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:

Characterization of the responses of brain macrophages to focused ultrasound-mediated blood–brain barrier opening

Abstract

The opening of the blood–brain barrier (BBB) by focused ultrasound (FUS) coupled with intravenously injected microbubbles can be leveraged as a form of immunotherapy for the treatment of neurodegenerative disorders. However, how FUS BBB opening affects brain macrophages is not well understood. Here by using single-cell sequencing to characterize the distinct responses of microglia and central nervous system-associated macrophages (CAMs) to FUS-mediated BBB opening in mice, we show that the treatment remodels the immune landscape via the recruitment of CAMs and the proliferation of microglia and via population size increases in disease-associated microglia. Both microglia and CAMs showed early and late increases in population sizes, yet only the proliferation of microglia increased at both timepoints. The population of disease-associated microglia also increased, accompanied by the upregulation of genes associated with gliogenesis and phagocytosis, with the depletion of brain macrophages significantly decreasing the duration of BBB opening.

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: scRNA-seq identifies distinct microglia and CAM clusters.
Fig. 2: Treatment with FUS BBB opening promotes microglia proliferation.
Fig. 3: DAM population increases in size and exhibits an inflammatory signature in response to FUS BBB opening.
Fig. 4: Treatment with FUS BBB opening promotes central nervous system-associated macrophage migration to the area and inflammatory signature.
Fig. 5: Protein analysis confirms brain-macrophage changes.
Fig. 6: Macrophage depletion increases BBB opening volume and duration.
Fig. 7: FUS BBB opening remodels the immune landscape.

Similar content being viewed by others

Data availability

All sequencing data is available in both raw FASTQ and processed-table form on Gene Expression Omnibus via the accession code 22573264. The GRCm38 genome dataset is publicly available at https://www.ncbi.nlm.nih.gov/assembly/GCF_000001635.20.

Code availability

All custom code for analysis is available at https://github.com/ark2173/Seq1-2022.

References

  1. Konofagou, E. et al. Ultrasound-induced blood–brain barrier opening. Curr. Pharm. Biotechnol. 13, 1332–1345 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Stockwell, J. et al. Novel central nervous system drug delivery systems. Chem. Biol. Drug Des. 83, 507–520 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Karakatsani, M. et al. Unilateral focused ultrasound-induced blood–brain barrier opening reduces phosphorylated tau from the rtg4510 mouse model. Theranostics 9, 5396–5411 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Leinenga, G. & Götz, J. Scanning ultrasound removes amyloid-β and restores memory in an Alzheimer’s disease mouse model. Sci. Transl. Med. 7, 278ra33 (2015).

    Article  PubMed  Google Scholar 

  5. Burgess, A. et al. Alzheimer disease in a mouse model: MR imaging-guided focused ultrasound targeted to the hippocampus opens the blood–brain barrier and improves pathologic abnormalities and behavior. Radiology 7, 736–745 (2014).

    Article  Google Scholar 

  6. Shen, Y. et al. Ultrasound with microbubbles improves memory, ameliorates pathology and modulates hippocampal proteomic changes in a triple transgenic mouse model of Alzheimer’s disease. Theranostics 10, 11794–11819 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kugelman, T. et al. Analysis of Focused Ultrasound with Microbubbles Induced BBB Disruption on Tight Junction Morphology (IEEE International Ultrasonics Symposium, 2017).

  8. Ji, R. et al. Cavitation-modulated inflammatory response following focused ultrasound blood–brain barrier opening. J. Control. Release 337, 458–471 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Choi, J., Pernot, M., Small, S. & Konofagou, E. Non-invasive, transcranial and localized opening of the blood–brain barrier using focused ultrasound in mice. Ultrasound Med. Biol. 33, 95–104 (2007).

    Article  PubMed  Google Scholar 

  10. Wu, S. et al. Efficient blood–brain barrier opening in primates with neuronavigation-guided ultrasound and real-time acoustic mapping. Sci. Rep. 8, 7978–7989 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Haut, M. et al. Non-invasive hippocampal blood–brain barrier opening in Alzheimer’s disease with focused ultrasound. Proc. Natl Acad. Sci. USA 117, 9180–9182 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Shin, J. et al. Focused ultrasound-induced blood–brain barrier opening improves adult hippocampal neurogenesis and cognitive function in a cholinergic degeneration dementia rat model. Alzheimers Res. Ther. 11, 1–15 (2019).

    Article  Google Scholar 

  13. Mooney, S. et al. Antidepressant effects of focused ultrasound induced blood–brain barrier opening. Behav. Brain Res. 16, 57–61 (2018).

    Article  Google Scholar 

  14. Choi, J. et al. Non-invasive and transient blood–brain barrier opening in the hippocampus of Alzheimer’s double transgenic mice using focused ultrasound. Ultrason. Imaging 30, 189–200 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Burgess, A. et al. Alzheimer disease in a mouse model: MR imaging–guided focused ultrasound targeted to the hippocampus opens the blood–brain barrier and improves pathologic abnormalities and behavior. Radiology 273, 736–745 (2014).

    Article  PubMed  Google Scholar 

  16. Mooney, S. et al. Focused ultrasound-induced neurogenesis requires an increase in blood–brain barrier permeability. PLoS ONE 11, 0159892 (2016).

    Article  Google Scholar 

  17. McMahon, D., Bendayan, R. & Hynynen, K. Acute effects of focused ultrasound-induced increases in blood–brain barrier permeability on rat microvascular transcriptome. Sci. Rep. 7, 1–15 (2017).

    Article  CAS  Google Scholar 

  18. Downs, M. et al. Long-term safety of repeated blood–brain barrier opening via focused ultrasound with microbubbles in non-human primates performing a cognitive task. PLoS ONE 10, 0125911 (2015).

    Google Scholar 

  19. Wu, Y. et al. Microglia: dynamic mediators of synapse development and plasticity. Trends Immunol. 36, 605–613 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Chintamen, S., Imessadouene, F. & Kernie, S. Immune regulation of adult neurogenic niches in health and disease. Front. Cell. Neurosci. 14, 571071 (2020).

    Article  CAS  PubMed  Google Scholar 

  21. Rogers, J. et al. Cx3cr1 deficiency leads to impairment of hippocampal cognitive function and synaptic plasticity. J. Neurosci. 31, 16241–16250 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pascal, A. et al. Histologic evaluation of activation of acute inflammatory response in a mouse model following ultrasound-mediated blood–brain barrier using different acoustic pressures and microbubble doses. Nanotheranostics 4, 210–223 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Hove, H., Martens, L. & Scheyltjens, I. A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment. Nat. Neurosci. 22, 1021–1035 (2019).

    Article  PubMed  Google Scholar 

  24. Kierdorf, K. et al. Macrophages at CNS interfaces: ontogeny and function in health and disease. Nat. Rev. Neurosci. 20, 547–562 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Sevenich, L. Brain-resident microglia and blood-borne macrophages orchestrate central nervous system inflammation in neurodegenerative disorders and brain cancer. Front. Immunol. 9, 697 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Tan, Y., Yuan, Y. & Tian, L. Microglial regional heterogeneity and its role in the brain. Mol. Psychiatry 25, 351–367 (2020).

    Article  PubMed  Google Scholar 

  27. Keren-Shaul, H. et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169, 1276–1290 (2017).

    Article  CAS  PubMed  Google Scholar 

  28. Li, Q. et al. Developmental heterogeneity of microglia and brain myeloid cells revealed by deep single-cell RNA sequencing. Neuron 101, 207–223.e10 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Masuda, T. et al. Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature 566, 388–392 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Matcovitch-Natan, O. et al. Microglia development follows a stepwise program to regulate brain homeostasis. Science 353, 789 (2016).

    Article  CAS  Google Scholar 

  31. Réu, P. et al. The lifespan and turnover of microglia in the human brain. Cell Rep. 20, 779–784 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Mildner, A. et al. Distinct and non-redundant roles of microglia and myeloid subsets in mouse models of Alzheimer’s disease. J. Neurosci. 31, 11159–11171 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kim, J. et al. A binary cre transgenic approach dissects microglia and CNS border-associated macrophages. Immunity 54, 176–190 (2021).

    Article  CAS  PubMed  Google Scholar 

  34. Goldmann, T. et al. Origin, fate and dynamics of macrophages at central nervous system interfaces. Nat. Immunol. 17, 797–805 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Mrdjen, D. et al. High-dimensional single-cell mapping of central nervous system immune cells reveals distinct myeloid subsets in health, aging, and disease. Immunity 48, 380–395 (2018).

    Article  CAS  PubMed  Google Scholar 

  36. Deczkowska, A. et al. Disease-associated microglia: a universal immune sensor of neurodegeneration. Cell 173, 1073–1081 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. McDonough, A., Lee, R. & Weinstein, J. Microglial interferon signaling and white matter. Neurochem. Res. 42, 2625–2638 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Vukovic, J. et al. Microglia modulate hippocampal neural precursor activity in response to exercise and aging. J. Neurosci. 32, 6435–6443 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hammond, T. et al. Single-cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes. Immunity 50, 253–271 (2019).

    Article  CAS  PubMed  Google Scholar 

  40. Zhao, N. et al. Microglia-targeting nanotherapeutics for neurodegenerative diseases. APL Bioeng. 4, 030902 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhao, R. et al. Phosphatidylserine-microbubble targeting-activated microglia/macrophage in inflammation combined with ultrasound for breaking through the blood–brain barrier. J. Neuroinflammation 15, 334 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Menon, V. Clustering single cells: a review of approaches on high-and low-depth single-cell RNA-seq data. Brief. Funct. Genomics 17, 240–245 (2019).

    Article  Google Scholar 

  44. Jung, S. et al. Analysis of fractalkine receptor cx3cr1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol. Cell. Biol. 20, 4106–4114 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pedragosa, J. et al. CNS-border associated macrophages respond to acute ischemic stroke attracting granulocytes and promoting vascular leakage. Acta Neuropathol. Commun. 6, 76 (2018); https://doi.org/10.1186/s40478-018-0581-6

  46. Rajan, W. et al. Defining molecular identity and fates of CNS-border associated macrophages after ischemic stroke in rodents and humans. Neurobiol. Dis. 137, 104722 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. Zelco, A. et al. Single-cell atlas reveals meningeal leukocyte heterogeneity in the developing mouse brain. Genes & Dev. 35, 1190–1207 (2021); http://www.genesdev.org/cgi/doi/10.1101/gad.348190.120

  48. Han, J., Harris, R. & Zhang, X. An updated assessment of microglia depletion: current concepts and future directions. Mol. Brain 10, 25 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Han, J. et al. Underestimated peripheral effects following pharmacological and conditional genetic microglial depletion. Int. J. Mol. Sci. 21, 8603 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kovacs, Z. et al. Disrupting the blood–brain barrier by focused ultrasound induces sterile inflammation. Proc. Natl Acad. Sci. USA 114, 75–84 (2017).

    Article  Google Scholar 

  51. Willis, E. et al. Repopulating microglia promote brain repair in an IL-6-dependent manner. Cell 180, 833–846 (2020).

    Article  CAS  PubMed  Google Scholar 

  52. Lee, S. et al. Microglia depletion increase brain injury after acute ischemic stroke in aged mice. Exp. Neurol. 336, 113530 (2020).

    PubMed Central  Google Scholar 

  53. Mathew, A., Gorick, C. & Price, R. Single-cell mapping of focused ultrasound-transfected brain. Gene Ther. 30, 255–263 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Mathew, A., Gorick, C. M. & Price, R. J. Multiple regression analysis of a comprehensive transcriptomic data assembly elucidates mechanically- and biochemically-driven responses to focused ultrasound blood-brain barrier disruption. Theranostics 11, 9847–9858 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Leinenga, G. et al. Transcriptional signature in microglia isolated from an Alzheimer’s disease mouse model treated with scanning ultrasound. Bioeng. Transl. Med. 8, e10329 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Eguchi, K. et al. Whole-brain low-intensity pulsed ultrasound therapy markedly improves cognitive dysfunctions in mouse models of dementia—crucial roles of endothelial nitric oxide synthase. Brain Stimul. 11, 959–973 (2018).

    Article  PubMed  Google Scholar 

  57. Bobola, M. et al. Transcranial focused ultrasound, pulsed at 40 hz, activates microglia acutely and reduces aβ load chronically, as demonstrated in vivo. Brain Stimul. 13, 1014–1023 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Blackmore, D. G. et al. Low-intensity ultrasound restores long-term potentiation and memory in senescent mice through pleiotropic mechanisms including NMDAR signaling. Mol. Psychiatry 26, 6975–6991 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Elmore, M. et al. Replacement of microglia in the aged brain reverses cognitive, synaptic, and neuronal deficits in mice. Aging Cell 17, e12832 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Lloyd, A. et al. Central nervous system regeneration is driven by microglia necroptosis and repopulation. Nat. Neurosci. 22, 1046–1052 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wang, S. et al. Microbubble type and distribution dependence of focused ultrasound-induced blood–brain barrier opening. Ultrasound Med. Biol. 40, 103–107 (2014).

    Article  Google Scholar 

  62. Macosko, E. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research was funded by the National Institutes of Health R01AG038961 (EEK), R01NS095803 (SGK) and National Science Foundation DGE-2036197 (RLN).

Author information

Authors and Affiliations

Authors

Contributions

The original scRNA-seq experiments were executed, designed and performed by A.R.K.-S., S.C., S.G.K. and E.E.K., with input from V.M. Bioinformatics analyses were performed by A.R.K.-S. and S.C., with input from V.M. and oversight from E.E.K. S.G.K. and E.E.K. performed the data interpretation. IHC validation experiments were performed by A.R.K.-S., with support from M.J.W., M.R.D., R.L.N., A.J.B. and N.K. Flow cytometry validation experiments were performed by A.R.K.-S., M.J.W. and M.R.D. Manuscript preparation was performed by A.R.K.-S., with input and editing from V.M., S.G.K. and E.E.K. E.E.K., S.Z., S.G.K. and C.-C.W. obtained funding for this work.

Corresponding author

Correspondence to Elisa E. Konofagou.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Evandro Fang, Jurgen Gotz, Rao P. Gullapalli, Kazuyuki Takata and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 figs. 1 and 2.

Reporting Summary

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

Kline-Schoder, A.R., Chintamen, S., Willner, M.J. et al. Characterization of the responses of brain macrophages to focused ultrasound-mediated blood–brain barrier opening. Nat. Biomed. Eng (2023). https://doi.org/10.1038/s41551-023-01107-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41551-023-01107-0

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