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:

Treatment of infarcted heart tissue via the capture and local delivery of circulating exosomes through antibody-conjugated magnetic nanoparticles

Abstract

The systemic biodistribution of endogenous extracellular vesicles is central to the maintenance of tissue homeostasis. Here, we show that angiogenesis and heart function in infarcted heart tissue can be ameliorated by the local accumulation of exosomes collected from circulation using magnetic nanoparticles. The nanoparticles consist of a Fe3O4 core and a silica shell that is decorated with poly (ethylene glycol) conjugated through hydrazone bonds to two types of antibody, which bind either to CD63 antigens on the surface of extracellular vesicles or to myosin-light-chain surface markers on injured cardiomyocytes. On application of a local magnetic field, accumulation of the nanoparticles and cleavage of the hydrazone bonds under the acidic pH of injured cardiac tissue lead to the local release of the captured exosomes. In rabbit and rat models of myocardial infarction, the magnetic-guided accumulation of captured CD63-expressing exosomes in infarcted tissue led to reductions in infarct size as well as improved left-ventricle ejection fraction and angiogenesis. The approach could be used to manipulate endogenous exosome biodistribution for the treatment of other diseases.

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: Schematic of the vesicle-shuttle approach.
Fig. 2: Synthesis of GMNPs and their characterization.
Fig. 3: Fabrication and in vitro biofunction of the GMNPEC nanoparticles.
Fig. 4: Dual-targeting ability of GMNPEC in vitro and in vivo.
Fig. 5: Exosome-release characteristics of GMNPEC nanoparticles in vitro and in vivo.
Fig. 6: Injection of GMNPEC improves cardiac functions after MI.
Fig. 7: Systemic injection of GMNPEC improves cardiac functions in a rabbit MI model.

Similar content being viewed by others

Data availability

The main data supporting the results of this study are available within the paper and its Supplementary Information. The data used to make the graphs are provided as Supplementary Information. The raw and analysed datasets generated during the study are too large to be publicly shared, yet they are available for research purposes from the corresponding authors on reasonable request.

References

  1. Thery, C., Zitvogel, L. & Amigorena, S. Exosomes: composition, biogenesis and function. Nat. Rev. Immunol. 2, 569–579 (2002).

    CAS  Google Scholar 

  2. 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).

    CAS  Google Scholar 

  3. Steinbichler, T. B., Dudas, J., Riechelmann, H. & Skvortsova, I. I. The role of exosomes in cancer metastasis. Semin. Cancer Biol. 44, 170–181 (2017).

    CAS  Google Scholar 

  4. Skog, J. et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat. Cell Biol. 10, 1470–1476 (2008).

    CAS  Google Scholar 

  5. Aznar, E. et al. Gated materials for on-command release of guest molecules. Chem. Rev. 116, 561–718 (2016).

    CAS  Google Scholar 

  6. Huo, M., Wang, L., Chen, Y. & Shi, J. Tumor-selective catalytic nanomedicine by nanocatalyst delivery. Nat. Commun. 8, 357 (2017).

    Google Scholar 

  7. Xing, P. & Zhao, Y. Multifunctional nanoparticles self-assembled from small organic building blocks for biomedicine. Adv. Mater. 28, 7304–7339 (2016).

    CAS  Google Scholar 

  8. Chen, X. et al. Light-Induced hydrogel based on tumor-targeting mesoporous silica nanoparticles as a theranostic platform for sustained cancer treatment. ACS Appl. Mater. Inter. 8, 15857–15863 (2016).

    CAS  Google Scholar 

  9. Chen, X. et al. Dual bioresponsive mesoporous silica nanocarrier as an “AND” logic gate for targeted drug delivery cancer cells. Adv. Funct. Mater. 24, 6999–7006 (2014).

    CAS  Google Scholar 

  10. Nahrendorf, M., Pittet, M. J. & Swirski, F. K. Monocytes: protagonists of infarct inflammation and repair after myocardial infarction. Circulation 121, 2437–2445 (2010).

    Google Scholar 

  11. Vicencio, J. M. et al. Plasma exosomes protect the myocardium from ischemia-reperfusion injury. J. Am. Coll. Cardiol. 65, 1525–1536 (2015).

    CAS  Google Scholar 

  12. Sahoo, S. & Losordo, D. W. Exosomes and cardiac repair after myocardial infarction. Circ. Res. 114, 333–344 (2014).

    CAS  Google Scholar 

  13. Giricz, Z. et al. Cardioprotection by remote ischemic preconditioning of the rat heart is mediated by extracellular vesicles. J. Mol. Cell. Cardiol. 68, 75–78 (2014).

    CAS  Google Scholar 

  14. Armstrong, J. P., Holme, M. N. & Stevens, M. M. Re-engineering extracellular vesicles as smart nanoscale therapeutics. ACS Nano 11, 69–83 (2017).

    CAS  Google Scholar 

  15. Bian, S. et al. Extracellular vesicles derived from human bone marrow mesenchymal stem cells promote angiogenesis in a rat myocardial infarction model. J. Mol. Med. 92, 387–397 (2014).

    CAS  Google Scholar 

  16. Gallet, R. et al. Exosomes secreted by cardiosphere-derived cells reduce scarring, attenuate adverse remodelling, and improve function in acute and chronic porcine myocardial infarction. Eur. Heart J. 38, 201–211 (2017).

    CAS  Google Scholar 

  17. Bei, Y. et al. Exercise-induced circulating extracellular vesicles protect against cardiac ischemia-reperfusion injury. Basic Res. Cardiol. 112, 38 (2017).

    Google Scholar 

  18. Cheng, M. et al. Circulating myocardial microRNAs from infarcted hearts are carried in exosomes and mobilise bone marrow progenitor cells. Nat. Commun. 10, 959 (2019).

    Google Scholar 

  19. Li, H. et al. Coronary serum exosomes derived from patients with myocardial ischemia regulate angiogenesis through the miR-939-mediated nitric oxide signaling pathway. Theranostics 8, 2079–2093 (2018).

    CAS  Google Scholar 

  20. Zile, M. R. et al. Relationship between the temporal profile of plasma microRNA and left ventricular remodeling in patients after myocardial infarction. Circ. Cardiovasc. Genet. 4, 614–619 (2011).

    CAS  Google Scholar 

  21. Luther, K. M. et al. Exosomal miR-21a-5p mediates cardioprotection by mesenchymal stem cells. J. Mol. Cell. Cardiol. 119, 125–137 (2018).

    CAS  Google Scholar 

  22. Dong, S. et al. MicroRNA expression signature and the role of microRNA-21 in the early phase of acute myocardial infarction. J. Biol. Chem. 284, 29514–29525 (2009).

    CAS  Google Scholar 

  23. Mathieu, M., Martin-Jaular, L., Lavieu, G. & Thery, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 21, 9–17 (2019).

    CAS  Google Scholar 

  24. Kamerkar, S. et al. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 546, 498–503 (2017).

    CAS  Google Scholar 

  25. Thery, C., Amigorena, S., Raposo, G. & Clayton, A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr. Protoc. Cell Biol. 30, 3.22.1–3.22.29 (2006).

  26. Vandergriff, A. C. et al. Magnetic targeting of cardiosphere-derived stem cells with ferumoxytol nanoparticles for treating rats with myocardial infarction. Biomaterials 35, 8528–8539 (2014).

    CAS  Google Scholar 

  27. Garbern, J. C., Minami, E., Stayton, P. S. & Murry, C. E. Delivery of basic fibroblast growth factor with a pH-responsive, injectable hydrogel to improve angiogenesis in infarcted myocardium. Biomaterials 32, 2407–2416 (2011).

    CAS  Google Scholar 

  28. Lott, J. A. Serum enzyme determinations in the diagnosis of acute myocardial infarction: an update. Hum. Pathol. 15, 706–716 (1984).

    CAS  Google Scholar 

  29. Wolf, R. E. et al. Evaluation of serum creatine kinase and lactate dehydrogenase in experimental myocardial infarction, atriotomies, and thoracotomies. Ann. Thorac. Surg. 41, 378–386 (1986).

    CAS  Google Scholar 

  30. Xu, R., Greening, D. W., Zhu, H. J., Takahashi, N. & Simpson, R. J. Extracellular vesicle isolation and characterization: toward clinical application. J. Clin. Invest. 126, 1152–1162 (2016).

    Google Scholar 

  31. Farooqi, A. A. et al. Exosome biogenesis, bioactivities and functions as new delivery systems of natural compounds. Biotechnol. Adv. 36, 328–334 (2018).

    CAS  Google Scholar 

  32. Peinado, H. et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 18, 883–891 (2012).

    CAS  Google Scholar 

  33. Becker, A. et al. Extracellular vesicles in cancer: cell-to-cell mediators of metastasis. Cancer Cell 30, 836–848 (2016).

    CAS  Google Scholar 

  34. Tran, T. H., Mattheolabakis, G., Aldawsari, H. & Amiji, M. Exosomes as nanocarriers for immunotherapy of cancer and inflammatory diseases. Clin. Immunol. 160, 46–58 (2015).

    CAS  Google Scholar 

  35. Im, H. et al. Label-free detection and molecular profiling of exosomes with a nano-plasmonic sensor. Nat. Biotechnol. 32, 490–495 (2014).

    CAS  Google Scholar 

  36. Shao, H. et al. Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma. Nat. Commun. 6, 6999 (2015).

    CAS  Google Scholar 

  37. Tavallaie, R. et al. Nucleic acid hybridization on an electrically reconfigurable network of gold-coated magnetic nanoparticles enables microRNA detection in blood. Nat. Nanotechnol. 13, 1066–1071 (2018).

    CAS  Google Scholar 

  38. Melo, S. A. et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 523, 177–182 (2015).

    CAS  Google Scholar 

  39. Colao, I. L., Corteling, R., Bracewell, D. & Wall, I. Manufacturing exosomes: a promising therapeutic platform. Trends Mol. Med. 24, 242–256 (2018).

    CAS  Google Scholar 

  40. Alvarez-Erviti, L. et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 29, 341–345 (2011).

    CAS  Google Scholar 

  41. Koppers-Lalic, D., Hogenboom, M. M., Middeldorp, J. M. & Pegtel, D. M. Virus-modified exosomes for targeted RNA delivery; a new approach in nanomedicine. Adv. Drug Deliv. Rev. 65, 348–356 (2013).

    CAS  Google Scholar 

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

    CAS  Google Scholar 

  43. Headland, S. E. et al. Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis. Sci. Transl. Med. 7, 315ra190 (2015).

    Google Scholar 

  44. Zhang, B. et al. HucMSC-exosome mediated-Wnt4 signaling is required for cutaneous wound healing. Stem Cells 33, 2158–2168 (2015).

    CAS  Google Scholar 

  45. Hoshino, A. et al. Tumour exosome integrins determine organotropic metastasis. Nature 527, 329–335 (2015).

    CAS  Google Scholar 

  46. Gutierrez-Vazquez, C., Villarroya-Beltri, C., Mittelbrunn, M. & Sanchez-Madrid, F. Transfer of extracellular vesicles during immune cell-cell interactions. Immunol. Rev. 251, 125–142 (2013).

    Google Scholar 

  47. Fleshner, M. & Crane, C. R. Exosomes, DAMPs and miRNA: features of stress physiology and immune homeostasis. Trends Immunol. 38, 768–776 (2017).

    CAS  Google Scholar 

  48. Kowal, J. et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl Acad. Sci. USA 113, E968–E977 (2016).

    CAS  Google Scholar 

  49. Cheng, K. et al. Magnetic antibody-linked nanomatchmakers for therapeutic cell targeting. Nat. Commun. 5, 4880 (2014).

    CAS  Google Scholar 

  50. Zhang, H. et al. Ultrasmall ferrite nanoparticles synthesized via dynamic simultaneous thermal decomposition for high-performance and multifunctional T1 magnetic resonance imaging contrast agent. ACS Nano 11, 3614–3631 (2017).

    CAS  Google Scholar 

  51. Liu, Q. et al. Genetic targeting of sprouting angiogenesis using Apln-CreER. Nat. Commun. 6, 6020 (2015).

    CAS  Google Scholar 

  52. Kapnisi, M. et al. Auxetic cardiac patches with tunable mechanical and conductive properties toward treating myocardial infarction. Adv. Funct. Mater. 28, 1800618 (2018).

    Google Scholar 

  53. Ferreira, J. C. B. et al. A selective inhibitor of mitofusin 1-βIIPKC association improves heart failure outcome in rats. Nat. Commun. 10, 329 (2019).

    Google Scholar 

  54. Yu, W. et al. GATA4 regulates Fgf16 to promote heart repair after injury. Development 143, 936–949 (2016).

    CAS  Google Scholar 

  55. Ottersbach, A. et al. Improved heart repair upon myocardial infarction: combination of magnetic nanoparticles and tailored magnets strongly increases engraftment of myocytes. Biomaterials 155, 176–190 (2018).

    CAS  Google Scholar 

  56. Qi, H. et al. Blood exosomes endowed with magnetic and targeting properties for cancer therapy. ACS Nano 10, 3323–3333 (2016).

    CAS  Google Scholar 

  57. Mahaffey, K. W., Raya, T. E., Pennock, G. D., Morkin, E. & Goldman, S. Left ventricular performance and remodeling in rabbits after myocardial infarction. Effects of a thyroid hormone analogue. Circulation 91, 794–801 (1995).

    CAS  Google Scholar 

  58. Makkar, R. R. et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet 379, 895–904 (2012).

    Google Scholar 

Download references

Acknowledgements

This work was supported by the National Key Research and Development Program of China (no. 2016YFC1101400 to Y.J.), the National Natural Science Foundation of China (no. 81601606 to X.C. and no. 31670995 to S.L.), the Young Elite Scientist Sponsorship Program by CAST (no. 2017QNRC001 to S.L.), the Fundamental Research Funds for the Central Universities (no. 2016qngz02 to X.C.), the National Natural Science Foundation of Shaanxi Province (no. 2017JM5023 to X.C.), the open fund of the State Key Laboratory of Military Stomatology (no. 2017KA02 to X.C.) and the ARC Centre of Excellence in Convergent Bio-Nano Science and Technology (no. CE140100036 to J.J.G.), and the ARC Laureate Fellowship (no. FL150100060 to J.J.G.).

Author information

Authors and Affiliations

Authors

Contributions

S.L., X.C., L.B. and T.L. contributed to the study design, data acquisition and interpretation. P.Y. and Y.B. characterized the properties of magnetic nanoparticles. X.Y. and X.Q. contributed to data analysis and interpretation. J.X. and F.P. performed the animal experiments. J.J.G. refined the manuscript. S.L., X.C. and Y.J. developed the concept and supervised experiments. All of the authors contributed to writing the manuscript.

Corresponding authors

Correspondence to Xin Chen or Yan Jin.

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 figures.

Reporting Summary

Supplementary Dataset 1

The data used to generate the figures.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, S., Chen, X., Bao, L. et al. Treatment of infarcted heart tissue via the capture and local delivery of circulating exosomes through antibody-conjugated magnetic nanoparticles. Nat Biomed Eng 4, 1063–1075 (2020). https://doi.org/10.1038/s41551-020-00637-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-020-00637-1

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