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:

Barcoded viral tracing identifies immunosuppressive astrocyte–glioma interactions

Abstract

Glioblastoma (GBM) is the most lethal primary brain malignancy1. Immunosuppression in the GBM tumour microenvironment (TME) is an important barrier to immune-targeted therapies, but our understanding of the mechanisms of immune regulation in the GBM TME is limited2. Here we describe a viral barcode interaction-tracing approach3 to analyse TME cell–cell communication in GBM clinical samples and preclinical models at single-cell resolution. We combine it with single-cell and bulk RNA-sequencing analyses, human organotypic GBM cultures, in vivo cell-specific CRISPR–Cas9-driven genetic perturbations as well as human and mouse experimental systems to identify an annexin A1–formyl peptide receptor 1 (ANXA1–FPR1) bidirectional astrocyte–GBM communication pathway that limits tumour-specific immunity. FPR1 inhibits immunogenic necroptosis in tumour cells, and ANXA1 suppresses NF-κB and inflammasome activation in astrocytes. ANXA1 expression in astrocytes and FPR1 expression in cancer cells are associated with poor outcomes in individuals with GBM. The inactivation of astrocyte–glioma ANXA1–FPR1 signalling enhanced dendritic cell, T cell and macrophage responses, increasing infiltration by tumour-specific CD8+ T cells and limiting T cell exhaustion. In summary, we have developed a method to analyse TME cell–cell interactions at single-cell resolution in clinical samples and preclinical models, and used it to identify bidirectional astrocyte–GBM communication through ANXA1–FPR1 as a driver of immune evasion and tumour progression.

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: Patient-tissue RABID-seq identifies ANXA1+ astrocytes in human GBM.
Fig. 2: Tumour-anchored RABID-seq detects astrocyte–glioma communication.
Fig. 3: FPR1 expression in GBM suppresses necroptosis and is associated with worse prognosis.
Fig. 4: ANXA1 inhibits astrocyte immune responses.
Fig. 5: Disruption of astrocyte–glioma ANXA1–FPR1 crosstalk enhances anti-tumour immunity.

Similar content being viewed by others

Data availability

Sequencing data have been deposited in the Gene Expression Omnibus (GEO) under the respective SuperSeries accession codes: human data, GSE263612; mouse single cell data, GSE263613; mouse bulk RNA-seq data, GL261 sgFprl versus GL261 sgRandom, GSE263737; AAV sgGFAP-Anxal versus AAV GFAP-sgRosa, GSE263738. Publicly available datasets: scRNA-seq dataset from ref. 11, https://github.com/kpetrecca/NeuroOncology2022.git; spatial transcriptomics datasets from ref. 15, https://datadryad.org/dataset/doi:10.5061/dryad.h70rxwdmj; Ivy Glioblastoma Atlas (ref. 27), http://glioblastoma.alleninstitute.org; TCGA/GLASS, http://www.cbioportal.org; GRCh38 reference genome, https://ftp.ensembl.org/pub/release-113/fasta/homo_sapiens/dna/. All other data are available from the corresponding authors upon reasonable request. Correspondence and request for materials should be addressed to F.J.Q.

Source data are provided with this paper.

References

  1. van den Bent, M. J. et al. Primary brain tumours in adults. Lancet 402, 1564–1579 (2023).

    Article  PubMed  Google Scholar 

  2. Andersen, B. M. et al. Glial and myeloid heterogeneity in the brain tumour microenvironment. Nat. Rev. Cancer 21, 786–802 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Clark, I. C. et al. Barcoded viral tracing of single-cell interactions in central nervous system inflammation. Science 372, eabf1230 (2021).

  4. Mathewson, N. D. et al. Inhibitory CD161 receptor identified in glioma-infiltrating T cells by single-cell analysis. Cell 184, 1281–1298.e26 (2021).

  5. Takenaka, M. C. et al. Control of tumor-associated macrophages and T cells in glioblastoma via AHR and CD39. Nat. Neurosci. 22, 729–740 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kirschenbaum, D. et al. Time-resolved single-cell transcriptomics defines immune trajectories in glioblastoma. Cell 187, 149–165 (2024).

    Article  CAS  PubMed  Google Scholar 

  7. Hara, T. et al. Interactions between cancer cells and immune cells drive transitions to mesenchymal-like states in glioblastoma. Cancer Cell 39, 779–792 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Friebel, E. et al. Single-cell mapping of human brain cancer reveals tumor-specific instruction of tissue-invading leukocytes. Cell 181, 1626–1642 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Lee, H.-G., Wheeler, M. A. & Quintana, F. J. Function and therapeutic value of astrocytes in neurological diseases. Nat. Rev. Drug Discov. 21, 339–358 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sofroniew, M. V. Astrocyte barriers to neurotoxic inflammation. Nat. Rev. Neurosci. 16, 249–263 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Couturier, C. P. et al. Glioblastoma scRNA-seq shows treatment-induced, immune-dependent increase in mesenchymal cancer cells and structural variants in distal neural stem cells. Neuro Oncol. 24, 1494–1508 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Müller, S. et al. Single-cell sequencing maps gene expression to mutational phylogenies in PDGF- and EGF-driven gliomas. Mol. Syst. Biol. 12, 889 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Gan, H. K., Kaye, A. H. & Luwor, R. B. The EGFRvIII variant in glioblastoma multiforme. J. Clin. Neurosci. 16, 748–754 (2009).

    Article  CAS  PubMed  Google Scholar 

  14. Keskin, D. B. et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature 565, 234–239 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Ravi, V. M. et al. Spatially resolved multi-omics deciphers bidirectional tumor-host interdependence in glioblastoma. Cancer Cell 40, 639–655 (2022).

    Article  CAS  PubMed  Google Scholar 

  16. Greenwald, A. C. et al. Integrative spatial analysis reveals a multi-layered organization of glioblastoma. Cell 187, 2485–2501 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Saunders, A. et al. Ascertaining cells’ synaptic connections and RNA expression simultaneously with barcoded rabies virus libraries. Nat. Commun. 13, 6993 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Weinlich, R., Oberst, A., Beere, H. M. & Green, D. R. Necroptosis in development, inflammation and disease. Nat. Rev. Mol. Cell Biol. 18, 127–136 (2017).

    Article  CAS  PubMed  Google Scholar 

  19. Callow, M. G. et al. CRISPR whole-genome screening identifies new necroptosis regulators and RIPK1 alternative splicing. Cell Death Dis. 9, 261 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Levy, A. et al. CD38 deficiency in the tumor microenvironment attenuates glioma progression and modulates features of tumor-associated microglia/macrophages. Neuro Oncol. 14, 1037–1049 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ma, K., Chen, S., Chen, X., Zhao, X. & Yang, J. CD93 is associated with glioma-related malignant processes and immunosuppressive cell infiltration as an inspiring biomarker of survivance. J. Mol. Neurosci. 72, 2106–2124 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yang, F., Zhang, X., Wang, X., Xue, Y. & Liu, X. The new oncogene transmembrane protein 60 is a potential therapeutic target in glioma. Front. Genet. 13, 1029270 (2022).

    Article  CAS  PubMed  Google Scholar 

  23. Mu, L. et al. Pan-cancer analysis of ASB3 and the potential clinical implications for immune microenvironment of glioblastoma multiforme. Front. Immunol. 13, 842524 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lu, B. et al. Basic transcription factor 3 like 4 enhances malignant phenotypes through modulating tumor cell function and immune microenvironment in glioma. Am. J. Pathol. 194, 772–784 (2024).

    Article  CAS  PubMed  Google Scholar 

  25. Ma, K., Chen, S., Chen, X., Yang, C. & Yang, J. S100A10 is a new prognostic biomarker related to the malignant molecular features and immunosuppression process of adult gliomas. World Neurosurg. 165, e650–e663 (2022).

    Article  PubMed  Google Scholar 

  26. Walther, A., Riehemann, K. & Gerke, V. A novel ligand of the formyl peptide receptor: annexin I regulates neutrophil extravasation by interacting with the FPR. Mol. Cell 5, 831–840 (2000).

    Article  CAS  PubMed  Google Scholar 

  27. Puchalski, R. B. et al. An anatomic transcriptional atlas of human glioblastoma. Science 360, 660–663 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Guo, K. et al. A novel necroptosis-related gene signature for predict prognosis of glioma based on single-cell and bulk RNA sequencing. Front. Mol. Biosci. 9, 984712 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sun, L. et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 148, 213–227 (2012).

    Article  CAS  PubMed  Google Scholar 

  30. Chesnokov, M., Khan, I. & Chefetz, I. Induction and detection of necroptotic cell death in mammalian cell culture. Methods Mol. Biol. 2255, 119–134 (2021).

    Article  CAS  PubMed  Google Scholar 

  31. Orozco, S. et al. RIPK1 both positively and negatively regulates RIPK3 oligomerization and necroptosis. Cell Death Differ. 21, 1511–1521 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Iizuka, Y. et al. Identification of a glioma antigen, GARC-1, using cytotoxic T lymphocytes induced by HSV cancer vaccine. Int. J. Cancer 118, 942–949 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Gerke, V. et al. Annexins—a family of proteins with distinctive tastes for cell signaling and membrane dynamics. Nat. Commun. 15, 1574 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Franchi, L. et al. Cytosolic double-stranded RNA activates the NLRP3 inflammasome via MAVS-induced membrane permeabilization and K+ efflux. J. Immunol. 193, 4214–4222 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Li, Z. et al. Formyl peptide receptor 1 signaling potentiates inflammatory brain injury. Sci. Transl. Med. 13, eabe9890 (2021).

  36. Weiß, E. & Kretschmer, D. Formyl-peptide receptors in infection, inflammation, and cancer. Trends Immunol. 39, 815–829 (2018).

    Article  PubMed  Google Scholar 

  37. Sanmarco, L. M. et al. Gut-licensed IFNγ+ NK cells drive LAMP1+TRAIL+ anti-inflammatory astrocytes. Nature 590, 473–479 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wheeler, M. A. et al. MAFG-driven astrocytes promote CNS inflammation. Nature 578, 593–599 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Wheeler, M. A. et al. Environmental control of astrocyte pathogenic activities in CNS inflammation. Cell 176, 581–596 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wheeler, M. A. et al. Droplet-based forward genetic screening of astrocyte-microglia cross-talk. Science 379, 1023–1030 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kiss, M. G. et al. Interleukin-3 coordinates glial-peripheral immune crosstalk to incite multiple sclerosis. Immunity 56, 1502–1514 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Schnell, A. et al. Stem-like intestinal Th17 cells give rise to pathogenic effector T cells during autoimmunity. Cell 184, 6281–6298 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Meiser, P. et al. A distinct stimulatory cDC1 subpopulation amplifies CD8+ T cell responses in tumors for protective anti-cancer immunity. Cancer Cell 41, 1498–1515 (2023).

    Article  CAS  PubMed  Google Scholar 

  44. Bowman-Kirigin, J. A. et al. The conventional dendritic cell 1 subset primes CD8+ T cells and traffics tumor antigen to drive antitumor immunity in the brain. Cancer Immunol. Res. 11, 20–37 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Maas, R. R. et al. The local microenvironment drives activation of neutrophils in human brain tumors. Cell 186, 4546–4566.e27 (2023).

  46. Priego, N. et al. STAT3 labels a subpopulation of reactive astrocytes required for brain metastasis. Nat. Med. 24, 1024–1035 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Heiland, D. H. et al. Tumor-associated reactive astrocytes aid the evolution of immunosuppressive environment in glioblastoma. Nat. Commun. 10, 2541 (2019).

    Article  Google Scholar 

  48. Pepinsky, R. B. et al. Purification and partial sequence analysis of a 37-kDa protein that inhibits phospholipase A2 activity from rat peritoneal exudates. J. Biol. Chem. 261, 4239–4246 (1986).

    Article  CAS  PubMed  Google Scholar 

  49. Wu, C. C. et al. Lipocortin 1 mediates the inhibition by dexamethasone of the induction by endotoxin of nitric oxide synthase in the rat. Proc. Natl Acad. Sci. USA 92, 3473–3477 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ferlazzo, V. et al. Anti-inflammatory effects of annexin-1: stimulation of IL-10 release and inhibition of nitric oxide synthesis. Int. Immunopharmacol. 3, 1363–1369 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Sanches, J. M. et al. Annexin A1 regulates NLRP3 inflammasome activation and modifies lipid release profile in isolated peritoneal macrophages. Cells 9, 926 (2020).

  52. Chao, C.-C. et al. Metabolic control of astrocyte pathogenic activity via cPLA2-MAVS. Cell 179, 1483–1498 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zhou, Y. et al. Formylpeptide receptor FPR and the rapid growth of malignant human gliomas. J. Natl Cancer Inst. 97, 823–835 (2005).

    Article  CAS  PubMed  Google Scholar 

  54. Yatim, N. et al. RIPK1 and NF-κB signaling in dying cells determines cross-priming of CD8+ T cells. Science 350, 328–334 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Han, M. et al. Regulated cell death in glioma: promising targets for natural small-molecule compounds. Front. Oncol. 14, 1273841 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sun, W. et al. Cytosolic calcium mediates RIP1/RIP3 complex-dependent necroptosis through JNK activation and mitochondrial ROS production in human colon cancer cells. Free Radic. Biol. Med. 108, 433–444 (2017).

    Article  CAS  PubMed  Google Scholar 

  57. Nomura, M., Ueno, A., Saga, K., Fukuzawa, M. & Kaneda, Y. Accumulation of cytosolic calcium induces necroptotic cell death in human neuroblastoma. Cancer Res. 74, 1056–1066 (2014).

    Article  CAS  PubMed  Google Scholar 

  58. Venkataramani, V. et al. Glutamatergic synaptic input to glioma cells drives brain tumour progression. Nature 573, 532–538 (2019).

    Article  CAS  PubMed  Google Scholar 

  59. Bagley, S. J. et al. Intrathecal bivalent CAR T cells targeting EGFR and IL13Rα2 in recurrent glioblastoma: phase 1 trial interim results. Nat. Med. 30, 1320–1329 (2024).

    Article  PubMed  Google Scholar 

  60. Lim, M. et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro Oncol. 24, 1935–1949 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Lee, Y., Messing, A., Su, M. & Brenner, M. GFAP promoter elements required for region-specific and astrocyte-specific expression. Glia 56, 481–493 (2008).

    Article  PubMed  Google Scholar 

  62. Hunker, A. C. et al. Conditional single vector CRISPR/SaCas9 viruses for efficient mutagenesis in the adult mouse nervous system. Cell Rep. 30, 4303–4316 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Challis, R. C. et al. Systemic AAV vectors for widespread and targeted gene delivery in rodents. Nat. Protoc. 14, 379–414 (2019).

    Article  CAS  PubMed  Google Scholar 

  64. Opitz, C. A. et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 478, 197–203 (2011).

    Article  CAS  PubMed  Google Scholar 

  65. Gut, G., Herrmann, M. D. & Pelkmans, L. Multiplexed protein maps link subcellular organization to cellular states. Science 361, eaar7042 (2018).

  66. Brennan, C. W. et al. The somatic genomic landscape of glioblastoma. Cell 155, 462–477 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Barthel, F. P. et al. Longitudinal molecular trajectories of diffuse glioma in adults. Nature 576, 112–120 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  PubMed  Google Scholar 

  69. Barbie, D. A. et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature 462, 108–112 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Chu, T., Wang, Z., Pe’er, D. & Danko, C. G. Cell type and gene expression deconvolution with BayesPrism enables Bayesian integrative analysis across bulk and single-cell RNA sequencing in oncology. Nat. Cancer 3, 505–517 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Verhaak, R. G. W. et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17, 98–110 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Maire, C. L. et al. Glioma escape signature and clonal development under immune pressure. J. Clin. Invest. 130, 5257–5271 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  74. Müller, S., Cho, A., Liu, S. J., Lim, D. A. & Diaz, A. CONICS integrates scRNA-seq with DNA sequencing to map gene expression to tumor sub-clones. Bioinformatics 34, 3217–3219 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Zorita, E., Cuscó, P. & Filion, G. J. Starcode: sequence clustering based on all-pairs search. Bioinformatics 31, 1913–1919 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Lun, A. T. L. et al. EmptyDrops: distinguishing cells from empty droplets in droplet-based single-cell RNA sequencing data. Genome Biol. 20, 63 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Griffiths, J. A., Richard, A. C., Bach, K., Lun, A. T. L. & Marioni, J. C. Detection and removal of barcode swapping in single-cell RNA-seq data. Nat. Commun. 9, 2667 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all Quintana lab members for discussion related. This work was supported by grants NS102807, ES02530, ES029136 and AI126880 from the NIH; RG4111A1 and JF2161-A-5 from the NMSS; RSG-14-198-01-LIB from the American Cancer Society; and PA-1604-08459 from the International Progressive MS Alliance. B.M.A. was supported by the Training Program in Nervous System Tumors (K12CA090354) from NCI/NIH; the Career Enhancement Program (CEP) for the NCI/NIH SPORE at Harvard Cancer Center (P50CA165962) from NCI/NIH; the Post-Doctoral Fellowship in Translational Medicine from the PhRMA Foundation; Cancer Neuroscience grant T32CA272386 from NCI/NIH; and 1IK2BX006568-01A1 from the Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development, Biomedical Laboratory Research and Development. C.F.A. was supported by a scholarship from the German Academic Exchange Service (DAAD). M.D. was supported by the Swiss National Science Foundation (project number 199310) and the German Research Foundation (IMM‐PACT‐programme, 413517907). L.F. was funded by the Gemeinnützige Hertie-Stiftung (medMS-Doktorandenprogramm). M.A.W. was supported by NINDS, NIMH, NIDA and NCI (R01MH130458, R00NS114111, T32CA20720, R01MH132632 and R01DA061199). J.-H.L was supported by the Basic Science Research Program funded by the National Research Foundation of Korea (NRF)/Ministry of Education (2022R1A6A3A03071157), and a long-term postdoctoral fellowship funded by the Human Frontier Science Program (LT0015/2023-L). T.I. was supported by an EMBO postdoctoral fellowship (ALTF, 1009-2021). H.-G.L. was supported by a Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2021R1A6A3A14039088). K.L.L. was supported by NCI/NIH SPORE at Harvard Cancer Center P50CA165962. C.M.P. was supported by the National Multiple Sclerosis Society (FG-2307-42209) and the Mayer Foundation. We thank R. Krishnan for technical assistance with flow cytometry and FACS; L. Ding at the Brigham and Women’s Hospital NeuroTechnology Studio (NTS) for technical assistance with confocal microscopy; staff at the NTS for use of their 10× Chromium controller; K. Grieco and S. Valentin, the BWH pathologist assistants and staff at the Tissue and Blood Repository for tissue support.

Author information

Authors and Affiliations

Authors

Contributions

B.M.A., C.F.A., M.A.W., D.A.R., E.A.C. and F.J.Q. designed the study. B.M.A., C.F.A., M.K., J.M.R., A.M., J.E.K., J-H.L., H-G.L., C.M.P., L.G., G.P., T.I., J.J.Y., A.G., J.M., E.N.C., L.S., D.F., L.E.F., A.M.S., L.M.S., F.G., M.C., C.G-V. and R.N. performed or assisted with in vitro and in vivo experiments. K.L.L., E.A.C., D.A.R., M.M.C., E.L., C.C.B. and P.S.P. provided neuropathology expertise, tissue resources and patient models. M.P., M.D. and L.F. performed GBM multiplex immunofluorescence. Z.L. and C.F.A. performed bioinformatic analysis. D.M., J.H.W., and G.G. performed multivariate survival analyses. B.M.A., C.F.A. and F.J.Q. wrote the manuscript with input from co-authors. F.J.Q. directed and supervised the study.

Corresponding author

Correspondence to Francisco J. Quintana.

Ethics declarations

Competing interests

F.J.Q and M.A.W have filed a provisional patent regarding the RABID-seq technology used in this paper. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature thanks Michael Platten 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.

Extended data figures and tables

Extended Data Fig. 1 scRNA-seq, spatial transcriptomic, and ptRABID-seq analysis of astrocytes in GBM.

(a) Biological relevance score of astrocyte DEGs; GBM vs. controls11. (b) Feature plot of ANXA1 in control and GBM scRNA-seq astrocytes. (c) Violin plot of ANXA1 expression by cell type in control and GBM. (d) Volcano plot of DEGs between ANXA1+ and ANXA1- astrocytes in human GBM scRNA-seq. (e) Control and GBM specimen staining of SOX2 to identify malignant cells. (f) Quantification of SOX2+ cells in control and GBM specimens. (n = 4 specimens each; p = 0.0286 by two-sided unpaired Mann-Whitney U-test). (g) ANXA1+ tumor cell (GFAP+ SOX2+) and astrocyte (GFAP+SOX2-) density at the infiltrating tumor zone (ITZ). Each point represents a separate brain, (n = 4 specimens each; p value from Mann-Whitney U-test). (h) ANXA1 staining in astrocytes in n = 5 GBMs in which malignant cells are distinguished by OLIG2 staining. Regions adjacent to tumor core (ITZ) were compared with regions distant from the tumor (leading edge; LE) when available. (i) ANXA1+ astrocyte density at LE vs. ITZ, using OLIG2 as a tumor marker. In total, n = 3,641 GBM-associated astrocytes at the ITZ and n = 1,213 GBM-associated astrocytes at the LE; two-tailed unpaired U-test. (j) ANXA1+ tumor cell (GFAP+ OLIG2+) and astrocyte (GFAP+OLIG2-) density at the ITZ. p = 0.0159 by Mann-Whitney U-test. (k,l) Spatial transcriptomics of the reactive astrocyte gene signature correlated to spatially defined layers programs (k) and to ANXA1 expression (l) in n = 13 GBMs. (m) Dot plot (top) and signature score (bottom) of cell type markers of cells analyzed by ptRABID-seq (n = 1,520 cells). (n) UMAP of CNV analysis (top, n = 3,157 malignant and 1,520 non-malignant cells), and cells grouped by patient (bottom) in ptRABID-seq. (o) Number of barcodes shared between astrocytes and other cell types (top) and divided by patient (bottom), as detected by RABID-seq. (p) Proportions of cell types in ptRABID-seq. (q, r) Average (q) and total (r) barcode numbers detected in each cell type in ptRABID-seq. (s) Shared barcodes between each cell type in ptRABID-seq. Data are mean and error bars show ± SEM (f, g, i, j).

Source Data

Extended Data Fig. 2 Detection of lentivirus transduction and rabies virus infection in human organotypic cultures.

(a) Quantification of GFP+ staining (indicating transduction by the helper lentivirus that delivers TVA and G) as determined by brightfield immunofluorescence detection (each dot represents a separate culture, and n = 6 organotypic cultures per condition from epilepsy surgery (non-GBM)). (b) Quantification of GFP+ cells by cell type. Each slide was co-stained for GFP, DAPI, and one cell type marker. Each dot represents a separate culture, and n = 5 organotypic cultures per condition from epilepsy surgery. (c) Quantification of apoptotic GFP+ cells by cell type using cleaved caspase-3 as a marker of apoptosis. Each slide was co-stained for GFP, one cell type marker, and cleaved caspase 3. Each dot represents a separate culture, and n = 5 organotypic cultures per condition from epilepsy surgery (non-GBM). (d) Quantification of mCherry+ staining (indicating rabies infection) as determined by brightfield immunofluorescence detection. Each dot represents a separate culture, and n = 6 organotypic cultures per condition. (e) Quantification of mCherry+ cells by cell type. Each slide was co-stained for mCherry, DAPI, and one cell type marker. Each dot represents a separate culture, and n = 5 organotypic cultures for astrocytes, n = 3 cultures for other cell types. (f) Quantification of apoptotic mCherry+ cells by cell type using cleaved caspase 3 as a marker of apoptosis. Each slide was co-stained for DAPI, mCherry, one cell type marker, and cleaved caspase 3. Each dot represents a separate culture, and N = 5 organotypic cultures for astrocytes and n = 3 for other cell types. (g-p) Representative images of control and lentivirus transduced cultures. (q-z) Representative images of control and rabies virus-infected cultures. Data are mean and error bars show ± SEM (a-f).

Source Data

Extended Data Fig. 3 Analysis of tumor-anchored RABID-seq.

GL261-TVA-G bearing mice were injected with 1,000 infectious units of rabies one week after tumor implantation; GL261-TVA-G bearing mice were used as controls (n = 5 each). (a-c) Quantification of total (a), cell-type specific (b) mCherry+ staining (indicating rabies infection), and cleaved caspase-3+ staining of mCherry+ cells (c) as determined by brightfield immunofluorescence detection. Each dot represents one animal, and in a, statistics were two sided unpaired t-test. (d) Overview images of control non-rabies injected tumor site (top) and rabies injected tumor site (bottom) from representative animals. Hematoxylin and eosin (H&E) reference images are taken from slices proximate to the immunofluorescence slices for reference purposes. (e-p) Representative images of rabies-injected and control staining by cell type. GFP-negative cells were used as controls when staining for GFAP and OLIG2 to exclude malignant cells. Data are mean and error bars show ± SEM (a-c).

Source Data

Extended Data Fig. 4 Tumor-anchored RABID-seq identifies regulators of glioma-TME interactions.

(a) Representative FACS plot of cells isolated from GL261-anchored RABID-seq one week after infection. (b) Dot plot showing cell type markers used for identification of cell types captured by taRABID-seq (n = 6 mice and 21,189 cells). (c) Cell type proportions by analyzed mouse sample. (d) Matrix showing shared barcodes between each cell type. (e) Anxa1 signature in mouse astrocytes isolated 14 or 21 days after tumor implantation. (f) Volcano plot showing DEGs of GL261 cells highly connected to astrocytes vs. lowly connected to astrocytes, with immunosuppression-related genes of interest labeled. (g) Kaplan-Meier plot of GL261 bearing mice with gRNA targeting Axl or a random gRNA sequence (n = 26 mice sgRandom and n = 16 mice sgAxl; p calculated from log-rank analysis; represents 3 pooled experiments). (h) Scatter plot of gene expression correlation between GL261 connected vs. not connected to activated microglia, by GL261 connected vs. not connected with infiltrating monocyte-derived macrophages.

Source Data

Extended Data Fig. 5 FPR expression in GBM and survival.

(a-c) FPR1 (a), FPR2 (b), and FPR3 (c) in a GBM scRNA-seq dataset11 (n = 25 patients). (d) Dot plot of FPR1 by cell type in GBM scRNA-seq data11. (e) RNA-seq of Fpr1,2,3 in GL261 cells. Each dot represents a mouse or cell culture replicate. n = 3 GL261 bearing mice, n = 3 cell culture replicates per group. (f) FPR1 expression in GBM cells (one dot per patient, and n = 18 patients; statistics by two-way ANOVA with Tukey’s multiple comparisons test; post-hoc unpaired two tailed t-tests were then performed). (g) Fpr1 inactivation in GL261-implanted mice (n = 3 mice per group; each dot represents average staining from a 9 high power field tile scan from one mouse’s tumor; statistics were from two-sided unpaired t-test). (h) Thymidine incorporation assay of GL261 sub-lines cultured for 6 h with thymidine. P values were calculated by unpaired two-sided t-test. (i) Survival of mice bearing CT2A harboring Fpr1 or random sequence gRNA (n = 20 mice per group, statistics were log rank; represents 3 pooled experiments). (j) Survival of mice bearing control GL261 or GL261 with Fpr2 gRNA (n = 20 mice for sgRandom; n = 10 mice for sgFpr2; statistical test by log rank; repeated twice with identical results). (k) Survival of Rag2–/– mice bearing GL261 harboring Fpr1 or random control gRNA (n = 8 mice sgRandom and n = 7 sgFpr1 group; statistics by log rank; repeated twice with identical results). (l) Box and whisker plot of FPR1 expression vs. spatially defined layers programs16. (m) Box and whisker plot of FPR1 correlation to malignant metaprograms16. Boxplots display median, interquartile range (IQR) ± 1.5IQR and outlier values (n) Correlation of FPR1 and ANXA1 expression from the Ivy Glioblastoma Atlas27. (o) Transwell migration assay of GL261 sgRandom vs GL261 sgFpr1; p value from unpaired two-tailed t-test. *** p < 0.001, **p < 0.005. Data are mean and error bars show ± SEM (e-h, o). Illustrations in a created in BioRender (Lee, J. https://BioRender.com/0bkvepw; 2025).

Source Data

Extended Data Fig. 6 Immune response from necroptotic GL261.

(a-b) Immunofluorescence of pMLKL in two human GBM isolates following necroptosis induction in the presence of rhANXA1 and/or an FPR1 neutralizing antibody (each dot represents a separate well, with n = 3 wells per condition; p values were calculated by two-sided unpaired t-test and replicated twice). (c-d) In vitro demonstration of GL261-RIPK3act (c) or GL261-OVA-RIPK3act (d) cell death as measured 48 h after the addition of 50 μM B/B homodimerizer (n = 3 wells per condition; two-sided unpaired t-test; experiment was replicated 3 times). (e-k) Flow cytometry analysis of vehicle (n = 3) vs RIPK3-activated (n = 5 mice) GL261-infiltrating cDC1 (e), cDC2 (f;) subsets, and CD8+ T cells (n = 4 and n = 5 mice) for IFN-γ (g), IL-10 (h), PD1 and TIM3 expression (I); and FOXP3 expression (j) and IL10 expression (k) in the CD4+ population. Unpaired two-tailed t-test (e-k). (l) Schematic for antigen presentation assay in which lysate from RIPK3 activated or control GL261-OVA cells was pulsed onto splenic DCs, and co-cultured with transgenic SIINFEKL-specific OT1 CD8 T cells. (m-o) Mean fluorescence intensities of CD11c+ MHCII+ dendritic cells, I-a/I-e (m), CD80 (n), and CD86 (o). (p) Percent proliferating CD3+ CD8+ T cells from OT1 mice, as measured by Cell Trace dilution, after 72 h of co-culture with lysate-pulsed DCs. (q) Thymidine incorporation assay of co-cultures taken in the final 12 h. (r-w) Flow cytometry of OT1 CD8 T cells from co-culture, displaying upregulation of IL-2 (r), CD25 (s), CD44 (t), IFNγ (u), CD107a (v), and PD1 (w). n = 3 replicates per group, p values from unpaired two tailed t-test for m-w. Data are mean and error bars show ± SEM (a-k; m-w.

Source Data

Extended Data Fig. 7 Investigation of astrocyte ANXA1 signaling.

(a) ELISA quantification in supernatants of primary human astrocytes (n = 3 wells per group; unpaired two-tailed t-test; graph represents one replicate experiment which was performed 3 times with similar results). (b) Representative images from staining of human astrocytes for cleaved IL-1β. (c) Representative images corresponding to Fig. 4j, detecting nuclear NF-κB in human astrocyte-GBM co-cultures. (d-e) Validation of astrocyte-specific Anxa1 knockdown with AAV-PHP.eB and sgRNA. Representative image (d) and automated quantification (e) of ANXA1+ cells of GFAP+ astrocytes (n = 5 sgRosa-transduced and n = 5 sgAnxa1 transduced mice; p values from unpaired two-tailed t-test). (f) Schematic of lentivirus-mediated Anxa1 perturbation study. (g) Kaplan-Meier survival plot of GL261 bearing mice following astrocyte-specific Anxa1 inactivation with lentivirus. Transduction with a random gRNA was used as a control. (n = 9 mice for sgRandom and n = 10 mice for sgAnxa1 group; p values from log rank; experiment was repeated twice with similar results). (h-i) Volcano plot (h) and ingenuity pathway analysis (IPA; i) of astrocytes from sgAnxa1 vs. sgRosa transduced mice (n = 4 sgAnxa1 and n = 3 sgRosa mice per group). (j) Gene set enrichment pathway analysis (GSEA) of astrocyte specific knockdown of Anxa1 showing decreased G protein-coupled receptor signaling in GL261 cells (n = 3 sgRosa and n = 4 sgAnxa1 transduced mice). (k) Representative images of immunofluorescence staining of ASC, GFAP, and DAPI in peritumoral regions from GL261-bearing mice from day 24, corresponding to Fig. 4n. (l) Kaplan-Meier survival plot of GL261 bearing Rag2–/– mice that were pre-treated with AAV-PHP.eB transducing sgAnxa1 or Rosa26 control under the GFAP promoter, as in Fig. 4l,m. (n = 8 mice per group, and p value from log rank; experiment replicated twice). Data are mean and error bars show ± SEM (a, e).

Source Data

Extended Data Fig. 8 Flow gating, TAM RNA-seq in GL261sgFpr1 and AAV PHP-eB-GFAP-sgAnxa1.

(a) Flow cytometry gating scheme. (b,c) Ingenuity pathway analysis on microglia (b) and monocyte-derived macrophages (c), based on differential gene expression between sgFpr1 vs. sg Random GL261 bearing mice (n = 4 mice for sgRandom and n = 5 mice for sgFpr1 group).

Extended Data Fig. 9 Effect on T cells and DCs of Fpr1 perturbation and overexpression in GL261 cells.

(a-d) Analysis of CD4 T-cells in the presence or absence of GL261 cell inactivation of Fpr1. (a) FOXP3+ CD4+ T cells (n = 4 sgRandom and n = 5 sgFpr1 mice), (b) IL-10+ CD4+ T cells (n = 4 sgRandom and n = 5 sgFpr1 mice), (c) FOXP3-IL-10+ CD4+ T cells (n = 4 sgRandom and n = 5 sgFpr1 mice), and (d) IL-2+ CD4+ T cells were quantified (n = 4 sgRandom and n = 5 sgFpr1 mice). (e-h) CD8 T cell suppression upon over-expression of FPR1 in GL261, including tetramer staining for GARC-1 (e; n = 6 control and n = 7 Fpr1 OE mice) and CD107a (f; n = 6 control and n = 7 Fpr1 OE mice), IFN-γ (g; n = 6 control and n = 7 Fpr1 OE mice) and TNF (h; n = 6 control and n = 7 Fpr1 OE mice). (i-n) Conventional (cDC) and monocyte-derived dendritic cell (moDC) quantification and gene expression analysis following GL261 cell inactivation of Fpr1, including cDC1 abundance (i; n = 4 mice per group), pathway analysis (j; n = 3 sgRandom and n = 3 sgFpr1 mice), cDC2 abundance (k; n = 4 sg Random and n = 5 sgFpr1 mice per group) and pathway analysis (l; n = 3 sgRandom and n = 3 sgFpr1 mice), and monocyte-derived DC (moDC) abundance (m; n = 4 sgRandom and n = 6 sgFpr1 mice) and pathway analysis (n; n = 3 sgRandom and n = 3 sgFpr1 mice). (o, p) Exhaustion markers of CD8 T cells from animals receiving sgAnxa1 or sgRosa are shown (n = 10 for sgRosa and n = 7 for sgAnxa1; left, PD-1, and right, TIM-3). (p) contour plot of exhaustion stages of CD8+ T cells. (q-r). Pathway analysis on microglia (q) and monocyte-derived macrophages (r) based on RNA-seq between mice transduced with AAV-PHP.eB sgAnxa1 vs. sgRosa followed by GL261-Luc2 implantation (n = 3 mice per group). All p values were from two-sided unpaired t-test. Data are mean and error bars show ± SEM (a-i; k, m, o).

Source Data

Supplementary information

Supplementary Tables

Table 1: GBM patient characteristics for patient tissue RABID-seq. Table 2: GBM isolates for in vitro mechanistic studies.

Reporting Summary

Supplementary Data

Supplementary Data 1–22.

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

Andersen, B.M., Faust Akl, C., Wheeler, M.A. et al. Barcoded viral tracing identifies immunosuppressive astrocyte–glioma interactions. Nature 644, 1097–1106 (2025). https://doi.org/10.1038/s41586-025-09191-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41586-025-09191-9

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