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.

  • Letter
  • Published:

Efficiency of siRNA delivery by lipid nanoparticles is limited by endocytic recycling

Abstract

Despite efforts to understand the interactions between nanoparticles and cells, the cellular processes that determine the efficiency of intracellular drug delivery remain unclear. Here we examine cellular uptake of short interfering RNA (siRNA) delivered in lipid nanoparticles (LNPs) using cellular trafficking probes in combination with automated high-throughput confocal microscopy. We also employed defined perturbations of cellular pathways paired with systems biology approaches to uncover protein-protein and protein–small molecule interactions. We show that multiple cell signaling effectors are required for initial cellular entry of LNPs through macropinocytosis, including proton pumps, mTOR and cathepsins. siRNA delivery is substantially reduced as 70% of the internalized siRNA undergoes exocytosis through egress of LNPs from late endosomes/lysosomes. Niemann-Pick type C1 (NPC1) is shown to be an important regulator of the major recycling pathways of LNP-delivered siRNAs. NPC1-deficient cells show enhanced cellular retention of LNPs inside late endosomes and lysosomes, and increased gene silencing of the target gene. Our data suggest that siRNA delivery efficiency might be improved by designing delivery vehicles that can escape the recycling pathways.

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

Figure 1: Systems survey for endocytosis of lipid nanoparticles (LNP).
Figure 2: Cellular trafficking of LNPs.
Figure 3: Quantitative analysis for disassembly and recycling of LNPs.
Figure 4: Enhanced cellular retention and efficacy of siAF647-LNPs in NPC1-deficient MEFs.

Similar content being viewed by others

References

  1. Elbashir, S.M. et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 411, 494–498 (2001).

    Article  CAS  Google Scholar 

  2. Whitehead, K.A., Langer, R. & Anderson, D.G. Knocking down barriers: advances in siRNA delivery. Nat. Rev. Drug Discov. 8, 129–138 (2009).

    Article  CAS  Google Scholar 

  3. Davis, M.E. et al. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 464, 1067–1070 (2010).

    Article  CAS  Google Scholar 

  4. Sheridan, C. Proof of concept for next-generation nanoparticle drugs in humans. Nat. Biotechnol. 30, 471–473 (2012).

    Article  CAS  Google Scholar 

  5. Sahay, G., Alakhova, D.Y. & Kabanov, A.V. Endocytosis of nanomedicines. J. Control. Release 145, 182–195 (2010).

    Article  CAS  Google Scholar 

  6. Wang, J., Byrne, J.D., Napier, M.E. & DeSimone, J.M. More effective nanomedicines through particle design. Small 7, 1919–1931 (2011).

    Article  CAS  Google Scholar 

  7. Juliano, R.L., Ming, X. & Nakagawa, O. Cellular uptake and intracellular trafficking of antisense and siRNA oligonucleotides. Bioconjug. Chem. 23, 147–157 (2012).

    Article  CAS  Google Scholar 

  8. Sonawane, N.D., Szoka, F.C. Jr. & Verkman, A.S. Chloride accumulation and swelling in endosomes enhances DNA transfer by polyamine-DNA polyplexes. J. Biol. Chem. 278, 44826–44831 (2003).

    Article  CAS  Google Scholar 

  9. Basha, G. et al. Influence of cationic lipid composition on gene silencing properties of lipid nanoparticle formulations of siRNA in antigen-presenting cells. Mol. Ther. 19, 2186–2200 (2011).

    Article  CAS  Google Scholar 

  10. Suh, J. et al. Real-time gene delivery vector tracking in the endo-lysosomal pathway of live cells. Microsc. Res. Tech. 75, 691–697 (2012).

    Article  CAS  Google Scholar 

  11. Lu, J.J., Langer, R. & Chen, J. A novel mechanism is involved in cationic lipid-mediated functional siRNA delivery. Mol. Pharm. 6, 763–771 (2009).

    Article  CAS  Google Scholar 

  12. Nguyen, J. & Szoka, F.C. Nucleic acid delivery: the missing pieces of the puzzle? Acc. Chem. Res. 45, 1153–1162 (2012).

    Article  CAS  Google Scholar 

  13. Collinet, C. et al. Systems survey of endocytosis by multiparametric image analysis. Nature 464, 243–249 (2010).

    Article  CAS  Google Scholar 

  14. Doherty, G.J. & McMahon, H.T. Mechanisms of endocytosis. Annu. Rev. Biochem. 78, 857–902 (2009).

    Article  CAS  Google Scholar 

  15. Chen, D. et al. Rapid discovery of potent siRNA-containing lipid nanoparticles enabled by controlled microfluidic formulation. J. Am. Chem. Soc. 134, 6948–6951 (2012).

    Article  CAS  Google Scholar 

  16. Love, K.T. et al. Lipid-like materials for low-dose, in vivo gene silencing. Proc. Natl. Acad. Sci. USA 107, 1864–1869 (2010).

    Article  CAS  Google Scholar 

  17. Leuschner, F. et al. Therapeutic siRNA silencing in inflammatory monocytes. Nat. Biotechnol. 29, 1005–1010 (2011).

    Article  CAS  Google Scholar 

  18. Zoncu, R. et al. mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H(+)-ATPase. Science 334, 678–683 (2011).

    Article  CAS  Google Scholar 

  19. Carette, J.E. et al. Ebola virus entry requires the cholesterol transporter Niemann-Pick C1. Nature 477, 340–343 (2011).

    Article  CAS  Google Scholar 

  20. Khalil, I.A., Kogure, K., Akita, H. & Harashima, H. Uptake pathways and subsequent intracellular trafficking in nonviral gene delivery. Pharmacol. Rev. 58, 32–45 (2006).

    Article  CAS  Google Scholar 

  21. Huss, M. et al. Concanamycin A, the specific inhibitor of V-ATPases, binds to the V(o) subunit c. J. Biol. Chem. 277, 40544–40548 (2002).

    Article  CAS  Google Scholar 

  22. Bayer, N. et al. Effect of bafilomycin A1 and nocodazole on endocytic transport in HeLa cells: implications for viral uncoating and infection. J. Virol. 72, 9645–9655 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Ravikumar, B. et al. Regulation of mammalian autophagy in physiology and pathophysiology. Physiol. Rev. 90, 1383–1435 (2010).

    Article  CAS  Google Scholar 

  24. Sarkar, S. et al. Complex inhibitory effects of nitric oxide on autophagy. Mol. Cell 43, 19–32 (2011).

    Article  CAS  Google Scholar 

  25. Akinc, A. et al. Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based mechanisms. Mol. Ther. 18, 1357–1364 (2010).

    Article  CAS  Google Scholar 

  26. Alabi, C.A. et al. FRET-labeled siRNA probes for tracking assembly and disassembly of siRNA nanocomplexes. ACS Nano 6, 6133–6141 (2012).

    Article  CAS  Google Scholar 

  27. Ikonen, E. Cellular cholesterol trafficking and compartmentalization. Nat. Rev. Mol. Cell Biol. 9, 125–138 (2008).

    Article  CAS  Google Scholar 

  28. Ko, D.C., Gordon, M.D., Jin, J.Y. & Scott, M.P. Dynamic movements of organelles containing Niemann-Pick C1 protein: NPC1 involvement in late endocytic events. Mol. Biol. Cell 12, 601–614 (2001).

    Article  CAS  Google Scholar 

  29. Zhang, M. et al. Cessation of rapid late endosomal tubulovesicular trafficking in Niemann–Pick type C1 disease. Proc. Natl. Acad. Sci. USA 98, 4466–4471 (2001).

    Article  CAS  Google Scholar 

  30. Koivusalo, M., Jansen, M., Somerharju, P. & Ikonen, E. Endocytic trafficking of sphingomyelin depends on its acyl chain length. Mol. Biol. Cell 18, 5113–5123 (2007).

    Article  CAS  Google Scholar 

  31. Lai, S.K., Hida, K., Chen, C. & Hanes, J. Characterization of the intracellular dynamics of a non-degradative pathway accessed by polymer nanoparticles. J. Control. Release 125, 107–111 (2008).

    Article  CAS  Google Scholar 

  32. Linder, M.D. et al. Rab8-dependent recycling promotes endosomal cholesterol removal in normal and sphingolipidosis cells. Mol. Biol. Cell 18, 47–56 (2007).

    Article  CAS  Google Scholar 

  33. Stenmark, H. & Olkkonen, V.M. The Rab GTPase family. Genome Biol. 2, REVIEWSS3007 (2001).

    Article  Google Scholar 

  34. Ostrowski, M. et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 12, 19–30 (2010).

    Article  CAS  Google Scholar 

  35. Hölttä-Vuori, M., Tanhuanpää, K., Möbius, W., Somerharju, P. & Ikonen, E. Modulation of cellular cholesterol transport and homeostasis by Rab11. Mol. Biol. Cell 13, 3107–3122 (2002).

    Article  Google Scholar 

  36. Gibbings, D.J., Ciaudo, C., Erhardt, M. & Voinnet, O. Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nat. Cell Biol. 11, 1143–1149 (2009).

    Article  CAS  Google Scholar 

  37. Lee, Y.S. et al. Silencing by small RNAs is linked to endosomal trafficking. Nat. Cell Biol. 11, 1150–1156 (2009).

    Article  CAS  Google Scholar 

  38. Siomi, H. & Siomi, M.C. RISC hitches onto endosome trafficking. Nat. Cell Biol. 11, 1049–1051 (2009).

    Article  CAS  Google Scholar 

  39. Stalder, L. et al. The rough endoplasmatic reticulum is a central nucleation site of siRNA-mediated RNA silencing. EMBO J. 32, 1115–1127 (2013).

    Article  CAS  Google Scholar 

  40. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    Article  CAS  Google Scholar 

  41. Maere, S., Heymans, K. & Kuiper, M. BiNGO: a Cytoscape plugin to assess overrepresentation of gene ontology categories in biological networks. Bioinformatics 21, 3448–3449 (2005).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We dedicate this work to the memory of MIT police officer Sean Collier who valiantly gave his life for the protection of the MIT community. We would like to thank P. Lobel and L. Huang from Rutgers University for NPC1 primary and immortalized cell lines. We wish to thank W. Salmon and N. Watson of the Whitehead Institute Core facility at MIT for help with confocal imaging. We would also like to thank D. Brown (Harvard MGH), K. Whitehead, R. Bogorad (MIT) and H. Yin (MIT) for healthy discussion. We would also like to thank J. Maraganore (Alnylam) and M. Invernale (MIT) for critical reading of the manuscript. Special thanks to D. Alakhova at University of Nebraska Medical Center for her help with graphic design. We would also like to thank Alnylam Pharmaceuticals, Control release grant EB000244 for funding. This work was supported by the National Heart, Lung, and Blood Institute, US National Institutes of Health, as a Program of Excellence in Nanotechnology (PEN) Award, Contract #HHSN268201000045C.

Author information

Authors and Affiliations

Authors

Contributions

G.S., R.L. and D.G.A. conceived the idea, G.S., W.Q., R.L. and D.G.A. designed research, G.S., W.Q. and C.Z. performed high-throughput microscopy. G.S., C.A., A.E., K.L., D.C. and A.S. designed LNPs and their probes, G.S., C.A., A.E., S.S. and Y.B. performed studies with NPC1 deficient and competent cells, G.S. and S.S. performed studies with autophagy based studies, G.S. and R.Z. performed TIRF microscopy, G.S. and E.K. performed systems biology, G.S., W.Q., R.L. and D.G.A. wrote the manuscript.

Corresponding author

Correspondence to Daniel G Anderson.

Ethics declarations

Competing interests

R.L. is a shareholder and member of the Scientific Advisory Board of Alnylam. D.G.A. is a consultant with Alnylam. R.L and D.G.A have research grants sponsored by Alnylam. W.Q. and C.Z. are employed by Alnylam.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11, Supplementary Table 1 and Supplementary Data (PDF 1842 kb)

Supplementary Movie 1

LNP trafficking at the cell membrane (LNP Alone.avi) (AVI 4104 kb)

Supplementary Movie 2

Effect of proton pump inhibitor on LNP trafficking at the cell membrane (LNP in presence of baf.avi) (AVI 1959 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sahay, G., Querbes, W., Alabi, C. et al. Efficiency of siRNA delivery by lipid nanoparticles is limited by endocytic recycling. Nat Biotechnol 31, 653–658 (2013). https://doi.org/10.1038/nbt.2614

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.2614

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research