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:

Extracellular vesicles incorporating retrovirus-like capsids for the enhanced packaging and systemic delivery of mRNA into neurons

Abstract

The blood–brain barrier (BBB) restricts the systemic delivery of messenger RNAs (mRNAs) into diseased neurons. Although leucocyte-derived extracellular vesicles (EVs) can cross the BBB at inflammatory sites, it is difficult to efficiently load long mRNAs into the EVs and to enhance their neuronal uptake. Here we show that the packaging of mRNA into leucocyte-derived EVs and the endocytosis of the EVs by neurons can be enhanced by engineering leucocytes to produce EVs that incorporate retrovirus-like mRNA-packaging capsids. We transfected immortalized and primary bone-marrow-derived leucocytes with DNA or RNA encoding the capsid-forming activity-regulated cytoskeleton-associated (Arc) protein as well as capsid-stabilizing Arc 5’-untranslated-region RNA elements. These engineered EVs inherit endothelial adhesion molecules from donor leukocytes, recruit endogenous enveloping proteins to their surface, cross the BBB, and enter the neurons in neuro-inflammatory sites. Produced from self-derived donor leukocytes, the EVs are immunologically inert, and enhanced the neuronal uptake of the packaged mRNA in a mouse model of low-grade chronic neuro-inflammation.

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: Enhanced stability of Arc EVs via the A5U motif.
Fig. 2: Enhanced mRNA loading in EVs and Arc capsids via A5U in donor cells.
Fig. 3: Enhanced and sustained mRNA uptake and translation with A5U in recipient cells.
Fig. 4: eraEVs target and deliver mRNAs for translation in the ageing brain.
Fig. 5: Neuronal mRNA delivery and translation via BM-DC/M–eraEVs in aged brains.

Similar content being viewed by others

Data availability

The data supporting the results in this study are available within the paper and its Supplementary Information. The raw patient data are available from the authors, subject to Institutional Review Board approval. Source data for the figures are provided with this paper. The raw and analysed datasets generated during the study are available for research purposes from the corresponding authors on reasonable request. Source data are provided with this paper.

Code availability

The code for the custom macro generated for this study is provided in Supplementary Information.

References

  1. Dong, X. Current strategies for brain drug delivery. Theranostics 8, 1481–1493 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Pardridge, W. M. Blood-brain barrier and delivery of protein and gene therapeutics to brain. Front. Aging Neurosci. 11, 373 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Sweeney, M. D., Zhao, Z., Montagne, A., Nelson, A. R. & Zlokovic, B. V. Blood-brain barrier: from physiology to disease and back. Physiol. Rev. 99, 21–78 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Varatharaj, A. & Galea, I. The blood-brain barrier in systemic inflammation. Brain Behav. Immun. 60, 1–12 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Nian, K., Harding, I. C., Herman, I. M. & Ebong, E. E. Blood-brain barrier damage in ischemic stroke and its regulation by endothelial mechanotransduction. Front. Physiol. 11, 605398 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Banks, W. A. et al. Transport of extracellular vesicles across the blood-brain barrier: brain pharmacokinetics and effects of inflammation. Int. J. Mol. Sci. 21, 4407 (2020).

  7. Yuan, D. F. et al. Macrophage exosomes as natural nanocarriers for protein delivery to inflamed brain. Biomaterials 142, 1–12 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Haney, M. J. et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J. Control. Release 207, 18–30 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Man, S., Ubogu, E. E. & Ransohoff, R. M. Inflammatory cell migration into the central nervous system: a few new twists on an old tale. Brain Pathol. 17, 243–250 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Seguin, R., Biernacki, K., Rotondo, R. L., Prat, A. & Antel, J. P. Regulation and functional effects of monocyte migration across human brain-derived endothelial cells. J. Neuropathol. Exp. Neurol. 62, 412–419 (2003).

    Article  CAS  PubMed  Google Scholar 

  11. Erickson, M. A. & Banks, W. A. Age-associated changes in the immune system and blood-brain barrier functions. Int. J. Mol. Sci. 20, 1632 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Aslan, C. et al. Exosomes for mRNA delivery: a novel biotherapeutic strategy with hurdles and hope. BMC Biotechnol. 21, 20 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Li, M. et al. Analysis of the RNA content of the exosomes derived from blood serum and urine and its potential as biomarkers. Phil. Trans. R. Soc. Lond. B 369, 20130502 (2014).

  14. Chevillet, J. R. et al. Quantitative and stoichiometric analysis of the microRNA content of exosomes. Proc. Natl Acad. Sci. USA 111, 14888–14893 (2014).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  15. Valadi, H. et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9, 654–659 (2007).

    Article  CAS  PubMed  Google Scholar 

  16. Luan, X. et al. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol. Sin. 38, 754–763 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Usman, W. M. et al. Efficient RNA drug delivery using red blood cell extracellular vesicles. Nat. Commun. 9, 2359 (2018).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  18. Momen-Heravi, F., Bala, S., Bukong, T. & Szabo, G. Exosome-mediated delivery of functionally active miRNA-155 inhibitor to macrophages. Nanomedicine 10, 1517–1527 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Wang, J. H. et al. Anti-HER2 scFv-directed extracellular vesicle-mediated mRNA-based gene delivery inhibits growth of HER2-positive human breast tumor xenografts by prodrug activation. Mol. Cancer Ther. 17, 1133–1142 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Pastuzyn, E. D. et al. The neuronal gene arc encodes a repurposed retrotransposon gag protein that mediates intercellular RNA transfer. Cell 172, 275–288.e18 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Segel, M. et al. Mammalian retrovirus-like protein PEG10 packages its own mRNA and can be pseudotyped for mRNA delivery. Science 373, 882–889 (2021).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  22. Ashley, J. et al. Retrovirus-like Gag Protein Arc1 binds RNA and traffics across synaptic boutons. Cell 172, 262–274.e11 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Comas-Garcia, M., Davis, S. R. & Rein, A. On the selective packaging of genomic RNA by HIV-1. Viruses 8, 246 (2016).

  24. Dynes, J. L. & Steward, O. Arc mRNA docks precisely at the base of individual dendritic spines indicating the existence of a specialized microdomain for synapse-specific mRNA translation. J. Comp. Neurol. 520, 3105–3119 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Fila, M., Diaz, L., Szczepanska, J., Pawlowska, E. & Blasiak, J. mRNA trafficking in the nervous system: a key mechanism of the involvement of activity-regulated cytoskeleton-associated protein (Arc) in synaptic plasticity. Neural Plast. 2021, 3468795 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Paolantoni, C. et al. Arc 3’ UTR splicing leads to dual and antagonistic effects in fine-tuning arc expression upon BDNF signaling. Front. Mol. Neurosci. 11, 145 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Giorgi, C. et al. The EJC factor eIF4AIII modulates synaptic strength and neuronal protein expression. Cell 130, 179–191 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Booth, A. M. et al. Exosomes and HIV Gag bud from endosome-like domains of the T cell plasma membrane. J. Cell Biol. 172, 923–935 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Comas-Garcia, M. et al. Dissection of specific binding of HIV-1 Gag to the ‘packaging signal’ in viral RNA. Elife 6, e27055 (2017).

  30. Brigham, B. S., Kitzrow, J. P., Reyes, J. C., Musier-Forsyth, K. & Munro, J. B. Intrinsic conformational dynamics of the HIV-1 genomic RNA 5’UTR. Proc. Natl Acad. Sci. USA 116, 10372–10381 (2019).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  31. Blakemore, R. J. et al. Stability and conformation of the dimeric HIV-1 genomic RNA 5’UTR. Biophys. J. 120, 4874–4890 (2021).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  32. Carlson, L. A., Bai, Y., Keane, S. C., Doudna, J. A. & Hurley, J. H. Reconstitution of selective HIV-1 RNA packaging in vitro by membrane-bound Gag assemblies. Elife 5, e14663 (2016).

  33. Eriksen, M. S. et al. Arc self-association and formation of virus-like capsids are mediated by an N-terminal helical coil motif. FEBS J. 288, 2930–2955 (2021).

    Article  CAS  PubMed  Google Scholar 

  34. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  35. Rossler, K. et al. Expression of leucocyte adhesion molecules at the human blood-brain barrier (BBB). J. Neurosci. Res. 31, 365–374 (1992).

    Article  CAS  PubMed  Google Scholar 

  36. Granger, D. N. & Senchenkova, E. Inflammation and the Microcirculation (Morgan & Claypool Life Sciences, 2010).

  37. Owens, T., Bechmann, I. & Engelhardt, B. Perivascular spaces and the two steps to neuroinflammation. J. Neuropathol. Exp. Neurol. 67, 1113–1121 (2008).

    Article  PubMed  Google Scholar 

  38. Zozulya, A. L. et al. Dendritic cell transmigration through brain microvessel endothelium is regulated by MIP-1alpha chemokine and matrix metalloproteinases. J. Immunol. 178, 520–529 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. Larochelle, C., Alvarez, J. I. & Prat, A. How do immune cells overcome the blood-brain barrier in multiple sclerosis? FEBS Lett. 585, 3770–3780 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank S. Wilkens from SUNY Upstate Medical University for assistance with TEM imaging; R. Williams, T. Abratte and the BRC Imaging Facility at the Cornell Institute of Biotechnology (RRID:SCR_021741) for imaging experiments, with NIH S10OD025049 for the IVIS-spectrum optical imager in Cornell’s BRC Imaging Facility; P. Schweitzer and the BRC Genomics Facility (RRID:SCR_021727) at the Cornell Institute of Biotechnology for the sequencing experiments; L. Tesfa, J. Mahoney and the BRC Flow Cytometry Facility (RRID:SCR_021740) for flow cytometry data; T. Totman, E. Feldman, F. Burgus and the CARE at Cornell for service and advice on care of animals; R. Felt and the IACUC, Cornell for help composing and managing animal protocols; A. Recknagel for advice on imaging; Herbert Fountain for inspiring discussions about anti-aging therapies; and P. Miller for help in maintaining high-standard laboratory practices. S.J. acknowledges start-up support from Cornell University, including the Robert Langer ’70 Family and Friends Professorship, Cornell NEXT Nano Initiative and Cornell Engineering’s inaugural Sprout Awards.

Author information

Authors and Affiliations

Authors

Contributions

W. Gu, R.L. and S.J. conceptualized the work. W. Gu, S.L., K.L., S.C., N.E., Y.Y., M.C., Y.-W.C., W. Gao and T.S. acquired the data. W. Gu, S.L., S.C., Y.Y., Y.-W.C. and W. Gao contributed to data analyses and to the interpretation of the results. W. Gu, Z.Y., K.S.-G., R.H., P.M., C.W., C. Seo, A.G., C. Schaffer, N.N., R.C., Q.Y., M.W., R.L. and S.J. provided advice, support and supervision. W. Gu wrote the manuscript. W. Gu, C. Schaffer, N.N., R.C., M.W., R.L. and S.J. edited the manuscript.

Corresponding authors

Correspondence to Robert Langer or Shaoyi Jiang.

Ethics declarations

Competing interests

S.J., W. Gu, S.L. and Z.Y. are authors of a patent application related to this work (PCT/US2022/027568) filed by Cornell University. All other authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Koen Breyne, Takahiro Ochiya and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1 Optimization of CMDR dye staining for the quantification of total EV uptake.

To aid in quantifying EVs both in solution and within recipient cells, we employed plasma membrane staining using CMDR. We further refined the number of EVs introduced to recipient cells. The final parameters settled on a 1:5000 CMDR dye concentration and a 2X EV concentration, indicating EVs derived from 2 donor cells (over a span of 24-40 hours) were designated for a single recipient cell.

Extended Data Fig. 2 Density-related expression of the EV markers CD63 and CD81 in capsid+/ stabilizer+ eraEVs.

(a) Western blot analysis illustrating CD63 and Arc expression across various EV subpopulations, differentiated by density through density gradient centrifugation. The 70% sucrose cushion layer—representing the highest density—displayed a marked increase in both CD63 and Arc. Equal amounts of total proteins were loaded across all lanes. Displayed is one representative trial out of three independent experiments. (b) Immunomagnetic positive selection was conducted to isolate CD81+ EVs from filtered supernatant cell culture media laden with both control and engineered EVs. Following isolation, the CD81+ EVs were lysed to facilitate protein quantification and subsequent western blot analysis. (c) Utilizing the Pierce 660 nm Protein Assay, total proteins harvested from CD81+ EVs were quantified. Notably, there was a 2.3-fold upsurge in the protein count extracted from CD81 + /Arc+ engineered EVs in contrast to the capsid‒ control. (d) Western blotting of the CD81 + EV lysates affirmed the presence of Arc proteins. The absence of reducing agents in the sample buffer was deliberate since detection antibodies for CD9/CD63/CD81 often identify the disulfide bond associated with antigen epitopes. Under non-reducing conditions, substantial Arc oligomers are evident. Notably, despite comparable levels of monomer Arc, the quantity of Arc oligomers was significantly amplified in stabilizer+ EVs, underscoring the stabilization effect imparted by the A5U motif. Owing to our exclusion of an Arc knockout cell line, endogenous Arc was discernible in the no-transfection control set. Abbreviations: NT - no transfection control; AA - Arc/A5U-GFP; AG - Arc/GFP. (e-f) Quantitative breakdown depicting the distribution of Arc protein monomers and oligomers in each sample set, as determined through Western blot analysis. The figure presents one representative trial from three independent repeats. For uncropped western gels, refer to SD_ED_FIG2.

Source data

Supplementary information

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gu, W., Luozhong, S., Cai, S. et al. Extracellular vesicles incorporating retrovirus-like capsids for the enhanced packaging and systemic delivery of mRNA into neurons. Nat. Biomed. Eng (2024). https://doi.org/10.1038/s41551-023-01150-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41551-023-01150-x

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