar 

  51. Omabegho, T., Sha, R. & Seeman, N. C. A bipedal DNA Brownian motor with coordinated legs. Science 324, 67–71 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Derr, N. D. et al. Tug-of-war in motor protein ensembles revealed with a programmable DNA origami scaffold. Science 338, 662–665 (2012).

    Article  CAS  PubMed  Google Scholar 

  53. Wollman, A. J., Sanchez-Cano, C., Carstairs, H. M., Cross, R. A. & Turberfield, A. J. Transport and self-organization across different length scales powered by motor proteins and programmed by DNA. Nat. Nanotechnol. 9, 44–47 (2014).

    Article  CAS  PubMed  Google Scholar 

  54. Jung, C., Allen, P. B. & Ellington, A. D. A stochastic DNA walker that traverses a microparticle surface. Nat. Nanotechnol. 11, 157–163 (2016).

    Article  CAS  PubMed  Google Scholar 

  55. Li, J., Green, A. A., Yan, H. & Fan, C. Engineering nucleic acid structures for programmable molecular circuitry and intracellular biocomputation. Nat. Chem. 9, 1056–1067 (2017).

    Article  CAS  PubMed  Google Scholar 

  56. Amir, Y. et al. Universal computing by DNA origami robots in a living animal. Nat. Nanotechnol. 9, 353–357 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Burns, J. R., Seifert, A., Fertig, N. & Howorka, S. A biomimetic DNA-based channel for the ligand-controlled transport of charged molecular cargo across a biological membrane. Nat. Nanotechnol. 11, 152–156 (2016).

    Article  CAS  PubMed  Google Scholar 

  58. Langecker, M. et al. Synthetic lipid membrane channels formed by designed DNA nanostructures. Science 338, 932–936 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Bell, N. A. et al. DNA origami nanopores. Nano Lett. 12, 512–517 (2012).

    Article  CAS  PubMed  Google Scholar 

  60. Burns, J., Stulz, E. & Howorka, S. Self-assembled DNA nanopores that span lipid bilayers. Nano Lett. 13, 2351–2356 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Howorka, S. Building membrane nanopores. Nat. Nanotechnol. 12, 619–630 (2017).

    Article  CAS  PubMed  Google Scholar 

  62. Ayub, M. & Bayley, H. Engineered transmembrane pores. Curr. Opin. Chem. Biol. 34, 117–126 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Spiess, C. et al. Bispecific antibodies with natural architecture produced by co-culture of bacteria expressing two distinct half-antibodies. Nat. Biotechnol. 31, 753–758 (2013).

    Article  CAS  PubMed  Google Scholar 

  64. Brekke, O. H. & Sandlie, I. Therapeutic antibodies for human diseases at the dawn of the twenty-first century. Nat. Rev. Drug. Discov. 2, 52–62 (2003).

    Article  CAS  PubMed  Google Scholar 

  65. Kohler, G. & Milstein, C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 256, 495–497 (1975).

    Article  CAS  PubMed  Google Scholar 

  66. Boulianne, G. L., Hozumi, N. & Shulman, M. J. Production of functional chimaeric mouse/human antibody. Nature 312, 643–646 (1984).

    Article  CAS  PubMed  Google Scholar 

  67. Better, M., Chang, C. P., Robinson, R. R. & Horwitz, A. H. Escherichia coli secretion of an active chimeric antibody fragment. Science 240, 1041–1043 (1988).

    Article  CAS  PubMed  Google Scholar 

  68. Jones, P. T., Dear, P. H., Foote, J., Neuberger, M. S. & Winter, G. Replacing the complementarity-determining regions in a human antibody with those from a mouse. Nature 321, 522–525 (1986).

    Article  CAS  PubMed  Google Scholar 

  69. de Wildt, R. M., Mundy, C. R., Gorick, B. D. & Tomlinson, I. M. Antibody arrays for high-throughput screening of antibody-antigen interactions. Nat. Biotechnol. 18, 989–994 (2000).

    Article  CAS  PubMed  Google Scholar 

  70. Kontermann, R. E. & Brinkmann, U. Bispecific antibodies. Drug Discov. Today 20, 838–847 (2015).

    Article  CAS  PubMed  Google Scholar 

  71. Bargou, R. et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science 321, 974–977 (2008).

    Article  CAS  PubMed  Google Scholar 

  72. Brinkmann, U. & Kontermann, R. E. The making of bispecific antibodies. MAbs 9, 182–212 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Wu, A. M. & Senter, P. D. Arming antibodies: prospects and challenges for immunoconjugates. Nat. Biotechnol. 23, 1137–1146 (2005).

    Article  CAS  PubMed  Google Scholar 

  74. Gross, G., Waks, T. & Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl Acad. Sci. USA 86, 10024–10028 (1989).

  75. Hamers-Casterman, C. et al. Naturally occurring antibodies devoid of light chains. Nature 363, 446–448 (1993).

    Article  CAS  PubMed  Google Scholar 

  76. Stanfield, R. L., Dooley, H., Flajnik, M. F. & Wilson, I. A. Crystal structure of a shark single-domain antibody V region in complex with lysozyme. Science 305, 1770–1773 (2004).

    Article  CAS  PubMed  Google Scholar 

  77. Muyldermans, S. Nanobodies: natural single-domain antibodies. Annu. Rev. Biochem. 82, 775–797 (2013).

    Article  CAS  PubMed  Google Scholar 

  78. Dolk, E. et al. Isolation of llama antibody fragments for prevention of dandruff by phage display in shampoo. Appl. Environ. Microbiol. 71, 442–450 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Hussack, G. et al. Neutralization of Clostridium difficile toxin A with single-domain antibodies targeting the cell receptor binding domain. J. Biol. Chem. 286, 8961–8976 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Rothbauer, U. et al. Targeting and tracing antigens in live cells with fluorescent nanobodies. Nat. Methods 3, 887–889 (2006).

    Article  CAS  PubMed  Google Scholar 

  81. Jost, C. & Pluckthun, A. Engineered proteins with desired specificity: DARPins, other alternative scaffolds and bispecific IgGs. Curr. Opin. Struct. Biol. 27, 102–112 (2014).

    Article  CAS  PubMed  Google Scholar 

  82. Binz, H. K. et al. High-affinity binders selected from designed ankyrin repeat protein libraries. Nat. Biotechnol. 22, 575–582 (2004).

    Article  CAS  PubMed  Google Scholar 

  83. Huber, T., Steiner, D., Rothlisberger, D. & Pluckthun, A. In vitro selection and characterization of DARPins and Fab fragments for the co-crystallization of membrane proteins: The Na+-citrate symporter CitS as an example. J. Struct. Biol. 159, 206–221 (2007).

    Article  CAS  PubMed  Google Scholar 

  84. Pluckthun, A. Designed ankyrin repeat proteins (DARPins): binding proteins for research, diagnostics, and therapy. Annu. Rev. Pharmacol. Toxicol. 55, 489–511 (2015).

    Article  CAS  PubMed  Google Scholar 

  85. Park, K. et al. Control of repeat-protein curvature by computational protein design. Nat. Struct. Mol. Biol. 22, 167–174 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Marold, J. D., Kavran, J. M., Bowman, G. D. & Barrick, D. A. Naturally occurring repeat protein with high internal sequence identity defines a new class of TPR-like proteins. Structure 23, 2055–2065 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Urvoas, A. et al. Design, production and molecular structure of a new family of artificial alpha-helicoidal repeat proteins (alphaRep) based on thermostable HEAT-like repeats. J. Mol. Biol. 404, 307–327 (2010).

    Article  CAS  PubMed  Google Scholar 

  88. Boersma, Y. L. & Pluckthun, A. DARPins and other repeat protein scaffolds: advances in engineering and applications. Curr. Opin. Biotechnol. 22, 849–857 (2011).

    Article  CAS  PubMed  Google Scholar 

  89. Parmeggiani, F. & Huang, P. S. Designing repeat proteins: a modular approach to protein design. Curr. Opin. Struct. Biol. 45, 116–123 (2017).

    Article  CAS  PubMed  Google Scholar 

  90. Driggers, E. M., Hale, S. P., Lee, J. & Terrett, N. K. The exploration of macrocycles for drug discovery — an underexploited structural class. Nat. Rev. Drug. Discov. 7, 608–624 (2008).

    Article  CAS  PubMed  Google Scholar 

  91. Hosseinzadeh, P. et al. Comprehensive computational design of ordered peptide macrocycles. Science 358, 1461–1466 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Ng, E. W. et al. Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease. Nat. Rev. Drug. Discov. 5, 123–132 (2006).

    Article  CAS  PubMed  Google Scholar 

  93. Meng, H. M. et al. Aptamer-integrated DNA nanostructures for biosensing, bioimaging and cancer therapy. Chem. Soc. Rev. 45, 2583–2602 (2016).

    Article  CAS  PubMed  Google Scholar 

  94. Rinker, S., Ke, Y. G., Liu, Y., Chhabra, R. & Yan, H. Self-assembled DNA nanostructures for distance-dependent multivalent ligand-protein binding. Nat. Nanotechnol. 3, 418–422 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Fisher, M. A. & Tullman-Ercek, D. Change, exchange, and rearrange: protein engineering for the biotechnological production of fuels, pharmaceuticals, and other chemicals. Curr. Opin. Biotechnol. 24, 1010–1016 (2013).

    Article  CAS  PubMed  Google Scholar 

  96. Porter, J. L., Rusli, R. A. & Ollis, D. L. Directed evolution of enzymes for industrial biocatalysis. ChemBioChem 17, 197–203 (2016).

    Article  CAS  PubMed  Google Scholar 

  97. Bornscheuer, U. T. et al. Engineering the third wave of biocatalysis. Nature 485, 185–194 (2012).

    Article  CAS  PubMed  Google Scholar 

  98. Atsumi, S., Hanai, T. & Liao, J. C. Non-fermentative pathways for synthesis of branched-chain higher alcohols as biofuels. Nature 451, 86–89 (2008).

    Article  CAS  PubMed  Google Scholar 

  99. Steen, E. J. et al. Microbial production of fatty-acid-derived fuels and chemicals from plant biomass. Nature 463, 559–562 (2010).

    Article  CAS  PubMed  Google Scholar 

  100. Savile, C. K. et al. Biocatalytic asymmetric synthesis of chiral amines from ketones applied to sitagliptin manufacture. Science 329, 305–309 (2010).

    Article  CAS  PubMed  Google Scholar 

  101. O’Maille, P. E. et al. Quantitative exploration of the catalytic landscape separating divergent plant sesquiterpene synthases. Nat. Chem. Biol. 4, 617–623 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Lichman, B. R., Zhao, J., Hailes, H. C. & Ward, J. M. Enzyme catalysed Pictet-Spengler formation of chiral 1,1ʹ-disubstituted- and spiro-tetrahydroisoquinolines. Nat. Commun. 8, 14883 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Griengl, H., Schwab, H. & Fechter, M. The synthesis of chiral cyanohydrins by oxynitrilases. Trends Biotechnol. 18, 252–256 (2000).

    Article  CAS  PubMed  Google Scholar 

  104. DeSantis, G. et al. Creation of a productive, highly enantioselective nitrilase through gene site saturation mutagenesis (GSSM). J. Am. Chem. Soc. 125, 11476–11477 (2003).

    Article  CAS  PubMed  Google Scholar 

  105. Stemmer, W. P. Rapid evolution of a protein in vitro by DNA shuffling. Nature 370, 389–391 (1994).

    Article  CAS  PubMed  Google Scholar 

  106. Wells, J. A., Vasser, M. & Powers, D. B. Cassette mutagenesis: an efficient method for generation of multiple mutations at defined sites. Gene 34, 315–323 (1985).

    Article  CAS  PubMed  Google Scholar 

  107. Estell, D. A., Graycar, T. P. & Wells, J. A. Engineering an enzyme by site-directed mutagenesis to be resistant to chemical oxidation. J. Biol. Chem. 260, 6518–6521 (1985).

    CAS  PubMed  Google Scholar 

  108. Prier, C. K., Zhang, R. K., Buller, A. R., Brinkmann-Chen, S. & Arnold, F. H. Enantioselective, intermolecular benzylic C–H amination catalysed by an engineered iron-haem enzyme. Nat. Chem. 9, 629–634 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Hammer, S. C. et al. Anti-Markovnikov alkene oxidation by metal-oxo-mediated enzyme catalysis. Science 358, 215–218 (2017).

    Article  CAS  PubMed  Google Scholar 

  110. Giger, L. et al. Evolution of a designed retro-aldolase leads to complete active site remodeling. Nat. Chem. Biol. 9, 494–498 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Chao, F. A. et al. Structure and dynamics of a primordial catalytic fold generated by in vitro evolution. Nat. Chem. Biol. 9, 81–83 (2013).

    Article  CAS  PubMed  Google Scholar 

  112. Dodani, S. C. et al. Discovery of a regioselectivity switch in nitrating P450s guided by molecular dynamics simulations and Markov models. Nat. Chem. 8, 419–425 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Chen, I., Dorr, B. M. & Liu, D. R. A general strategy for the evolution of bond-forming enzymes using yeast display. Proc. Natl Acad. Sci. USA 108, 11399–11404 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Preiswerk, N. et al. Impact of scaffold rigidity on the design and evolution of an artificial Diels–Alderase. Proc. Natl Acad. Sci. USA 111, 8013–8018 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Rothlisberger, D. et al. Kemp elimination catalysts by computational enzyme design. Nature 453, 190–195 (2008).

    Article  CAS  PubMed  Google Scholar 

  116. Kries, H., Blomberg, R. & Hilvert, D. De novo enzymes by computational design. Curr. Opin. Chem. Biol. 17, 221–228 (2013).

    Article  CAS  PubMed  Google Scholar 

  117. Lu, Y., Yeung, N., Sieracki, N. & Marshall, N. M. Design of functional metalloproteins. Nature 460, 855–862 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Keys, T. G. et al. Engineering the product profile of a polysialyltransferase. Nat. Chem. Biol. 10, 437–442 (2014).

    Article  CAS  PubMed  Google Scholar 

  119. Fox, R. J. et al. Improving catalytic function by ProSAR-driven enzyme evolution. Nat. Biotechnol. 25, 338–344 (2007).

    Article  CAS  PubMed  Google Scholar 

  120. Burton, A. J., Thomson, A. R., Dawson, W. M., Brady, R. L. & Woolfson, D. N. Installing hydrolytic activity into a completely de novo protein framework. Nat. Chem. 8, 837–844 (2016).

    Article  CAS  PubMed  Google Scholar 

  121. Blomberg, R. et al. Precision is essential for efficient catalysis in an evolved Kemp eliminase. Nature 503, 418–421 (2013).

    Article  CAS  PubMed  Google Scholar 

  122. Merski, M. & Shoichet, B. K. Engineering a model protein cavity to catalyze the Kemp elimination. Proc. Natl Acad. Sci. USA 109, 16179–16183 (2012).

  123. Korendovych, I. V. et al. Design of a switchable eliminase. Proc. Natl Acad. Sci. USA 108, 6823–6827 (2011).

  124. Bhan, N., Xu, P. & Koffas, M. A. Pathway and protein engineering approaches to produce novel and commodity small molecules. Curr. Opin. Biotechnol. 24, 1137–1143 (2013).

    Article  CAS  PubMed  Google Scholar 

  125. Kuchler, A., Yoshimoto, M., Luginbuhl, S., Mavelli, F. & Walde, P. Enzymatic reactions in confined environments. Nat. Nanotechnol. 11, 409–420 (2016).

    Article  CAS  PubMed  Google Scholar 

  126. Agapakis, C. M., Boyle, P. M. & Silver, P. A. Natural strategies for the spatial optimization of metabolism in synthetic biology. Nat. Chem. Biol. 8, 527–535 (2012).

    Article  CAS  PubMed  Google Scholar 

  127. Kell, D. B., Swainston, N., Pir, P. & Oliver, S. G. Membrane transporter engineering in industrial biotechnology and whole cell biocatalysis. Trends Biotechnol. 33, 237–246 (2015).

    Article  CAS  PubMed  Google Scholar 

  128. Cuenoud, B. & Szostak, J. W. A. DNA metalloenzyme with DNA ligase activity. Nature 375, 611–614 (1995).

    Article  CAS  PubMed  Google Scholar 

  129. Ponce-Salvatierra, A., Wawrzyniak-Turek, K., Steuerwald, U., Hobartner, C. & Pena, V. Crystal structure of a DNA catalyst. Nature 529, 231–234 (2016).

    Article  CAS  PubMed  Google Scholar 

  130. Horning, D. P. & Joyce, G. F. Amplification of RNA by an RNA polymerase ribozyme. Proc. Natl Acad. Sci. USA 113, 9786–9791 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Samanta, B. & Joyce, G. F. A reverse transcriptase ribozyme. eLife 6, e31153 (2017).

    Article  Google Scholar 

  132. Cech, T. R. Structural biology. The ribosome is a ribozyme. Science 289, 878–879 (2000).

    Article  CAS  PubMed  Google Scholar 

  133. Ban, N., Nissen, P., Hansen, J., Moore, P. B. & Steitz, T. A. The complete atomic structure of the large ribosomal subunit at 2.4 A resolution. Science 289, 905–920 (2000).

    Article  CAS  PubMed  Google Scholar 

  134. Silverman, S. K. Catalytic DNA: scope, applications, and biochemistry of deoxyribozymes. Trends Biochem. Sci. 41, 595–609 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Pinheiro, V. B. et al. Synthetic genetic polymers capable of heredity and evolution. Science 336, 341–344 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Pinheiro, V. B. & Holliger, P. Towards XNA nanotechnology: new materials from synthetic genetic polymers. Trends Biotechnol. 32, 321–328 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Winnacker, M. & Kool, E. T. Artificial genetic sets composed of size-expanded base pairs. Angew. Chem. Int. Ed. 52, 12498–12508 (2013).

    Article  CAS  Google Scholar 

  138. Rioz-Martinez, A. & Roelfes, G. DNA-based hybrid catalysis. Curr. Opin. Chem. Biol. 25, 80–87 (2015).

    Article  CAS  PubMed  Google Scholar 

  139. Boersma, A. J. et al. Catalytic enantioselective syn hydration of enones in water using a DNA-based catalyst. Nat. Chem. 2, 991–995 (2010).

    Article  CAS  PubMed  Google Scholar 

  140. Gartner, Z. J. et al. DNA-templated organic synthesis and selection of a library of macrocycles. Science 305, 1601–1605 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. O’Reilly, R. K., Turberfield, A. J. & Wilks, T. R. The evolution of DNA-templated synthesis as a tool for materials discovery. Acc. Chem. Res. 50, 2496–2509 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Meng, W. et al. An autonomous molecular assembler for programmable chemical synthesis. Nat. Chem. 8, 542–548 (2016).

    Article  CAS  PubMed  Google Scholar 

  143. Alberts, B. et al. Molecular biology of the cell, Sixth edition. (Garland Publishing, 2014).

  144. Sleytr, U. B., Schuster, B., Egelseer, E. M. & Pum, D. S-layers: principles and applications. FEMS Microbiol. Rev. 38, 823–864 (2014).

    Article  CAS  PubMed  Google Scholar 

  145. Fagan, R. P. & Fairweather, N. F. Biogenesis and functions of bacterial S-layers. Nat. Rev. Microbiol. 12, 211–222 (2014).

    Article  CAS  PubMed  Google Scholar 

  146. Missiakas, D. & Schneewind, O. Assembly and function of the Bacillus anthracis S-layer. Annu. Rev. Microbiol. 71, 79–98 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Baranova, E. et al. SbsB structure and lattice reconstruction unveil Ca2+ triggered S-layer assembly. Nature 487, 119–122 (2012).

    Article  CAS  PubMed  Google Scholar 

  148. van Raaij, M. J., Mitraki, A., Lavigne, G. & Cusack, S. A triple beta-spiral in the adenovirus fibre shaft reveals a new structural motif for a fibrous protein. Nature 401, 935–938 (1999).

    Article  CAS  PubMed  Google Scholar 

  149. Tanaka, S., Sawaya, M. R. & Yeates, T. O. Structure and mechanisms of a protein-based organelle in Escherichia coli. Science 327, 81–84 (2010).

    Article  CAS  PubMed  Google Scholar 

  150. Sutter, M., Greber, B., Aussignargues, C. & Kerfeld, C. A. Assembly principles and structure of a 6.5-MDa bacterial microcompartment shell. Science 356, 1293–1297 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Kerfeld, C. A., Heinhorst, S. & Cannon, G. C. Bacterial microcompartments. Annu. Rev. Microbiol. 64, 391–408 (2010).

    Article  CAS  PubMed  Google Scholar 

  152. Yeates, T. O., Crowley, C. S. & Tanaka, S. Bacterial microcompartment organelles: protein shell structure and evolution. Annu. Rev. Biophys. 39, 185–205 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. McMahon, H. T. & Boucrot, E. Molecular mechanism and physiological functions of clathrin-mediated endocytosis. Nat. Rev. Mol. Cell Biol. 12, 517–533 (2011).

    Article  CAS  PubMed  Google Scholar 

  154. Peter, B. J. et al. BAR domains as sensors of membrane curvature: the amphiphysin BAR structure. Science 303, 495–499 (2004).

    Article  CAS  PubMed  Google Scholar 

  155. Johannes, L., Parton, R. G., Bassereau, P. & Mayor, S. Building endocytic pits without clathrin. Nat. Rev. Mol. Cell Biol. 16, 311–321 (2015).

    Article  CAS  PubMed  Google Scholar 

  156. Frost, A. et al. Structural basis of membrane invagination by F-BAR domains. Cell 132, 807–817 (2008).

  157. Woolfson, D. N. & Mahmoud, Z. N. More than just bare scaffolds: towards multi-component and decorated fibrous biomaterials. Chem. Soc. Rev. 39, 3464–3479 (2010).

    Article  CAS  PubMed  Google Scholar 

  158. Ryadnov, M. G. & Woolfson, D. N. Engineering the morphology of a self-assembling protein fibre. Nat. Mater. 2, 329–332 (2003).

    Article  CAS  PubMed  Google Scholar 

  159. Burgess, N. C. et al. Modular design of self-assembling peptide-based nanotubes. J. Am. Chem. Soc. 137, 10554–10562 (2015).

    Article  CAS  PubMed  Google Scholar 

  160. Xu, C. et al. Rational design of helical nanotubes from self-assembly of coiled-coil lock washers. J. Am. Chem. Soc. 135, 15565–15578 (2013).

    Article  CAS  PubMed  Google Scholar 

  161. Magnotti, E. L. et al. Self-assembly of an alpha-helical peptide into a crystalline two-dimensional nanoporous framework. J. Am. Chem. Soc. 138, 16274–16282 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Brodin, J. D. et al. Metal-directed, chemically tunable assembly of one-, two- and three-dimensional crystalline protein arrays. Nat. Chem. 4, 375–382 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Suzuki, Y. et al. Self-assembly of coherently dynamic, auxetic, two-dimensional protein crystals. Nature 533, 369–373 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Gradisar, H. et al. Design of a single-chain polypeptide tetrahedron assembled from coiled-coil segments. Nat. Chem. Biol. 9, 362–366 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Padilla, J. E., Colovos, C. & Yeates, T. O. Nanohedra: using symmetry to design self assembling protein cages, layers, crystals, and filaments. Proc. Natl Acad. Sci. USA 98, 2217–2221 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Lai, Y. T. et al. Structure of a designed protein cage that self-assembles into a highly porous cube. Nat. Chem. 6, 1065–1071 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. King, N. P. et al. Computational design of self-assembling protein nanomaterials with atomic level accuracy. Science 336, 1171–1174 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Mandell, D. J. & Kortemme, T. Computer-aided design of functional protein interactions. Nat. Chem. Biol. 5, 797–807 (2009).

    Article  CAS  PubMed  Google Scholar 

  169. Grueninger, D. et al. Designed protein–protein association. Science 319, 206–209 (2008).

    Article  CAS  PubMed  Google Scholar 

  170. Fletcher, J. M. et al. Self-assembling cages from coiled-coil peptide modules. Science 340, 595–599 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Kanekiyo, M. et al. Self-assembling influenza nanoparticle vaccines elicit broadly neutralizing H1N1 antibodies. Nature 499, 102–106 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Delpeyroux, F. et al. A poliovirus neutralization epitope expressed on hybrid hepatitis B surface antigen particles. Science 233, 472–475 (1986).

    Article  CAS  PubMed  Google Scholar 

  173. Schäffer, C. et al. Novel biocatalysts based on S-layer self-assembly of Geobacillus stearothermophilus NRS 2004/3a: a nanobiotechnological approach. Small 3, 1549–1559 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Ilk, N., Egelseer, E. M. & Sleytr, U. B. S-layer fusion proteins — construction principles and applications. Curr. Opin. Biotechnol. 22, 824–831 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Fan, C. et al. Short N-terminal sequences package proteins into bacterial microcompartments. Proc. Natl Acad. Sci. USA 107, 7509–7514 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  176. Kotterman, M. A. & Schaffer, D. V. Engineering adeno-associated viruses for clinical gene therapy. Nat. Rev. Genet. 15, 445–451 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Shenton, W., Pum, D., Sleytr, U. B. & Mann, S. Synthesis of cadmium sulphide superlattices using self-assembled bacterial S-layers. Nature 389, 585–587 (1997).

    Article  CAS  Google Scholar 

  178. Baneyx, F. & Matthaei, J. F. Self-assembled two-dimensional protein arrays in bionanotechnology: from S-layers to designed lattices. Curr. Opin. Biotechnol. 28, 39–45 (2014).

    Article  CAS  PubMed  Google Scholar 

  179. Howorka, S. Creating regular arrays of nanoparticles with self-assembling protein building blocks. J. Mater. Chem. 17, 2049–2053 (2007).

    Article  CAS  Google Scholar 

  180. Nam, Y. S. et al. Biologically templated photocatalytic nanostructures for sustained light-driven water oxidation. Nat. Nanotechnol. 5, 340–344 (2010).

    Article  CAS  PubMed  Google Scholar 

  181. Lee, Y. J. et al. Fabricating genetically engineered high-power lithium-ion batteries using multiple virus genes. Science 324, 1051–1055 (2009).

    CAS  PubMed  Google Scholar 

  182. Low, H. H. et al. Structure of a type IV secretion system. Nature 508, 550–553 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Winfree, E., Liu, F., Wenzler, L. A. & Seeman, N. C. Design and self-assembly of two-dimensional DNA crystals. Nature 394, 539–544 (1998).

    Article  CAS  PubMed  Google Scholar 

  184. Yan, H., Park, S. H., Finkelstein, G., Reif, J. H. & LaBean, T. H. DNA-templated self-assembly of protein arrays and highly conductive nanowires. Science 301, 1882–1884 (2003).

    Article  CAS  PubMed  Google Scholar 

  185. He, Y. et al. Hierarchical self-assembly of DNA into symmetric supramolecular polyhedra. Nature 452, 198–201 (2008).

    Article  CAS  PubMed  Google Scholar 

  186. Zheng, J. P. et al. From molecular to macroscopic via the rational design of a self-assembled 3D DNA crystal. Nature 461, 74–77 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Andersen, E. S. et al. Self-assembly of a nanoscale DNA box with a controllable lid. Nature 459, 73–76 (2009).

    Article  CAS  PubMed  Google Scholar 

  188. Douglas, S. M. et al. Self-assembly of DNA into nanoscale three-dimensional shapes. Nature 459, 414–418 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Castro, C. E. et al. A primer to scaffolded DNA origami. Nat. Methods 8, 221–229 (2011).

    Article  CAS  PubMed  Google Scholar 

  190. Douglas, S. M. et al. Rapid prototyping of 3D DNA-origami shapes with caDNAno. Nucleic Acids Res. 37, 5001–5006 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Benson, E. et al. DNA rendering of polyhedral meshes at the nanoscale. Nature 523, 441–444 (2015).

    Article  CAS  PubMed  Google Scholar 

  192. Veneziano, R. et al. Designer nanoscale DNA assemblies programmed from the top down. Science 352, 1534 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Dietz, H., Douglas, S. M. & Shih, W. M. Folding DNA into twisted and curved nanoscale shapes. Science 325, 725–730 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Han, D. et al. DNA origami with complex curvatures in three-dimensional space. Science 332, 342–346 (2011).

    Article  CAS  PubMed  Google Scholar 

  195. Kocabey, S. et al. Membrane-assisted growth of DNA origami nanostructure arrays. ACS Nano 9, 3530–3539 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Gerling, T., Wagenbauer, K. F., Neuner, A. M. & Dietz, H. Dynamic DNA devices and assemblies formed by shape-complementary, non-base pairing 3D components. Science 347, 1446–1452 (2015).

    Article  CAS  PubMed  Google Scholar 

  197. Woo, S. & Rothemund, P. W. Programmable molecular recognition based on the geometry of DNA nanostructures. Nat. Chem. 3, 620–627 (2011).

    Article  CAS  PubMed  Google Scholar 

  198. Goodman, R. P. et al. Rapid chiral assembly of rigid DNA building blocks for molecular nanofabrication. Science 310, 1661–1665 (2005).

    Article  CAS  PubMed  Google Scholar 

  199. Shih, W. M., Quispe, J. D. & Joyce, G. F. A. 1.7-kilobase single-stranded DNA that folds into a nanoscale octahedron. Nature 427, 618–621 (2004).

    Article  CAS  PubMed  Google Scholar 

  200. Zhang, F. et al. Complex wireframe DNA origami nanostructures with multi-arm junction vertices. Nat. Nanotechnol. 10, 779–784 (2015).

    Article  CAS  PubMed  Google Scholar 

  201. Wei, B., Dai, M. & Yin, P. Complex shapes self-assembled from single-stranded DNA tiles. Nature 485, 623–626 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Ke, Y., Ong, L. L., Shih, W. M. & Yin, P. Three-dimensional structures self-assembled from DNA bricks. Science 338, 1177–1183 (2012).

    Article  CAS  PubMed  Google Scholar 

  203. Zhang, Z. et al. Placing and shaping liposomes with reconfigurable DNA nanocages. Nat. Chem. 9, 653–659 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Howorka, S. Nanotechnology. Changing of the guard. Science 352, 890–891 (2016).

    Article  CAS  PubMed  Google Scholar 

  205. Franquelim, H. G., Khmelinskaia, A., Sobczak, J. P., Dietz, H. & Schwille, P. Membrane sculpting by curved DNA origami scaffolds. Nat. Commun. 9, 811 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Yoshina-Ishii, C. & Boxer, S. G. Arrays of mobile tethered vesicles on supported lipid bilayers. J. Am. Chem. Soc. 125, 3696–3697 (2003).

    Article  CAS  PubMed  Google Scholar 

  207. Baldelli Bombelli, F. et al. Closed nanoconstructs assembled by step-by-step ss-DNA coupling assisted by phospholipid membranes. Soft Matter 5, 1639–1645 (2009).

    Article  CAS  Google Scholar 

  208. Johnson-Buck, A., Jiang, S., Yan, H. & Walter, N. G. DNA-cholesterol barges as programmable membrane-exploring agents. ACS Nano 8, 5641–5649 (2014).

    Article  CAS  PubMed  Google Scholar 

  209. Suzuki, Y., Endo, M. & Sugiyama, H. Lipid-bilayer-assisted two-dimensional self-assembly of DNA origami nanostructures. Nat. Commun. 6, 8052 (2015).

    Article  CAS  PubMed  Google Scholar 

  210. List, J., Weber, M. & Simmel, F. C. Hydrophobic actuation of a DNA origami bilayer structure. Angew. Chem. Int. Ed. 53, 4236–4239 (2014).

    Article  CAS  Google Scholar 

  211. Czogalla, A. et al. Amphipathic DNA origami nanoparticles to scaffold and deform lipid membrane vesicles. Angew. Chem. Int. Ed. 54, 6501–6505 (2015).

    Article  CAS  Google Scholar 

  212. Edwardson, T. G., Carneiro, K. M., McLaughlin, C. K., Serpell, C. J. & Sleiman, H. F. Site-specific positioning of dendritic alkyl chains on DNA cages enables their geometry-dependent self-assembly. Nat. Chem. 5, 868–875 (2013).

    Article  CAS  PubMed  Google Scholar 

  213. Yang, Y. et al. Self-assembly of size-controlled liposomes on DNA nanotemplates. Nat. Chem. 8, 476–483 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Xu, W. et al. A programmable DNA origami platform to organize SNAREs for membrane fusion. J. Am. Chem. Soc. 138, 4439–4447 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Perrault, S. D. & Shih, W. M. Virus-inspired membrane encapsulation of DNA nanostructures to achieve in vivo stability. ACS Nano 8, 5132–5140 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Howorka, S. DNA nanotechnology: bringing lipid bilayers into shape. Nat. Chem. 9, 611–613 (2017).

    Article  CAS  PubMed  Google Scholar 

  217. Jungmann, R. et al. Single-molecule kinetics and super-resolution microscopy by fluorescence imaging of transient binding on DNA origami. Nano Lett. 10, 4756–4761 (2010).

    Article  CAS  PubMed  Google Scholar 

  218. Iinuma, R. et al. Polyhedra self-assembled from DNA tripods and characterized with 3D DNA-PAINT. Science 344, 65–69 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Modi, S. et al. A DNA nanomachine that maps spatial and temporal pH changes inside living cells. Nat. Nanotechnol. 4, 325–330 (2009).

    Article  CAS  PubMed  Google Scholar 

  220. Modi, S., Nizak, C., Surana, S., Halder, S. & Krishnan, Y. Two DNA nanomachines map pH changes along intersecting endocytic pathways inside the same cell. Nat. Nanotechnol. 8, 459–467 (2013).

    Article  CAS  PubMed  Google Scholar 

  221. Knudsen, J. B. et al. Routing of individual polymers in designed patterns. Nat. Nanotechnol. 10, 892–898 (2015).

    Article  CAS  PubMed  Google Scholar 

  222. Tan, S. J., Campolongo, M. J., Luo, D. & Cheng, W. Building plasmonic nanostructures with DNA. Nat. Nanotechnol. 6, 268–276 (2011).

    Article  CAS  PubMed  Google Scholar 

  223. Ozbay, E. Plasmonics: merging photonics and electronics at nanoscale dimensions. Science 311, 189–193 (2006).

    Article  CAS  PubMed  Google Scholar 

  224. Klinkova, A., Choueiri, R. M. & Kumacheva, E. Self-assembled plasmonic nanostructures. Chem. Soc. Rev. 43, 3976–3991 (2014).

    Article  CAS  PubMed  Google Scholar 

  225. Mirkin, C. A., Letsinger, R. L., Mucic, R. C. & Storhoff, J. J. A DNA-based method for rationally assembling nanoparticles into macroscopic materials. Nature 382, 607–609 (1996).

    Article  CAS  PubMed  Google Scholar 

  226. Alivisatos, A. P. et al. Organization of ‘nanocrystal molecules’ using DNA. Nature 382, 609–611 (1996).

    Article  CAS  PubMed  Google Scholar 

  227. Zheng, J. W. et al. Two-dimensional nanoparticle arrays show the organizational power of robust DNA motifs. Nano Lett. 6, 1502–1504 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Aldaye, F. A., Palmer, A. L. & Sleiman, H. F. Assembling materials with DNA as the guide. Science 321, 1795–1799 (2008).

    Article  CAS  PubMed  Google Scholar 

  229. Sharma, J. et al. Control of self-assembly of DNA tubules through integration of gold nanoparticles. Science 323, 112–116 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Kuzyk, A. et al. DNA-based self-assembly of chiral plasmonic nanostructures with tailored optical response. Nature 483, 311–314 (2012).

    Article  CAS  PubMed  Google Scholar 

  231. Acuna, G. P. et al. Fluorescence enhancement at docking sites of DNA-directed self-assembled nanoantennas. Science 338, 506–510 (2012).

    Article  CAS  PubMed  Google Scholar 

  232. Sun, W. et al. Casting inorganic structures with DNA molds. Science 346, 1258361 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Trinh, T. et al. DNA-imprinted polymer nanoparticles with monodispersity and prescribed DNA-strand patterns. Nat. Chem. 10, 184–192 (2018).

    Article  CAS  PubMed  Google Scholar 

  234. Edwardson, T. G., Lau, K. L., Bousmail, D., Serpell, C. J. & Sleiman, H. F. Transfer of molecular recognition information from DNA nanostructures to gold nanoparticles. Nat. Chem. 8, 162–170 (2016).

    Article  CAS  PubMed  Google Scholar 

  235. Liu, W. et al. Diamond family of nanoparticle superlattices. Science 351, 582–586 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Kershner, R. J. et al. Placement and orientation of individual DNA shapes on lithographically patterned surfaces. Nat. Nanotechnol. 4, 557–561 (2009).

    Article  CAS  PubMed  Google Scholar 

  237. Hung, A. M. et al. Large-area spatially ordered arrays of gold nanoparticles directed by lithographically confined DNA origami. Nat. Nanotechnol. 5, 121–126 (2010).

    Article  CAS  PubMed  Google Scholar 

  238. Gopinath, A., Miyazono, E., Faraon, A. & Rothemund, P. W. Engineering and mapping nanocavity emission via precision placement of DNA origami. Nature 535, 401–405 (2016).

    Article  CAS  PubMed  Google Scholar 

  239. Lee, M. J. et al. Engineered synthetic scaffolds for organizing proteins within the bacterial cytoplasm. Nat. Chem. Biol. 14, 142–147 (2017).

    Article  CAS  PubMed  Google Scholar 

  240. Dueber, J. E. et al. Synthetic protein scaffolds provide modular control over metabolic flux. Nat. Biotechnol. 27, 753–759 (2009).

    Article  CAS  PubMed  Google Scholar 

  241. Flavell, R. R. & Muir, T. W. Expressed protein ligation (EPL) in the study of signal transduction, ion conduction, and chromatin biology. Acc. Chem. Res. 42, 107–116 (2009).

    Article  CAS  PubMed  Google Scholar 

  242. Dawson, P. E., Muir, T. W., Clark-Lewis, I. & Kent, S. B. Synthesis of proteins by native chemical ligation. Science 266, 776–779 (1994).

    Article  CAS  PubMed  Google Scholar 

  243. Chin, J. W. Expanding and reprogramming the genetic code. Nature 550, 53–60 (2017).

    Article  CAS  PubMed  Google Scholar 

  244. Spicer, C. D. & Davis, B. G. Selective chemical protein modification. Nat. Commun. 5, 4740 (2014).

    Article  CAS  PubMed  Google Scholar 

  245. Zolot, R. S., Basu, S. & Million, R. P. Antibody-drug conjugates. Nat. Rev. Drug. Discov. 12, 259–260 (2013).

    Article  CAS  PubMed  Google Scholar 

  246. Chudasama, V., Maruani, A. & Caddick, S. Recent advances in the construction of antibody-drug conjugates. Nat. Chem. 8, 114–119 (2016).

    Article  CAS  PubMed  Google Scholar 

  247. Maruani, A. et al. A plug-and-play approach to antibody-based therapeutics via a chemoselective dual click strategy. Nat. Commun. 6, 6645 (2015).

    Article  CAS  PubMed  Google Scholar 

  248. Chen, T., Hongdilokkul, N., Liu, Z., Thirunavukarasu, D. & Romesberg, F. E. The expanding world of DNA and RNA. Curr. Opin. Chem. Biol. 34, 80–87 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Praetorius, F. et al. Biotechnological mass production of DNA origami. Nature 552, 84–87 (2017).

    Article  CAS  PubMed  Google Scholar 

  250. Agarwal, N. P., Matthies, M., Joffroy, B. & Schmidt, T. L. Structural transformation of wireframe DNA origami DNA polymerase assisted gap-filling. ACS Nano 12, 2546–2553 (2018).

  251. Surana, S., Shenoy, A. R. & Krishnan, Y. Designing DNA nanodevices for compatibility with the immune system of higher organisms. Nat. Nanotechnol. 10, 741–747 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Lee, D. S., Qian, H., Tay, C. Y. & Leong, D. T. Cellular processing and destinies of artificial DNA nanostructures. Chem. Soc. Rev. 45, 4199–4225 (2016).

    Article  CAS  PubMed  Google Scholar 

  253. Bell, N. A. & Keyser, U. F. Digitally encoded DNA nanostructures for multiplexed, single-molecule protein sensing with nanopores. Nat. Nanotechnol. 11, 645–651 (2016).

    Article  CAS  PubMed  Google Scholar 

  254. Dunn, K. E. et al. Guiding the folding pathway of DNA origami. Nature 525, 82–86 (2015).

    Article  CAS  PubMed  Google Scholar 

  255. Sacca, B. et al. Orthogonal protein decoration of DNA origami. Angew. Chem. Int. Ed. 49, 9378–9383 (2010).

    Article  CAS  Google Scholar 

  256. Wilner, O. I. et al. Enzyme cascades activated on topologically programmed DNA scaffolds. Nat. Nanotechnol. 4, 249–254 (2009).

    Article  CAS  PubMed  Google Scholar 

  257. Linko, V., Eerikainen, M. & Kostiainen, M. A. A modular DNA origami-based enzyme cascade nanoreactor. Chem. Commun. 51, 5351–5354 (2015).

    Article  CAS  Google Scholar 

  258. Zhao, Z. et al. Nanocaged enzymes with enhanced catalytic activity and increased stability against protease digestion. Nat. Commun. 7, 10619 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Fu, J. et al. Multi-enzyme complexes on DNA scaffolds capable of substrate channelling with an artificial swinging arm. Nat. Nanotechnol. 9, 531–536 (2014).

    Article  CAS  PubMed  Google Scholar 

  260. Grossi, G., Dalgaard Ebbesen Jepsen, M., Kjems, J. & Andersen, E. S. Control of enzyme reactions by a reconfigurable DNA nanovault. Nat. Commun. 8, 992 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  261. Delebecque, C. J., Lindner, A. B., Silver, P. A. & Aldaye, F. A. Organization of intracellular reactions with rationally designed RNA assemblies. Science 333, 470–474 (2011).

    Article  CAS  PubMed  Google Scholar 

  262. Angelin, A. et al. Multiscale origami structures as interface for cells. Angew. Chem. Int. Ed. 54, 15813–15817 (2015).

    Article  CAS  Google Scholar 

  263. Praetorius, F. & Dietz, H. Self-assembly of genetically encoded DNA-protein hybrid nanoscale shapes. Science 355, eaam5488 (2017).

    Article  CAS  Google Scholar 

  264. Schiffels, D., Szalai, V. A. & Liddle, J. A. Molecular precision at micrometer length scales: hierarchical assembly of DNA–protein nanostructures. ACS Nano 11, 6623–6629 (2017).

    Article  CAS  PubMed  Google Scholar 

  265. Henning-Knechtel, A., Knechtel, J. & Magzoub, M. DNA-assisted oligomerization of pore-forming toxin monomers into precisely-controlled protein channels. Nucleic Acids Res. 45, 12057–12068 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  266. Spruijt, E., Tusk, S. E. & Bayley, H. DNA scaffolds support stable and uniform peptide nanopores. Nat. Nanotechnol. https://doi.org/10.1038/s41565-018-0139-6 (2018).

  267. Zhao, Y. H., Abraham, M. H. & Zissimos, A. M. Fast calculation of van der Waals volume as a sum of atomic and bond contributions and its application to drug compounds. J. Org. Chem. 68, 7368–7373 (2003).

    Article  CAS  PubMed  Google Scholar 

  268. Choi, J. & Majima, T. Conformational changes of non-B DNA. Chem. Soc. Rev. 40, 5893–5909 (2011).

    Article  CAS  PubMed  Google Scholar 

  269. Hansel-Hertsch, R. et al. G-Quadruplex structures mark human regulatory chromatin. Nat. Genet. 48, 1267–1272 (2016).

    Article  CAS  PubMed  Google Scholar 

  270. Colin, P. Y., Zinchenko, A. & Hollfelder, F. Enzyme engineering in biomimetic compartments. Curr. Opin. Struct. Biol. 33, 42–51 (2015).

    Article  CAS  PubMed  Google Scholar 

  271. Fischlechner, M. et al. Evolution of enzyme catalysts caged in biomimetic gel-shell beads. Nat. Chem. 6, 791–796 (2014).

    Article  CAS  PubMed  Google Scholar 

  272. He, M. & Taussig, M. J. Eukaryotic ribosome display with in situ DNA recovery. Nat. Methods 4, 281–288 (2007).

    Article  CAS  PubMed  Google Scholar 

  273. Carlson, J. C., Badran, A. H., Guggiana-Nilo, D. A. & Liu, D. R. Negative selection and stringency modulation in phage-assisted continuous evolution. Nat. Chem. Biol. 10, 216–222 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Tizei, P. A., Csibra, E., Torres, L. & Pinheiro, V. B. Selection platforms for directed evolution in synthetic biology. Biochem. Soc. Trans 44, 1165–1175 (2016).

  275. Currin, A., Swainston, N., Day, P. J. & Kell, D. B. Synthetic biology for the directed evolution of protein biocatalysts: navigating sequence space intelligently. Chem. Soc. Rev. 44, 1172–1239 (2015).

    Article  CAS  PubMed  Google Scholar 

  276. Davids, T., Schmidt, M., Bottcher, D. & Bornscheuer, U. T. Strategies for the discovery and engineering of enzymes for biocatalysis. Curr. Opin. Chem. Biol. 17, 215–220 (2013).

    Article  CAS  PubMed  Google Scholar 

  277. Frushicheva, M. P. et al. Computer aided enzyme design and catalytic concepts. Curr. Opin. Chem. Biol. 21, 56–62 (2014).

    Article  CAS  PubMed  Google Scholar 

  278. Durao, P. et al. Opposing effects of folding and assembly chaperones on evolvability of Rubisco. Nat. Chem. Biol. 11, 148–155 (2015).

    Article  CAS  PubMed  Google Scholar 

  279. Balchin, D., Hayer-Hartl, M. & Hartl, F. U. In vivo aspects of protein folding and quality control. Science 353, aac4354 (2016).

    Article  CAS  PubMed  Google Scholar 

  280. Farías-Rico, J. A., Schmidt, S. & Höcker, B. Evolutionary relationship of two ancient protein superfolds. Nat. Chem. Biol. 10, 710–715 (2014).

  281. Höcker, B. Design of proteins from smaller fragments-learning from evolution. Curr. Opin. Struct. Biol. 27, 56–62 (2014).

  282. Ramakers, B. E., van Hest, J. C. & Lowik, D. W. Molecular tools for the construction of peptide-based materials. Chem. Soc. Rev. 43, 2743–2756 (2014).

    Article  CAS  PubMed  Google Scholar 

  283. Matteucci, M. D. & Caruthers, M. H. Synthesis of deoxyoligonucleotides on a polymer support. J. Am. Chem. Soc. 103, 3185–3191 (1981).

    Article  CAS  Google Scholar 

  284. Kukwikila, M., Gale, N., El-Sagheer, A. H., Brown, T. & Tavassoli, A. Assembly of a biocompatible triazole-linked gene by one-pot click-DNA ligation. Nat. Chem. 9, 1089–1098 (2017).

  285. Gibson, D. G. et al. Complete chemical synthesis, assembly, and cloning of a Mycoplasma genitalium genome. Science 319, 1215–1220 (2008).

    Article  CAS  PubMed  Google Scholar 

  286. Gibson, D. G. et al. Creation of a bacterial cell controlled by a chemically synthesized genome. Science 329, 52–56 (2010).

    Article  CAS  PubMed  Google Scholar 

  287. Shivalingam, A. & Brown, T. Synthesis of chemically modified DNA. Biochem. Soc. Trans 44, 709–715 (2016).

    Article  CAS  PubMed  Google Scholar 

  288. Chen, Z., Lichtor, P. A., Berliner, A. P., Chen, J. C. & Liu, D. R. Evolution of sequence-defined highly functionalized nucleic acid polymers. Nat. Chem. 10, 420–427 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  289. Harris, L. J., Larson, S. B., Hasel, K. W. & McPherson, A. Refined structure of an intact IgG2a monoclonal antibody. Biochemistry 36, 1581–1597 (1997).

    Article  CAS  PubMed  Google Scholar 

  290. Garces, F. et al. Structural evolution of glycan recognition by a family of potent HIV antibodies. Cell 159, 69–79 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  291. Guillard, S. et al. Structural and functional characterization of a DARPin which inhibits Ras nucleotide exchange. Nat. Commun. 8, 16111 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  292. Kato, K. et al. Structural basis for specific inhibition of Autotaxin by a DNA aptamer. Nat. Struct. Mol. Biol. 23, 395–401 (2016).

    Article  CAS  PubMed  Google Scholar 

  293. Jiang, L. et al. De novo computational design of retro-aldol enzymes. Science 319, 1387–1391 (2008).

  294. Sleytr, U. B. et al. Nanobiotechnology with S-layer proteins as building blocks. Prog. Mol. Biol. Transl Sci. 103, 277–352 (2011).

    Article  CAS  PubMed  Google Scholar 

  295. Marvin, D. A., Hale, R. D., Nave, C. & Helmer-Citterich, M. Molecular models and structural comparisons of native and mutant class I filamentous bacteriophages Ff (fd, f1, M13), If1 and IKe. J. Mol. Biol. 235, 260–286 (1994).

    Article  CAS  PubMed  Google Scholar 

  296. Schluenzen, F. et al. Structure of functionally activated small ribosomal subunit at 3.3 angstroms resolution. Cell 102, 615–623 (2000).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Supported the EPSRC (EP/N009282/1), the BBSRC (BB/M025373/1, BB/N017331/1), the Leverhulme Trust (RPG-2017-015) and Oxford Nanopore Technologies. The authors thank Keith Fox, Birte Höcker, and Derek Woolfson for critically reading the manuscript and suggesting improvements, and Katya Ahmad for calculating the van der Waals volumes of amino acids and nucleotides, and suggesting improvements for Box 1.

Author information

Authors and Affiliations

Authors

Contributions

G.C.P. and S.H. researched literature and wrote the article. J.R.B. contributed to the discussion of the content and editing of the manuscript, and generated the illustrations.

Corresponding author

Correspondence to Stefan Howorka.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pugh, G.C., Burns, J.R. & Howorka, S. Comparing proteins and nucleic acids for next-generation biomolecular engineering. Nat Rev Chem 2, 113–130 (2018). https://doi.org/10.1038/s41570-018-0015-9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41570-018-0015-9

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing