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.

  • Article
  • Published:

Radical polymerization inside living cells

Abstract

Polymerization reactions conducted inside cells must be compatible with the complex intracellular environment, which contains numerous molecules and functional groups that could potentially prevent or quench polymerization reactions. Here we report a strategy for directly synthesizing unnatural polymers in cells through free radical photopolymerization using a number of biocompatible acrylic and methacrylic monomers. This offers a platform to manipulate, track and control cellular behaviour by the in cellulo generation of macromolecules that have the ability to alter cellular motility, label cells by the generation of fluorescent polymers for long-term tracking studies, as well as generate a variety of nanostructures within cells. It is remarkable that free radical polymerization chemistry can take place within such complex cellular environments. This demonstration opens up a multitude of new possibilities for how chemists can modulate cellular function and behaviour and for understanding cellular behaviour in response to the generation of free radicals.

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: Intracellular polymerization.
Fig. 2: HeLa cells that underwent intracellular polymerization were less migratory.
Fig. 3: Actin cytoskeleton organization was altered in polymerized HeLa cells.
Fig. 4: Polymerization of NaSS in HeLa cells.
Fig. 5: Co-polymerization of HPMA with AOTCRhB in HeLa cells.
Fig. 6: Polymerization of FMMA in HeLa cells.

Similar content being viewed by others

Data availability

All relevant data are available within the Article or the Supplementary Information.

References

  1. Nicolas, J., Mura, S., Brambilla, D., Mackiewicz, N. & Couvreur, P. Design, functionalization strategies and biomedical applications of targeted biodegradable/biocompatible polymer-based nanocarriers for drug delivery. Chem. Soc. Rev. 42, 1147–1235 (2013).

    Article  CAS  Google Scholar 

  2. Green, J. J. & Elisseeff, J. H. Mimicking biological functionality with polymers for biomedical applications. Nature 540, 386–394 (2016).

    Article  CAS  Google Scholar 

  3. Howes, P. D., Chandrawati, R. & Stevens, M. M. Colloidal nanoparticles as advanced biological sensors. Science 346, 1247390 (2014).

    Article  Google Scholar 

  4. Tang, F., He, F., Cheng, H. & Li, L. Self-assembly of conjugated polymer-Ag@SiO2 hybrid fluorescent nanoparticles for application to cellular imaging. Langmuir 26, 11774–11778 (2010).

    Article  CAS  Google Scholar 

  5. Tonga, G. Y. et al. Supramolecular regulation of bioorthogonal catalysis in cells using nanoparticle-embedded transition metal catalysts. Nat. Chem. 7, 597–603 (2015).

    Article  CAS  Google Scholar 

  6. Yusop, R. M., Unciti-Broceta, A., Johansson, E. M. V., Sanchez-Martin, R. M. & Bradley, M. Palladium-mediated intracellular chemistry. Nat. Chem. 3, 239–243 (2011).

    Article  CAS  Google Scholar 

  7. Collins, M. N. & Birkinshaw, C. Hyaluronic acid based scaffolds for tissue engineering—a review. Carbohydr. Polym. 92, 1262–1279 (2013).

    Article  CAS  Google Scholar 

  8. Tasoglu, S. & Demirci, U. Bioprinting for stem cell research. Trends Biotechnol. 31, 10–19 (2013).

    Article  CAS  Google Scholar 

  9. Hutmacher, D. W. Scaffolds in tissue engineering bone and cartilage. Biomaterials 21, 2529–2543 (2000).

    Article  CAS  Google Scholar 

  10. Meyer, R. A. et al. Biodegradable nanoellipsoidal artificial antigen presenting cells for antigen specific T-cell activation. Small 11, 1519–1525 (2015).

    Article  CAS  Google Scholar 

  11. Hu, C.-M. J. et al. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc. Natl Acad. Sci. USA 108, 10980–10985 (2011).

    Article  CAS  Google Scholar 

  12. Lodish, H. et al. Molecular Cell Biology (W. H. Freeman, 2007).

  13. Zhu, Y., Huang, W., Lee, S. S. K. & Xu, W. Crystal structure of a polyphosphate kinase and its implications for polyphosphate synthesis. EMBO Rep. 6, 681–687 (2005).

    Article  CAS  Google Scholar 

  14. Anderson, A. J., Haywood, G. W. & Dawes, E. A. Biosynthesis and composition of bacterial poly(hydroxyalkanoates). Int. J. Biol. Macromol. 12, 102–105 (1990).

    Article  CAS  Google Scholar 

  15. Kessler, B. & Witholt, B. Factors involved in the regulatory network of polyhydroxyalkanoate metabolism. J. Biotechnol. 86, 97–104 (2001).

    Article  CAS  Google Scholar 

  16. Adams, D. J. Fungal cell wall chitinases and glucanases. Microbiology 150, 2029–2035 (2004).

    Article  CAS  Google Scholar 

  17. Arakawa, Y. et al. Genomic organization of the Klebsiella pneumoniae cps region responsible for serotype K2 capsular polysaccharide synthesis in the virulent strain Chedid. J. Bacteriol. 177, 1788–1796 (1995).

    Article  CAS  Google Scholar 

  18. Chien, L. J. & Lee, C. K. Enhanced hyaluronic acid production in Bacillus subtilis by coexpressing bacterial hemoglobin. Biotechnol. Prog. 23, 1017–1022 (2007).

    CAS  PubMed  Google Scholar 

  19. Brangwynne, C. P., Tompa, P. & von Pappu, R. Polymer physics of intracellular phase transitions. Nat. Phys. 11, 899–904 (2015).

    Article  CAS  Google Scholar 

  20. Rehm, B. H. A. Bacterial polymers: biosynthesis, modifications and applications. Nat. Rev. Microbiol. 8, 578–592 (2010).

    Article  CAS  Google Scholar 

  21. Yang, S. H. et al. Mussel-inspired encapsulation and functionalization of individual yeast cells. J. Am. Chem. Soc. 133, 2795–2797 (2011).

    Article  CAS  Google Scholar 

  22. Tytgat, L. et al. in 3D Printing and Biofabrication (eds Ovsianikov, A., Yoo, J. & Mironov, V.) Vol. 96, 1–43 (Springer International, 2017).

  23. Williams, C. G., Malik, A. N., Kim, T. K., Manson, P. N. & Elisseeff, J. H. Variable cytocompatibility of six cell lines with photoinitiators used for polymerizing hydrogels and cell encapsulation. Biomaterials 26, 1211–1218 (2005).

    Article  CAS  Google Scholar 

  24. Yang, J. et al. Nanoencapsulation of individual mammalian cells with cytoprotective polymer shell. Biomaterials 133, 253–262 (2017).

    Article  CAS  Google Scholar 

  25. Xia, Y. et al. Photopolymerized injectable water-soluble maleilated chitosan/poly(ethylene glycol) diacrylate hydrogels as potential tissue engineering scaffolds. J. Photopolym. Sci. Technol. 30, 33–40 (2017).

    Article  CAS  Google Scholar 

  26. Pathak, C. P., Sawhney, A. S. & Hubbell, J. A. Rapid photopolymerization of immunoprotective gels in contact with cells and tissue. J. Am. Chem. Soc. 114, 8311–8312 (1992).

    Article  CAS  Google Scholar 

  27. Magennis, E. P. et al. Bacteria-instructed synthesis of polymers for self-selective microbial binding and labelling. Nat. Mater. 13, 748–755 (2014).

    Article  CAS  Google Scholar 

  28. Kim, J. Y. et al. Cytocompatible polymer grafting from individual living cells by atom‐transfer radical polymerization. Angew. Chem. Int. Ed. 55, 15306–15309 (2016).

    Article  CAS  Google Scholar 

  29. Niu, J. et al. Engineering live cell surfaces with functional polymers via cytocompatible controlled radical polymerization. Nat. Chem. 9, 537–545 (2017).

    Article  CAS  Google Scholar 

  30. Ma, X. et al. Construction and potential applications of a functionalized cell with an intracellular mineral scaffold. Angew. Chem. Int. Ed. 50, 7414–7417 (2011).

    Article  CAS  Google Scholar 

  31. Sweeney, R. Y. et al. Bacterial biosynthesis of cadmium sulfide nanocrystals. Chem. Biol. 11, 1553–1559 (2004).

    Article  CAS  Google Scholar 

  32. Klaus, T., Joerger, R., Olsson, E. & Granqvist, C.-G. Silver-based crystalline nanoparticles, microbially fabricated. Proc. Natl Acad. Sci. USA 96, 13611–13614 (1999).

    Article  CAS  Google Scholar 

  33. Said, El,W. A., Cho, H. Y., Yea, C. H. & Choi, J. W. Synthesis of metal nanoparticles inside living human cells based on the intracellular formation process. Adv. Mater. 26, 910–918 (2014).

    Article  Google Scholar 

  34. Li, Y. et al. Mechanism-oriented controllability of intracellular quantum dots formation: the role of glutathione metabolic pathway. ACS Nano 7, 2240–2248 (2013).

    Article  CAS  Google Scholar 

  35. Cui, R. et al. Living yeast cells as a controllable biosynthesizer for fluorescent quantum dots. Adv. Funct. Mater. 19, 2359–2364 (2009).

    Article  CAS  Google Scholar 

  36. Bryant, S. J., Nuttelman, C. R. & Anseth, K. S. Cytocompatibility of UV and visible light photoinitiating systems on cultured NIH/3T3 fibroblasts in vitro. J. Biomater. Sci. Polym. Ed. 11, 439–457 (2000).

    Article  CAS  Google Scholar 

  37. Fedorovich, N. E. et al. The effect of photopolymerization on stem cells embedded in hydrogels. Biomaterials 30, 344–353 (2009).

    Article  CAS  Google Scholar 

  38. Schweikl, H., Spagnuolo, G. & Schmalz, G. Genetic and cellular toxicology of dental resin monomers. J. Dent. Res. 85, 870–877 (2006).

    Article  CAS  Google Scholar 

  39. Issa, Y., Watts, D. C., Brunton, P. A., Waters, C. M. & Duxbury, A. J. Resin composite monomers alter MTT and LDH activity of human gingival fibroblasts in vitro. Dent. Mater. 20, 12–20 (2004).

    Article  CAS  Google Scholar 

  40. Vasey, P. A. et al. Phase I clinical and pharmacokinetic study of PK1 [N-(2-hydroxypropyl)methacrylamide copolymer doxorubicin]: first member of a new class of chemotherapeutic agents—drug–polymer conjugates. Clin. Cancer Res. 5, 83–94 (1999).

    CAS  PubMed  Google Scholar 

  41. Callahan, J. & Kopeček, J. Semitelechelic HPMA copolymers functionalized with triphenylphosphonium as drug carriers for membrane transduction and mitochondrial localization. Biomacromolecules 7, 2347–2356 (2006).

    Article  CAS  Google Scholar 

  42. Kopecek, J. & Kopečková, P. HPMA copolymers: origins, early developments, present and future. Adv. Drug Deliv. Rev. 62, 122–149 (2010).

    Article  CAS  Google Scholar 

  43. Lilly, J. L., Romero, G., Xu, W., Shin, H. Y. & Berron, B. J. Characterization of molecular transport in ultrathin hydrogel coatings for cellular immunoprotection. Biomacromolecules 16, 541–549 (2015).

    Article  CAS  Google Scholar 

  44. Dröge, W. Free radicals in the physiological control of cell function. Physiol. Rev. 82, 47–95 (2002).

    Article  Google Scholar 

  45. Armstrong, D. Advanced Protocols in Oxidative Stress II (Humana, 2010).

  46. Ridley, A. J. et al. Cell migration: integrating signals from front to back. Science 302, 1704–1709 (2003).

    Article  CAS  Google Scholar 

  47. Hood, J. D. & Cheresh, D. A. Role of integrins in cell invasion and migration. Nat. Rev. Cancer 2, 91–100 (2002).

    Article  Google Scholar 

  48. Scharffetter-Kochanek, K. et al. UV-induced reactive oxygen species in photocarcinogenesis and photoaging. J. Biol. Chem. 378, 1247–1257 (1997).

    CAS  Google Scholar 

  49. Roca-Cusachs, P., Conte, V. & Trepat, X. Quantifying forces in cell biology. Nat. Cell Biol. 19, 742–751 (2017).

    Article  CAS  Google Scholar 

  50. Poirier, M. G. & Marko, J. F. Effect of internal friction on biofilament dynamics. Phys. Rev. Lett. 88, 228103 (2002).

    Article  Google Scholar 

  51. Mizuno, D., Tardin, C., Schmidt, C. F. & MacKintosh, F. C. Nonequilibrium mechanics of active cytoskeletal networks. Science 315, 370–373 (2007).

    Article  CAS  Google Scholar 

  52. Vasconcellos, C. A. et al. Reduction in viscosity of cystic-fibrosis sputum in-vitro by gelsolin. Science 263, 969–971 (1994).

    Article  CAS  Google Scholar 

  53. Nakamura, F., Osborn, E., Janmey, P. A. & Stossel, T. P. Comparison of filamin A-induced cross-linking and Arp2/3 complex-mediated branching on the mechanics of actin filaments. J. Biochem. 277, 9148–9154 (2002).

    CAS  Google Scholar 

  54. Blanchoin, L. & Pollard, T. D. Interaction of actin monomers with acanthamoeba actophorin (ADF/cofilin) and profilin. J. Biochem. 273, 25106–25111 (1998).

    CAS  Google Scholar 

  55. Püspöki, Z., Storath, M., Sage, D. & Unser, M. Focus on bio-image informatics, in Proc. AAECB Vol. 219, 69–93 (Springer, 2016).

  56. Boudaoud, A. et al. FibrilTool, an ImageJ plug-in to quantify fibrillar structures in raw microscopy images. Nat. Protoc. 9, 457–463 (2014).

    Article  CAS  Google Scholar 

  57. Wang, Z. et al. Long-term fluorescent cellular tracing by the aggregates of AIE bioconjugates. J. Am. Chem. Soc. 135, 8238–8245 (2013).

    Article  CAS  Google Scholar 

  58. Major, M. D. & Torkelson, J. M. Fluorescence of vinyl aromatic polyelectrolytes: effects of conformation, concentration, and molecular weight of sodium poly (styrene sulfonate). Macromolecules 1986, 2806–2810 (1986).

    Google Scholar 

  59. Ander, P. & Mahmoudhagh, M. K. Excimer formation of poly (styrenesulfonic acid) and its salts in solution. Macromolecules 15, 214–216 (1982).

    Article  Google Scholar 

  60. Yan, J. J. et al. Polymerizing nonfluorescent monomers without incorporating any fluorescent agent produces strong fluorescent polymers. Adv. Mater. 24, 5617–5624 (2012).

    Article  CAS  Google Scholar 

  61. Gerweck, L. E. & Seetharaman, K. Cellular pH gradient in tumor versus normal tissue: potential exploitation for the treatment of cancer. Cancer Res. 56, 1194–1198 (1996).

    CAS  PubMed  Google Scholar 

  62. Johnson, D. E., Ostrowski, P., Jaumouillé, V. & Grinstein, S. The position of lysosomes within the cell determines their luminal pH. J. Cell Biol. 212, 677–692 (2016).

    Article  CAS  Google Scholar 

  63. Bridges, J. W. & Williams, R. T. The fluorescence of indoles and aniline derivatives. Biochem. J. 107, 225–237 (1968).

    Article  CAS  Google Scholar 

  64. Goswami, T. K. et al. Ferrocene-conjugated copper(ii) complexes of l-methionine and phenanthroline bases: synthesis, structure and photocytotoxic activity. Organometallics 31, 3010–3021 (2012).

    Article  CAS  Google Scholar 

  65. Chen, S., Lu, J., Sun, C. & Ma, H. A highly specific ferrocene-based fluorescent probe for hypochlorous acid and its application to cell imaging. Analyst 135, 577–582 (2010).

    Article  CAS  Google Scholar 

  66. Kumar, K., Vulugundam, G., Kondaiah, P. & Bhattacharya, S. Co-liposomes of redox-active alkyl-ferrocene modified low MW branched PEI and DOPE for efficacious gene delivery in serum. J. Mater. Chem. B 3, 2318–2330 (2015).

    Article  CAS  Google Scholar 

  67. Vankayala, R., Kalluru, P., Tsai, H.-H., Chiang, C.-S. & Hwang, K. C. Effects of surface functionality of carbon nanomaterials on short-term cytotoxicity and embryonic development in zebrafish. J. Mater. Chem. B 2, 1038–1047 (2014).

    Article  CAS  Google Scholar 

  68. Blanazs, A., Ryan, A. J. & Armes, S. P. Predictive phase diagrams for RAFT aqueous dispersion polymerization: effect of block copolymer composition, molecular weight and copolymer concentration. Macromolecules 45, 5099–5107 (2012).

    Article  CAS  Google Scholar 

  69. Refojo, M. F. Hydrophobic interaction in poly(2-hydroxyethyl methacrylate) homogeneous hydrogel. J. Polym. Sci. A 5, 3103–3113 (1967).

    Google Scholar 

Download references

Acknowledgements

This work was supported by the European Research Council (advanced grant ADREEM ERC-2013-340469) and the Rosetrees Trust (A865). N.T. acknowledges support from the Commonwealth Scholarship Commission and W.L. from the Chinese Scholarship Council. The authors thank the Wellcome Trust for Multi-user Equipment Grant WT104915MA.

Author information

Authors and Affiliations

Authors

Contributions

J.G. and M.B. conceived, designed and directed the project. W.L. and Y.Z. conducted the control polymerizations and MTT assays. Y.Z. performed the actin experiments, Y.Z. and N.T. conducted the wound healing experiments. W.L., Y.Z. and J.C. carried out work with the fluorescent polymers. J.G., A.L. and M.B. co-wrote the manuscript. All authors analysed the data and contributed to the scientific discussion and revised the manuscript.

Corresponding author

Correspondence to Mark Bradley.

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.

Supplementary information

Supplementary Information

Supplementary Data, Supplementary Figures and Supplementary Methods.

Reporting Summary

Supplementary Movie 1

Polymerized sodium 4-styrenesulfonate inside cells

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Geng, J., Li, W., Zhang, Y. et al. Radical polymerization inside living cells. Nat. Chem. 11, 578–586 (2019). https://doi.org/10.1038/s41557-019-0240-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41557-019-0240-y

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