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.

  • Protocol
  • Published:

A flow cytometry-based protocol for syngenic isolation of neurovascular unit cells from mouse and human tissues

Abstract

The neurovascular unit (NVU), composed of endothelial cells, pericytes, juxtaposed astrocytes and microglia together with neurons, is essential for proper central nervous system functioning. The NVU critically regulates blood–brain barrier (BBB) function, which is impaired in several neurological diseases and is therefore a key therapeutic target. To understand the extent and cellular source of BBB dysfunction, simultaneous isolation and analysis of NVU cells is needed. Here, we describe a protocol for the EPAM-ia method, which is based on flow cytometry for simultaneous isolation and analysis of endothelial cells, pericytes, astrocytes and microglia. This method is based on differential processing of NVU cell types using enzymes, mechanical homogenization and filtration specific for each cell type followed by combining them for immunostaining and fluorescence-activated cell sorting. The gating strategy encompasses cell-type-specific and exclusion markers for contaminating cells to isolate the major NVU cell types. This protocol takes ~6 h for two sets of one or two animals. The isolation part requires experience in animal handling, fresh tissue processing and immunolabeling for flow cytometry. Sorted NVU cells can be used for downstream applications including transcriptomics, proteomics and cell culture. Multiple cell-type analyses using UpSet can then be applied to obtain robust targets from single or multiple NVU cell types in neurological diseases associated with BBB dysfunction. The EPAM-ia method is also amenable to isolation of several other cell types, including cancer cells and immune cells. This protocol is applicable to healthy and pathological tissue from mouse and human sources and to several cell types compared with similar protocols.

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: Overview of EPAM-ia method for the syngenic isolation of neurovascular unit cells from the adult mouse brain.
Fig. 2: Separation of vascular cells from glial cells using nylon mesh filtration for differential processing of NVU cells.
Fig. 3: Final EPAM-ia method gating strategy on single-brain and purity comparison with the control sample of murine stroke model.
Fig. 4: EPAM-ia method-based isolation of NVU and tumor cells applied to an animal model of brain metastasis.
Fig. 5: Representative FACS plots showing the gating strategy for the isolation of the NVU cells as well as immune and tumor cells in brain tissue from mouse glioblastoma model.
Fig. 6: Representative FACS plots showing the gating strategy for the isolation of NVU cells from human brain tissue using the EPAM-ia protocol.
Fig. 7: Analysis of NVU cells isolated with EPAM-ia method for cell-specific and multicellular targeting.
Fig. 8: Schematic for downstream handling and analyses post EPAM-ia method.

Similar content being viewed by others

Zixuan Zhao, Xinyi Chen, … Hanry Yu

Data availability

All the raw data and derived data of this manuscript are available from the corresponding author upon reasonable request. The transcriptomic datasets referred to in this manuscript have been uploaded to NCBI database GSE163752, and also to other servers (https://bioinformatics.mpi-bn.mpg.de/SGD_Stroke, http://vcg.github.io/upset/ followed by pasting the JSON file link https://raw.githubusercontent.com/SGD2020/mcao/master/mcao.json)

References

  1. Liebner, S. et al. Functional morphology of the blood–brain barrier in health and disease. Acta Neuropathol. 135, 311–336 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Sweeney, M. D., Sagare, A. P. & Zlokovic, B. V. Blood–brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 14, 133–150 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Banks, W. A. From blood–brain barrier to blood–brain interface: new opportunities for CNS drug delivery. Nat. Rev. Drug Discov. 15, 275–292 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Terstappen, G. C., Meyer, A. H., Bell, R. D. & Zhang, W. Strategies for delivering therapeutics across the blood–brain barrier. Nat. Rev. Drug Discov. 20, 362–383 (2021).

    Article  CAS  PubMed  Google Scholar 

  5. Janzer, R. C. & Raff, M. C. Astrocytes induce blood–brain barrier properties in endothelial cells. Nature 325, 253–257 (1987).

    Article  CAS  PubMed  Google Scholar 

  6. Abbott, N. J., Rönnbäck, L. & Hansson, E. Astrocyte–endothelial interactions at the blood-brain barrier. Nat. Rev. Neurosci. 7, 41–53 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Daneman, R., Zhou, L., Kebede, A. A. & Barres, B. A. Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature 468, 562–566 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Armulik, A. et al. Pericytes regulate the blood–brain barrier. Nature 468, 557–561 (2010).

    Article  CAS  PubMed  Google Scholar 

  9. da Fonseca, A. C. C. et al. The impact of microglial activation on blood–brain barrier in brain diseases. Front. Cell. Neurosci. 8, 362 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Archie, S. R., Shoyaib, Al,A. & Cucullo, L. Blood–brain barrier dysfunction in CNS disorders and putative therapeutic targets: an overview. Pharmaceutics 13, 1779 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Phoenix, T. N. et al. Medulloblastoma genotype dictates blood brain barrier phenotype. Cancer Cell 29, 508–522 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Cucullo, L. et al. Development of a humanized in vitro blood–brain barrier model to screen for brain penetration of antiepileptic drugs. Epilepsia 48, 505–516 (2007).

    Article  CAS  PubMed  Google Scholar 

  13. Lee, Y.-K., Uchida, H., Smith, H., Ito, A. & Sanchez, T. The isolation and molecular characterization of cerebral microvessels. Nat. Protoc. 14, 3059–3081 (2019).

    Article  CAS  PubMed  Google Scholar 

  14. Devraj, G. et al. HIF-1α is involved in blood–brain barrier dysfunction and paracellular migration of bacteria in pneumococcal meningitis. Acta Neuropathol. 140, 183–208 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Guerit, S. et al. Astrocyte-derived Wnt growth factors are required for endothelial blood-brain barrier maintenance. Prog. Neurobiol. 199, 101937 (2021).

  16. Crouch, E. E. & Doetsch, F. FACS isolation of endothelial cells and pericytes from mouse brain microregions. Nat. Protoc. 13, 738–751 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Lyck, R. et al. Culture-induced changes in blood–brain barrier transcriptome: implications for amino-acid transporters in vivo. J. Cereb. Blood Flow. Metab. 29, 1491–1502 (2009).

    Article  CAS  PubMed  Google Scholar 

  18. Vanlandewijck, M. et al. A molecular atlas of cell types and zonation in the brain vasculature. Nature 554, 475–480 (2018).

  19. Daneman, R. et al. The mouse blood–brain barrier transcriptome: a new resource for understanding the development and function of brain endothelial cells. PLoS ONE 5, e13741 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. He, L. et al. Analysis of the brain mural cell transcriptome. Sci. Rep. 6, 35108 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Winkler, E. A. et al. A single-cell atlas of the normal and malformed human brain vasculature. Science 375, eabi7377 (2022).

  23. Batiuk, M. Y. et al. An immunoaffinity-based method for isolating ultrapure adult astrocytes based on ATP1B2 targeting by the ACSA-2 antibody. J. Biol. Chem. 292, 8874–8891 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kantzer, C. G. et al. Anti-ACSA-2 defines a novel monoclonal antibody for prospective isolation of living neonatal and adult astrocytes. Glia 65, 990–1004 (2017).

    Article  PubMed  Google Scholar 

  25. Wylot, B., Konarzewska, K., Bugajski, L., Piwocka, K. & Zawadzka, M. Isolation of vascular endothelial cells from intact and injured murine brain cortex-technical issues and pitfalls in FACS analysis of the nervous tissue. Cytom. A 87, 908–920 (2015).

    Article  CAS  Google Scholar 

  26. Gurnik, S. et al. Angiopoietin-2-induced blood-brain barrier compromise and increased stroke size are rescued by VE-PTP-dependent restoration of Tie2 signaling. Acta Neuropathol. 131, 753–773 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Liebner, S. et al. Wnt/β-catenin signaling controls development of the blood–brain barrier. J. Cell Biol. 183, 409–417 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bennett, M. L. et al. New tools for studying microglia in the mouse and human CNS. Proc. Natl Acad. Sci. USA 113, E1738–E1746 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhang, Y. et al. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89, 37–53 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Foo, L. C. Purification of rat and mouse astrocytes by immunopanning. Cold Spring Harb. Protoc. 2013, 421–432 (2013).

    Article  PubMed  Google Scholar 

  31. Spitzer, D. et al. Profiling the neurovascular unit unveils detrimental effects of osteopontin on the blood–brain barrier in acute ischemic stroke. Acta Neuropathol. https://doi.org/10.1007/s00401-022-02452-1 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Srinivasan, K. et al. Untangling the brain’s neuroinflammatory and neurodegenerative transcriptional responses. Nat. Commun. 7, 11295 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sundstrøm, T. et al. Automated tracking of nanoparticle-labeled melanoma cells improves the predictive power of a brain metastasis model. Cancer Res. 73, 2445–2456 (2013).

    Article  PubMed  Google Scholar 

  34. Daphu, I. et al. In vitro treatment of melanoma brain metastasis by simultaneously targeting the MAPK and PI3K signaling pathways. Int. J. Mol. Sci. 15, 8773–8794 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Di Tacchio, M. et al. Tumor vessel normalization, immunostimulatory reprogramming, and improved survival in glioblastoma with combined inhibition of PD-1, angiopoietin-2, and VEGF. Cancer Immunol. Res. 7, 1910–1927 (2019).

    Article  PubMed  Google Scholar 

  36. Richardson, G. M., Lannigan, J. & Macara, I. G. Does FACS perturb gene expression? Cytom. A 87, 166–175 (2015).

    Article  Google Scholar 

  37. Grabert, K. et al. Microglial brain region-dependent diversity and selective regional sensitivities to aging. Nat. Neurosci. 19, 504–516 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Jurga, A. M., Paleczna, M. & Kuter, K. Z. Overview of general and discriminating markers of differential microglia phenotypes. Front. Cell. Neurosci. 14, 198 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Haage, V. et al. Comprehensive gene expression meta-analysis identifies signature genes that distinguish microglia from peripheral monocytes/macrophages in health and glioma. Acta Neuropathol. Commun. 7, 20 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Cahoy, J. D. et al. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J. Neurosci. 28, 264–278 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhou, L., Sohet, F. & Daneman, R. Purification and culture of central nervous system endothelial cells. Cold Spring Harb. Protoc. 2014, 44–46 (2014).

    PubMed  Google Scholar 

  42. Navone, S. E. et al. Isolation and expansion of human and mouse brain microvascular endothelial cells. Nat. Protoc. 8, 1680–1693 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Sheikh, B. N. et al. Systematic identification of cell–cell communication networks in the developing brain. iScience 21, 273–287 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Liu, L. & Shi, G.-P. CD31: beyond a marker for endothelial cells. Cardiovasc. Res. 94, 3–5 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Espina, V. et al. Laser-capture microdissection. Nat. Protoc. 1, 586–603 (2006).

    Article  CAS  PubMed  Google Scholar 

  46. Garrido-Gil, P., Fernandez-Rodríguez, P., Rodríguez-Pallares, J. & Labandeira-Garcia, J. L. Laser capture microdissection protocol for gene expression analysis in the brain. Histochem. Cell Biol. 148, 299–311 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. Ståhl, P. L. et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science 353, 78–82 (2016).

    Article  PubMed  Google Scholar 

  48. Vickovic, S. et al. High-definition spatial transcriptomics for in situ tissue profiling. Nat. Methods 16, 987–990 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Brady, L. et al. Inter- and intra-tumor heterogeneity of metastatic prostate cancer determined by digital spatial gene expression profiling. Nat. Commun. 12, 1426 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Devraj, K. et al. GLUT-1 glucose transporters in the blood–brain barrier: differential phosphorylation. J. Neurosci. Res. 89, 1913–1925 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Devraj, K. et al. BACE-1 is expressed in the blood–brain barrier endothelium and is upregulated in a murine model of Alzheimer’s disease. J. Cerebr. Blood Flow Metab. 36, 1281–1294 (2016).

  52. Song, H. W. et al. Transcriptomic comparison of human and mouse brain microvessels. Sci. Rep. 10, 12358 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Leisegang, M. S. et al. Long noncoding RNA MANTIS facilitates endothelial angiogenic function. Circulation 136, 65–79 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Devraj, K., Guerit, S., Macas, J. & Reiss, Y. An in vivo blood–brain barrier permeability assay in mice using fluorescently labeled tracers. JoVE 132, e57038–e57038 (2018).

  55. Lex, A. & Gehlenborg, N. Sets and intersections. Nat. Methods 11, 779 (2014).

    Article  CAS  Google Scholar 

  56. Lex, A., Gehlenborg, N., Strobelt, H., Vuillemot, R. & Pfister, H. UpSet: visualization of intersecting sets. IEEE Trans. Vis. Comput. Gr. 20, 1983–1992 (2014).

    Article  Google Scholar 

  57. Dubinski, D. et al. Controversial roles for dexamethasone in glioblastoma - Opportunities for novel vascular targeting therapies. J. Cereb. Blood Flow. Metab. 39, 1460–1468 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Bernatz, S. et al. Impact of docetaxel on blood–brain barrier function and formation of breast cancer brain metastases. J. Exp. Clin. Cancer Res. 38, 434 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the contributions of B. Yalcin, J. Macas and C. Czupalla with the FACS compensation and troubleshooting of the sorter and for inputs regarding the gating strategy. The authors thank S. Momma and I. Kur for neuronal staining. The authors would like to also thank L. Hansen and N. Lohfink for assistance with mice handling. The authors acknowledge SFB 1039 for funding D.S. and SFB TR23 to Y.R., K.H.P. and S.L., and GO-IN fellowship (291776) for funding Sy.G. and M.D.T., LI 911/5-1 to S.L., EU JUSTBRAIN consortium and Sphingonet consortium of LeducQ foundation for funding K.D. The authors acknowledge LOEWE CePTER consortium for funding Y.R., K.H.P., S.L. and K.D. The authors acknowledge the Edinger Institute for funding all the consumables and softwares with respect to FACS sorting and analysis.

Author information

Authors and Affiliations

Authors

Contributions

D.S., Sy.G., T.P. and K.D. performed tissue dissociation and FACS staining of WT healthy mice and human specimens; J.Z., X.J. and K.S. performed intracardial tumor cell injections and metastasis model characterization by immunohistochemistry; D.S., J.Z. and X.J. performed tissue dissociation and FACS staining for mouse metastasis model; Sy.G. and K.D. performed FACS operation and sorting; M.D.T., Sy.G., and K.D. analyzed and designed the FACS gating strategy with inputs from Y.R. and S.L.; D.S. and M.D.T. performed FACS analysis and prepared FACS plots; D.S., Sy.G., T.P. and M.I.K. performed qPCR analysis; St.G. performed the transcriptomic analysis. Sy.G. created the figures and performed statistics with assistance from K.D.; F.T. established the mouse melanoma model; T.M.F. operated the patients and performed the resection of the biopsies; K.F., K.H.P. and P.N.H. performed dissection and neuropathological analysis of human tissue; Sy.G., Y.R., S.L., M.D.T. and K.D. developed and improved the enzyme combinations and antigen selection. D.S., Sy.G. and K.D. wrote the manuscript with inputs from all the authors. K.D. and Sy.G. designed the overall protocol and also supervised the study.

Corresponding author

Correspondence to Kavi Devraj.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Protocols thanks Eduardo Candelario-Jalil, Elizabeth Crouch 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.

Related links

Key references using this protocol

Spitzer, D. et al. Acta Neuropathol. 144, 305–337 (2022): https://doi.org/10.1007/s00401-022-02452-1

Guérit, S. et al. Prog. Neurobiol. 199, 101937 (2021): https://doi.org/10.1016/j.pneurobio.2020.101937

Devraj, G. et al. Acta Neuropathol. 140, 183–208 (2020): https://doi.org/10.1007/s00401-020-02174-2

Extended data

Extended Data Fig. 1 Assessment of papain concentration in Digestion Mix 1 of the EPAM-ia method on cell yield and viability.

a) Gating strategy for the isolation of endothelial cells, pericytes (mural cells), astrocytes, and microglia from mouse cerebrum digested according to the EPAM-ia protocol with 0.1% of papain in the Digestion Mix 1, representative experiment of n=3. Initial FSC/SSC dot plot show 1,000,000 events. Percentages refer to the proportion of cells in the previous parental gate. (b) Normalized cell events of the four major NVU cell types isolated using different papain concentration in the Digestion Mix 1 (with 0.025%, n=4; 0.05% n=5; 0.1% n=3;). (c) Normalized number of dead cells using different concentrations of papain in the Digestion Mix 1.

Extended Data Fig. 2 Assessment of alternative kit-based Digestion Mix 1 and debris removal solution on cell yield, viability, and purity of the isolated NVU cells.

(a) Schematic indicating with an asterisk the two steps during the cell suspension preparation for which kit based solutions are used. (b) Identical gating strategy as in the fully defined EPAM-ia method is applied to obtained microglia, astrocytes, endothelial cells and pericytes (mural cells) from the same brain tissue sample with the kit based Digestion mix 1 and debris removal steps. The FSC/SSC plot shows 1,000,000 events. Percentages refer to the proportion of cells in the previous parental gate. (c) Normalized cell events of endothelial cells (EC), pericytes (PC), astrocytes (AC) and microglia (MG) isolated by flow cytometry after tissue dissociation using commercially available solutions for the Digestion Mix 1 and the debris removal (n=5). (d) Normalized number of dead cells. (e-h) Purity of kit-based and EPAM-ia method sorted cells is assessed by qPCR targeting cell type-specific markers of endothelial cells (e, Cdh5, Slca1a1), pericytes (f, Kcnj8), astrocytes (g, Aqp4, Slc1a3), and microglia (h, Tmem119). If no amplification was detected, the ∆Ct value is set at 15 by default (n=4). One way ANOVA followed by Dunnett’s multiple comparison test. *, †, § and $ indicate comparison to EC, PC, AC and MG respectively. *p≤0.05, **p≤0.01, ***p≤0.001, ****p≤0.0001 and ns: not significant. Panels a,eh adapted with permission from ref. 31, Springer.

Extended Data Fig. 3 Enrichment analysis of the isolated NVU cell types and the all negative population.

(a) Detailed transcripts reads for representative cell type markers genes of endothelial cells (EC), pericytes (PC), astrocytes (AC) and microglia (MG) in healthy single brain (n=1) or stroke contralateral hemisphere tissue (n=4, 3-4 mice/preparation). (b) Heatmap representing log2 expression of selected choroid plexus and neuronal cell-specific marker genes in endothelial cells, pericytes, astrocytes, microglia and all negative (AN) population isolated from a single mouse cerebrum (n=1). (c) FACS plots showing isolation of NeuN positive neurons by EPAM-ia method. Cells isolated by EPAM-ia method were preselected with ACSA2 magnetic beads followed by brief fixation (0.4% PFA) and permeabilization (0.1% Triton X-100) before immunostaining (n=2). Percentages refer to the proportion of cells in the previous parent gate. First SSC/FSC gate shows 200,000 events.

Supplementary information

Supplementary Information

Supplementary Figs. 1–8.

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

Spitzer, D., Khel, M.I., Pütz, T. et al. A flow cytometry-based protocol for syngenic isolation of neurovascular unit cells from mouse and human tissues. Nat Protoc 18, 1510–1542 (2023). https://doi.org/10.1038/s41596-023-00805-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-023-00805-y

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer