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:

Intercellular nanotubes mediate mitochondrial trafficking between cancer and immune cells

Abstract

Cancer progresses by evading the immune system. Elucidating diverse immune evasion strategies is a critical step in the search for next-generation immunotherapies for cancer. Here we report that cancer cells can hijack the mitochondria from immune cells via physical nanotubes. Mitochondria are essential for metabolism and activation of immune cells. By using field-emission scanning electron microscopy, fluorophore-tagged mitochondrial transfer tracing and metabolic quantification, we demonstrate that the nanotube-mediated transfer of mitochondria from immune cells to cancer cells metabolically empowers the cancer cells and depletes the immune cells. Inhibiting the nanotube assembly machinery significantly reduced mitochondrial transfer and prevented the depletion of immune cells. Combining a farnesyltransferase and geranylgeranyltransferase 1 inhibitor, namely, L-778123, which partially inhibited nanotube formation and mitochondrial transfer, with a programmed cell death protein 1 immune checkpoint inhibitor improved the antitumour outcomes in an aggressive immunocompetent breast cancer model. Nanotube-mediated mitochondrial hijacking can emerge as a novel target for developing next-generation immunotherapy agents for cancer.

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: Cancer cells and effector immune cells connect via physical nanotubes.
Fig. 2: Nanotubes mediate organelle transfer between immune cells and cancer cells.
Fig. 3: Metabolic effect of mitochondrial hijacking.
Fig. 4: Mechanism underlying nanotube formation and mitochondrial transfer.
Fig. 5: Targeting nanotube-mediated mitochondrial hijacking augments antitumour immune response in vivo.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding authors upon reasonable request.

References

  1. Wei, S. C. et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 170, 1120–1133.e17 (2017).

    Article  CAS  Google Scholar 

  2. Sharma, P. & Allison, J. P. Dissecting the mechanisms of immune checkpoint therapy. Nat. Rev. Immunol. 20, 75–76 (2020).

    Article  CAS  Google Scholar 

  3. Wolchok, J. Putting the immunologic brakes on cancer. Cell 175, 1452–1454 (2018).

    Article  CAS  Google Scholar 

  4. Kulkarni, A. et al. A designer self-assembled supramolecule amplifies macrophage immune responses against aggressive cancer. Nat. Biomed. Eng. 2, 589–599 (2018).

    Article  CAS  Google Scholar 

  5. Wei, S. C., Duffy, C. R. & Allison, J. P. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 8, 1069–1086 (2018).

    Article  Google Scholar 

  6. Tseng, D. et al. Anti-CD47 antibody–mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response. Proc. Natl Acad. Sci. USA 110, 11103–11108 (2013).

    Article  CAS  Google Scholar 

  7. Sharma, P. & Allison, J. P. The future of immune checkpoint therapy. Science 348, 56–61 (2015).

    Article  CAS  Google Scholar 

  8. Önfelt, B., Nedvetzki, S., Yanagi, K. & Davis, D. M. Cutting edge: membrane nanotubes connect immune cells. J. Immunol. 173, 1511–1513 (2004).

    Article  Google Scholar 

  9. Sowinski, S. et al. Membrane nanotubes physically connect T cells over long distances presenting a novel route for HIV-1 transmission. Nat. Cell Biol. 10, 211–219 (2008).

    Article  Google Scholar 

  10. Gousset, K. et al. Prions hijack tunnelling nanotubes for intercellular spread. Nat. Cell Biol. 11, 328–336 (2009).

    Article  Google Scholar 

  11. Osswald, M. et al. Brain tumour cells interconnect to a functional and resistant network. Nature 528, 93–98 (2015).

    Article  Google Scholar 

  12. Connor, Y. et al. Physical nanoscale conduit-mediated communication between tumour cells and the endothelium modulates endothelial phenotype. Nat. Commun. 6, 8671 (2015).

  13. Rustom, A., Saffrich, R., Markovic, I., Walther, P. & Gerdes, H.-H. Nanotubular highways for intercellular organelle transport. Science 303, 1007–1010 (2004).

    Article  CAS  Google Scholar 

  14. Ahmad, T. et al. Miro1 regulates intercellular mitochondrial transport & enhances mesenchymal stem cell rescue efficacy. EMBO J. 33, 994–1010 (2014).

    CAS  Google Scholar 

  15. Wang, X. & Gerdes, H. H. Transfer of mitochondria via tunneling nanotubes rescues apoptotic PC12 cells. Cell Death Differ. 22, 1181–1191 (2015).

    Google Scholar 

  16. Lu, J. et al. Tunneling nanotubes promote intercellular mitochondria transfer followed by increased invasiveness in bladder cancer cells. Oncotarget 8, 15539–15552 (2017).

    Article  Google Scholar 

  17. Sena, L. A. et al. Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity 38, 225–236 (2013).

    Article  CAS  Google Scholar 

  18. Kumar, A. et al. Enhanced oxidative phosphorylation in NKT cells is essential for their survival and function. Proc. Natl Acad. Sci. USA 116, 7439–7448 (2019).

    Article  CAS  Google Scholar 

  19. Vyas, S., Zaganjor, E. & Haigis, M. C. Mitochondria and cancer. Cell 166, 555–566 (2016).

    Article  CAS  Google Scholar 

  20. Goldman, A. et al. Targeting tumor phenotypic plasticity and metabolic remodeling in adaptive cross-drug tolerance. Sci. Signal. 12, eaas8779 (2019).

    Article  Google Scholar 

  21. Clutton, G., Mollan, K., Hudgens, M. & Goonetilleke, N. A reproducible, objective method using MitoTracker® fluorescent dyes to assess mitochondrial mass in T cells by flow cytometry. Cytometry 95, 450–456 (2019).

    Article  CAS  Google Scholar 

  22. Pham, A. H., McCaffery, J. M. & Chan, D. C. Mouse lines with photo-activatable mitochondria to study mitochondrial dynamics. Genesis 50, 833–843 (2012).

    Article  CAS  Google Scholar 

  23. Pelletier, M., Billingham, L. K., Ramaswamy, M. & Siegel, R. M. in Methods Enzymol, Vol. 542 (eds Galluzzi, L. & Kroemer, G.) 125–149 (Academic Press, 2014).

  24. Kaplon, J. et al. A key role for mitochondrial gatekeeper pyruvate dehydrogenase in oncogene-induced senescence. Nature 498, 109–112 (2013).

    Article  Google Scholar 

  25. Hase, K. et al. M-Sec promotes membrane nanotube formation by interacting with Ral and the exocyst complex. Nat. Cell Biol. 11, 1427–1432 (2009).

    Article  Google Scholar 

  26. Hashimoto, M. et al. Potential role of the formation of tunneling nanotubes in HIV-1 spread in macrophages. J. Immunol. 196, 1832–1841 (2016).

    Article  CAS  Google Scholar 

  27. Moskalenko, S. et al. The exocyst is a Ral effector complex. Nat. Cell Biol. 4, 66–72 (2002).

    Article  CAS  Google Scholar 

  28. Hanna, S. J. et al. The role of Rho-GTPases and actin polymerization during macrophage tunneling nanotube biogenesis. Sci. Rep. 7, 8547 (2017).

    Article  Google Scholar 

  29. Guo, W., Tamanoi, F. & Novick, P. Spatial regulation of the exocyst complex by Rho1 GTPase. Nat. Cell Biol. 3, 353–360 (2001).

    Article  CAS  Google Scholar 

  30. Fransson, Å., Ruusala, A. & Aspenström, P. The atypical Rho GTPases Miro-1 and Miro-2 have essential roles in mitochondrial trafficking. Biochem. Biophys. Res. Commun. 344, 500–510 (2006).

    Article  CAS  Google Scholar 

  31. Glater, E. E., Megeath, L. J., Stowers, R. S. & Schwarz, T. L. Axonal transport of mitochondria requires milton to recruit kinesin heavy chain and is light chain independent. J. Cell Biol. 173, 545–557 (2006).

    Article  CAS  Google Scholar 

  32. Arkwright, P. D. et al. Fas stimulation of T lymphocytes promotes rapid intercellular exchange of death signals via membrane nanotubes. Cell Res. 20, 72–88.

  33. Bustelo, X. R., Sauzeau, V. & Berenjeno, I. M. GTP-binding proteins of the Rho/Rac family: regulation, effectors and functions in vivo. Bioessays 29, 356–370 (2007).

    Article  CAS  Google Scholar 

  34. Majumder, B. et al. Predicting clinical response to anticancer drugs using an ex vivo platform that captures tumour heterogeneity. Nat. Commun. 6, 6169 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work is supported by grants from the National Institute of Health (NIH AR073135_HLJ, CA236702_SS_HLJ, CA214411_SS and CA229772_SS_Co-I), American Lung Association Discovery Grant (LCD-618834_SS) and Department of Defense (DoD PC180355_HLJ and CA201065_HLJ). This work was performed in part at the Center for Nanoscale Systems (CNS), Harvard University, a member of the National Nanotechnology Coordinated Infrastructure Network (NNCI), supported by the National Science Foundation. We thank M. Haigis and A. Goldman for help with the PhAM mouse studies. K.K. is a Gilead Sciences Fellow of the Life Sciences Research Foundation.

Author information

Authors and Affiliations

Authors

Contributions

T.S. conceived the project, designed and executed the experiments, analysed the data, prepared the figures, and contributed to writing the manuscript. C.D. and R.J. helped in designing and executing the in vitro experiments. S.K. and J.M. helped in designing and executing the in vivo studies and analysed results. A.K. performed the genotyping experiment, analysed results and helped write the manuscript. K.K. helped with the Dendra2 mice and Seahorse studies. P.K.M. and A.B. helped with human tumour explant studies and contributed to the manuscript writing. H.L.J. and S.S. conceptualized and supervised the project, and guided the experimental design, data analysis and manuscript writing.

Corresponding authors

Correspondence to Hae Lin Jang or Shiladitya Sengupta.

Ethics declarations

Competing interests

S.S. is a co-founder and owns equity in Vyome Therapeutics, Akamara Therapeutics and Invictus Oncology, and receives fees from Famygen and Advamedica. H.L.J. is a founder and owns equity in Curer. A.B. is involved in the following consulting/advisory boards: Pfizer, Novartis, Genentech, Merck, Radius Health, Immunomedics, Taiho, Sanofi, Daiichi Pharma/AstraZeneca, Puma, Biothernostics, Phillips, Eli Lilly and Foundation Medicine; A.B. is also contracted via research/grant to the following institutions: Genentech, Novartis, Pfizer, Merck, Sanofi, Radius Health, Immunomedics, Daiichi Pharma/AstraZeneca and Natera. T.S., C.D., R.J., S.K., A.K., K.K., J.M. and P.K.M. declare no competing interests.

Additional information

Peer review information Nature Nanotechnology thanks the anonymous reviewers for their contribution to the peer review of this work.

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

Supplementary information

Supplementary Information

Supplementary Figs. 1–29 and Table 1.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Saha, T., Dash, C., Jayabalan, R. et al. Intercellular nanotubes mediate mitochondrial trafficking between cancer and immune cells. Nat. Nanotechnol. 17, 98–106 (2022). https://doi.org/10.1038/s41565-021-01000-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-021-01000-4

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