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.

  • Perspective
  • Published:

Genetically encodable materials for non-invasive biological imaging

Abstract

Many questions in basic biology and medicine require the ability to visualize the function of specific cells and molecules inside living organisms. In this context, technologies such as ultrasound, optoacoustics and magnetic resonance provide non-invasive imaging access to deep-tissue regions, as used in many laboratories and clinics to visualize anatomy and physiology. In addition, recent work has enabled these technologies to image the location and function of specific cells and molecules inside the body by coupling the physics of sound waves, nuclear spins and light absorption to unique protein-based materials. These materials, which include air-filled gas vesicles, capsid-like nanocompartments, pigment-producing enzymes and transmembrane transporters, enable new forms of biomolecular and cellular contrast. The ability of these protein-based contrast agents to be genetically encoded and produced by cells creates opportunities for unprecedented in vivo studies of cellular function, while their amenability to genetic engineering enables atomic-level design of their physical, chemical and biological properties.

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: Small proteins as genetically encoded contrast agents for non-invasive imaging.

panel e adapted with permission from ref. 22, Springer Nature Ltd.

Fig. 2: Proteinaceous nanocompartments as multiscale contrast agents.

panels adapted with permission from: b, ref. 43, Springer Nature Ltd; d, ref. 44, American Chemical Society.

Fig. 3: Genetically encodable air-filled protein nanostructures as multimodality contrast agents.

panels adapted with permission from: ce, ref. 59, AAAS; f, ref. 61, Springer Nature Ltd; g, ref. 64, American Chemical Society.

Similar content being viewed by others

References

  1. Piraner, D. I. et al. Going deeper: biomolecular tools for acoustic and magnetic imaging and control of cellular function. Biochemistry 56, 5202–5209 (2017).

    Article  CAS  Google Scholar 

  2. Marblestone, A. H. et al. Physical principles for scalable neural recording. Front. Comput. Neurosci. 7, 137 (2013).

    Article  Google Scholar 

  3. Wang, L. V. & Yao, J. A practical guide to photoacoustic tomography in the life sciences. Nat. Methods 13, 627–638 (2016).

    Article  CAS  Google Scholar 

  4. Maresca, D. et al. Biomolecular ultrasound and sonogenetics. Annu. Rev. Chem. Biomol. Eng. 9, 229–252 (2018).

    Article  Google Scholar 

  5. Mukherjee, A., Davis, H. C., Ramesh, P., Lu, G. J. & Shapiro, M. G. Biomolecular MRI reporters: evolution of new mechanisms. Prog. Nucl. Magn. Reson. Spectrosc. 102–103, 32–42 (2017).

    Article  Google Scholar 

  6. Paefgen, V., Doleschel, D. & Kiessling, F. Evolution of contrast agents for ultrasound imaging and ultrasound-mediated drug delivery. Front. Pharmacol. 6, 197 (2015).

    Article  Google Scholar 

  7. Wahsner, J., Gale, E. M., Rodríguez-Rodríguez, A. & Caravan, P. Chemistry of MRI contrast agents: current challenges and new frontiers. Chem. Rev. 119, 957–1057 (2019).

    Article  CAS  Google Scholar 

  8. Chung, J.-K. Sodium iodide symporter: its role in nuclear medicine. J. Nucl. Med. 43, 1188–1200 (2002).

    CAS  Google Scholar 

  9. Kircher, M. F., Gambhir, S. S. & Grimm, J. Noninvasive cell-tracking methods. Nat. Rev. Clin. Oncol. 8, 677–688 (2011).

    Article  CAS  Google Scholar 

  10. Louie, A. Y. et al. In vivo visualization of gene expression using magnetic resonance imaging. Nat. Biotechnol. 18, 321–325 (2000).

    Article  CAS  Google Scholar 

  11. Genove, G., DeMarco, U., Xu, H., Goins, W. F. & Ahrens, E. T. A new transgene reporter for in vivo magnetic resonance imaging. Nat. Med. 11, 450–454 (2005).

    Article  CAS  Google Scholar 

  12. Cohen, B., Dafni, H., Meir, G., Harmelin, A. & Neeman, M. Ferritin as an endogenous MRI reporter for noninvasive imaging of gene expression in C6 glioma tumors. Neoplasia 7, 109–117 (2005).

    Article  CAS  Google Scholar 

  13. Duewell, S., Kasserra, C. E., Jezzard, P. & Balaban, R. S. Evaluation of methemoglobin as an autologous intravascular MRI contrast agent. Magn. Reson. Med. 35, 787–789 (1996).

    Article  CAS  Google Scholar 

  14. Shapiro, M. G. et al. Directed evolution of a magnetic resonance imaging contrast agent for noninvasive imaging of dopamine. Nat. Biotechnol. 28, 264–270 (2010).

    Article  CAS  Google Scholar 

  15. Yang, J. J. et al. Rational design of protein-based MRI contrast agents. J. Am. Chem. Soc. 130, 9260–9267 (2008).

    Article  CAS  Google Scholar 

  16. Deans, A. E. et al. Cellular MRI contrast via coexpression of transferrin receptor and ferritin. Magn. Reson. Med. 56, 51–59 (2006).

    Article  CAS  Google Scholar 

  17. Patrick, P. S. et al. Dual-modality gene reporter for in vivo imaging. Proc. Natl Acad. Sci. USA 111, 415–420 (2014).

    Article  CAS  Google Scholar 

  18. Gilad, A. A. et al. Artificial reporter gene providing MRI contrast based on proton exchange. Nat. Biotechnol. 25, 217–219 (2007).

    Article  CAS  Google Scholar 

  19. Yuan, Y. et al. Furin-mediated intracellular self-assembly of olsalazine nanoparticles for enhanced magnetic resonance imaging and tumour therapy. Nat. Mater. 18, 1376–1383 (2019).

    Article  CAS  Google Scholar 

  20. Mukherjee, A., Wu, D., Davis, H. C. & Shapiro, M. G. Non-invasive imaging using reporter genes altering cellular water permeability. Nat. Commun. 7, 13891 (2016).

    Article  CAS  Google Scholar 

  21. Schilling, F. et al. MRI measurements of reporter-mediated increases in transmembrane water exchange enable detection of a gene reporter. Nat. Biotechnol. 35, 75–80 (2017).

    Article  CAS  Google Scholar 

  22. Desai, M., Slusarczyk, A. L., Chapin, A., Barch, M. & Jasanoff, A. Molecular imaging with engineered physiology. Nat. Commun. 7, 13607 (2016).

    Article  CAS  Google Scholar 

  23. Ohlendorf, R. et al. Target-responsive vasoactive probes for ultrasensitive molecular imaging. Nat. Commun. 11, 2399 (2020).

    Article  Google Scholar 

  24. Ntziachristos, V. Going deeper than microscopy: the optical imaging frontier in biology. Nat. Methods 7, 603–614 (2010).

    Article  CAS  Google Scholar 

  25. Wang, L. V. & Hu, S. Photoacoustic tomography: in vivo imaging from organelles to organs. Science 335, 1458–1462 (2012).

    Article  CAS  Google Scholar 

  26. Shu, X. et al. Mammalian expression of infrared fluorescent proteins engineered from a bacterial phytochrome. Science 324, 804–807 (2009).

    Article  Google Scholar 

  27. Filonov, G. S. et al. Bright and stable near-infrared fluorescent protein for in vivo imaging. Nat. Biotechnol. 29, 757–761 (2011).

    Article  CAS  Google Scholar 

  28. Fuenzalida Werner, J. P. et al. Structure-based mutagenesis of phycobiliprotein smURFP for optoacoustic imaging. ACS Chem. Biol. 14, 1896–1903 (2019).

    Article  CAS  Google Scholar 

  29. Stiel, A. C. et al. High-contrast imaging of reversibly switchable fluorescent proteins via temporally unmixed multispectral optoacoustic tomography. Opt. Lett. 40, 367–370 (2015).

    Article  CAS  Google Scholar 

  30. Deán-Ben, X. L. et al. Light fluence normalization in turbid tissues via temporally unmixed multispectral optoacoustic tomography. Opt. Lett. 40, 4691–4694 (2015).

    Article  Google Scholar 

  31. Yao, J. et al. Multiscale photoacoustic tomography using reversibly switchable bacterial phytochrome as a near-infrared photochromic probe. Nat. Methods 13, 67–73 (2016).

    Article  CAS  Google Scholar 

  32. Deán-Ben, X. L. et al. Functional optoacoustic neuro-tomography for scalable whole-brain monitoring of calcium indicators. Light Sci. Appl. 5, e16201 (2016).

    Article  Google Scholar 

  33. Qian, Y. et al. A genetically encoded near-infrared fluorescent calcium ion indicator. Nat. Methods 16, 171–174 (2019).

    Article  CAS  Google Scholar 

  34. Jutz, G., van Rijn, P., Santos Miranda, B. & Böker, A. Ferritin: a versatile building block for bionanotechnology. Chem. Rev. 115, 1653–1701 (2015).

    Article  CAS  Google Scholar 

  35. Gossuin, Y., Gillis, P., Hocq, A., Vuong, Q. L. & Roch, A. Magnetic resonance relaxation properties of superparamagnetic particles. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 1, 299–310 (2009).

    Article  CAS  Google Scholar 

  36. Iordanova, B., Robison, C. S. & Ahrens, E. T. Design and characterization of a chimeric ferritin with enhanced iron loading and transverse NMR relaxation rate. J. Biol. Inorg. Chem. 15, 957–965 (2010).

    Article  CAS  Google Scholar 

  37. Matsumoto, Y., Chen, R., Anikeeva, P. & Jasanoff, A. Engineering intracellular biomineralization and biosensing by a magnetic protein. Nat. Commun. 6, 8721 (2015).

    Article  CAS  Google Scholar 

  38. Liu, X. et al. Engineering genetically-encoded mineralization and magnetism via directed evolution. Sci. Rep. 6, 38019 (2016).

    Article  CAS  Google Scholar 

  39. Douglas, T. et al. Protein engineering of a viral cage for constrained nanomaterials synthesis. Adv. Mater. 14, 415–418 (2002).

    Article  CAS  Google Scholar 

  40. McHugh, C. A. et al. A virus capsid-like nanocompartment that stores iron and protects bacteria from oxidative stress. EMBO J. 33, 1896–1911 (2014).

    Article  CAS  Google Scholar 

  41. He, D. et al. Structural characterization of encapsulated ferritin provides insight into iron storage in bacterial nanocompartments. eLife 5, e18972 (2016).

  42. Giessen, T. W. & Silver, P. A. Widespread distribution of encapsulin nanocompartments reveals functional diversity. Nat. Microbiol. 2, 17029 (2017).

    Article  Google Scholar 

  43. Sigmund, F. et al. Bacterial encapsulins as orthogonal compartments for mammalian cell engineering. Nat. Commun. 9, 1990 (2018).

    Article  Google Scholar 

  44. Sigmund, F. et al. Iron-sequestering nanocompartments as multiplexed electron microscopy gene reporters. ACS Nano 13, 8114–8123 (2019).

    Article  CAS  Google Scholar 

  45. Ramesh, P. et al. Ultraparamagnetic cells formed through intracellular oxidation and chelation of paramagnetic iron. Angew. Chem. Int. Ed. Engl. 57, 12385–12389 (2018).

    Article  CAS  Google Scholar 

  46. Komeili, A. Molecular mechanisms of compartmentalization and biomineralization in magnetotactic bacteria. FEMS Microbiol. Rev. 36, 232–255 (2012).

    Article  CAS  Google Scholar 

  47. Kolinko, I. et al. Biosynthesis of magnetic nanostructures in a foreign organism by transfer of bacterial magnetosome gene clusters. Nat. Nanotechnol. 9, 193–197 (2014).

    Article  CAS  Google Scholar 

  48. Stritzker, J. et al. Vaccinia virus-mediated melanin production allows MR and optoacoustic deep tissue imaging and laser-induced thermotherapy of cancer. Proc. Natl Acad. Sci. USA 110, 3316–3320 (2013).

    Article  CAS  Google Scholar 

  49. Jiang, Y. et al. Violacein as a genetically-controlled, enzymatically amplified and photobleaching-resistant chromophore for optoacoustic bacterial imaging. Sci. Rep. 5, 11048 (2015).

    Article  CAS  Google Scholar 

  50. Lauri, A. et al. Whole-cell photoacoustic sensor based on pigment relocalization. ACS Sens. 4, 603–612 (2019).

    Article  CAS  Google Scholar 

  51. Shapiro, M. G. et al. Biogenic gas nanostructures as ultrasonic molecular reporters. Nat. Nanotechnol. 9, 311–316 (2014).

    Article  CAS  Google Scholar 

  52. Maresca, D. et al. Nonlinear ultrasound imaging of nanoscale acoustic biomolecules. Appl. Phys. Lett. 110, 073704 (2017).

  53. Maresca, D., Sawyer, D. P., Renaud, G., Lee-Gosselin, A. & Shapiro, M. G. Nonlinear X-wave ultrasound imaging of acoustic biomolecules. Phys. Rev. X 8, 041002 (2018).

    CAS  Google Scholar 

  54. Lakshmanan, A. et al. Molecular engineering of acoustic protein nanostructures. ACS Nano 10, 7314–7322 (2016).

    Article  CAS  Google Scholar 

  55. Cherin, E. et al. Acoustic behavior of halobacterium salinarum gas vesicles in the high-frequency range: experiments and modeling. Ultrasound Med. Biol. 43, 1016–1030 (2017).

    Article  Google Scholar 

  56. Lakshmanan, A. et al. Preparation of biogenic gas vesicle nanostructures for use as contrast agents for ultrasound and MRI. Nat. Protoc. 12, 2050–2080 (2017).

    Article  CAS  Google Scholar 

  57. Bourdeau, R. W. et al. Acoustic reporter genes for noninvasive imaging of microorganisms in mammalian hosts. Nature 553, 86–90 (2018).

    Article  CAS  Google Scholar 

  58. Riglar, D. T. & Silver, P. A. Engineering bacteria for diagnostic and therapeutic applications. Nat. Rev. Microbiol. 16, 214–225 (2018).

    Article  CAS  Google Scholar 

  59. Farhadi, A., Ho, G. H., Sawyer, D. P., Bourdeau, R. W. & Shapiro, M. G. Ultrasound imaging of gene expression in mammalian cells. Science 365, 1469–1475 (2019).

    Article  CAS  Google Scholar 

  60. Lakshmanan, A. et al. Acoustic biosensors for ultrasound imaging of enzyme activity. Nat. Chem. Biol. 16, 988–996 (2020).

    Article  CAS  Google Scholar 

  61. Lu, G. J. et al. Acoustically modulated magnetic resonance imaging of gas-filled protein nanostructures. Nat. Mater. 17, 456–463 (2018).

    Article  CAS  Google Scholar 

  62. Shapiro, M. G. et al. Genetically encoded reporters for hyperpolarized xenon magnetic resonance imaging. Nat. Chem. 6, 629–634 (2014).

    Article  CAS  Google Scholar 

  63. Wang, Y., Roose, B. W., Palovcak, E. J., Carnevale, V. & Dmochowski, I. J. A genetically encoded β-lactamase reporter for ultrasensitive 129Xe NMR in mammalian cells. Angew. Chem. Int. Ed. 55, 8984–8987 (2016).

    Article  CAS  Google Scholar 

  64. Lu, G. J. et al. Genetically encodable contrast agents for optical coherence tomography. ACS Nano 14, 7823–7831 (2020).

    Article  CAS  Google Scholar 

  65. Farhadi, A. et al. Genetically encoded phase contrast agents for digital holographic microscopy. Nano Lett. 20, 8127–8134 (2020).

    Article  CAS  Google Scholar 

  66. Wu, D. et al. Genetically encoded nanostructures enable acoustic manipulation of engineered cells. Preprint at bioRxiv https://doi.org/10.1101/691105 (2019).

  67. Bar-Zion, A. et al. Acoustically detonated biomolecules for genetically encodable inertial cavitation. Preprint at bioRxiv https://doi.org/10.1101/620567 (2019).

  68. Gilbert, C. & Ellis, T. Biological engineered living materials: growing functional materials with genetically programmable properties. ACS Synth. Biol. 8, 1–15 (2019).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  70. Yang, K. K., Wu, Z. & Arnold, F. H. Machine-learning-guided directed evolution for protein engineering. Nat. Methods 16, 687–694 (2019).

    Article  CAS  Google Scholar 

  71. Lang, K. & Chin, J. W. Cellular incorporation of unnatural amino acids and bioorthogonal labeling of proteins. Chem. Rev. 114, 4764–4806 (2014).

    Article  CAS  Google Scholar 

  72. Gilad, A. A. & Shapiro, M. G. Molecular imaging in synthetic biology, and synthetic biology in molecular imaging. Mol. Imaging Biol. 19, 373–378 (2017).

    Article  CAS  Google Scholar 

  73. Banani, S. F., Lee, H. O., Hyman, A. A. & Rosen, M. K. Biomolecular condensates: organizers of cellular biochemistry. Nat. Rev. Mol. Cell Biol. 18, 285–298 (2017).

    Article  CAS  Google Scholar 

  74. Bracha, D., Walls, M. T. & Brangwynne, C. P. Probing and engineering liquid-phase organelles. Nat. Biotechnol. 37, 1435–1445 (2019).

    Article  CAS  Google Scholar 

  75. Palmer, A. E., Qin, Y., Park, J. G. & McCombs, J. E. Design and application of genetically encoded biosensors. Trends Biotechnol. 29, 144–152 (2011).

    Article  CAS  Google Scholar 

  76. Szablowski, J. O., Bar-Zion, A. & Shapiro, M. G. Achieving spatial and molecular specificity with ultrasound-targeted biomolecular nanotherapeutics. Acc. Chem. Res. 52, 2427–2434 (2019).

    Article  CAS  Google Scholar 

  77. Suetens, P. Fundamentals of Medical Imaging (Cambridge Univ. Press, 2017).

  78. Errico, C. et al. Ultrafast ultrasound localization microscopy for deep super-resolution vascular imaging. Nature 527, 499–502 (2015).

    Article  CAS  Google Scholar 

  79. Luís Dean-Ben, X. & Razansky, D. Localization optoacoustic tomography. Light Sci. Appl. 7, 18004 (2018).

    Article  Google Scholar 

  80. Seeger, M. et al. Pushing the boundaries of optoacoustic microscopy by total impulse response characterization. Nat. Commun. 11, 2910 (2020).

    Article  CAS  Google Scholar 

  81. Goddard, T. D. et al. UCSF ChimeraX: meeting modern challenges in visualization and analysis. Protein Sci. 27, 14–25 (2018).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to members of the Shapiro and Westmeyer laboratories for helpful discussions. Relevant research in the Shapiro laboratory was supported by the National Institutes of Health (grant nos. R01EB018975 and U54CA199090), the Human Frontier Science Program (RGP0050/2016), the Heritage Medical Research Institute, the Packard Foundation, the Pew Charitable Trust, the Sontag Foundation, the Dana Foundation and the Burroughs Wellcome Fund. A.F. was supported by an NSERC graduate fellowship. Relevant research in the Westmeyer laboratory was supported by the European Research Council under grant agreement nos. ERC-StG 311552 and ERC-COG 865710, the Deutsche Forschungsgemeinschaft through the TUM International Graduate School of Science and Engineering and the Federation of European Biochemical Societies.

Author information

Authors and Affiliations

Authors

Contributions

All authors wrote the manuscript.

Corresponding authors

Correspondence to Gil Gregor Westmeyer or Mikhail G. Shapiro.

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

Farhadi, A., Sigmund, F., Westmeyer, G.G. et al. Genetically encodable materials for non-invasive biological imaging. Nat. Mater. 20, 585–592 (2021). https://doi.org/10.1038/s41563-020-00883-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41563-020-00883-3

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