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:

CRISPR screens decode cancer cell pathways that trigger γδ T cell detection

Abstract

γδ T cells are potent anticancer effectors with the potential to target tumours broadly, independent of patient-specific neoantigens or human leukocyte antigen background1,2,3,4,5. γδ T cells can sense conserved cell stress signals prevalent in transformed cells2,3, although the mechanisms behind the targeting of stressed target cells remain poorly characterized. Vγ9Vδ2 T cells—the most abundant subset of human γδ T cells4—recognize a protein complex containing butyrophilin 2A1 (BTN2A1) and BTN3A1 (refs. 6,7,8), a widely expressed cell surface protein that is activated by phosphoantigens abundantly produced by tumour cells. Here we combined genome-wide CRISPR screens in target cancer cells to identify pathways that regulate γδ T cell killing and BTN3A cell surface expression. The screens showed previously unappreciated multilayered regulation of BTN3A abundance on the cell surface and triggering of γδ T cells through transcription, post-translational modifications and membrane trafficking. In addition, diverse genetic perturbations and inhibitors disrupting metabolic pathways in the cancer cells, particularly ATP-producing processes, were found to alter BTN3A levels. This induction of both BTN3A and BTN2A1 during metabolic crises is dependent on AMP-activated protein kinase (AMPK). Finally, small-molecule activation of AMPK in a cell line model and in patient-derived tumour organoids led to increased expression of the BTN2A1–BTN3A complex and increased Vγ9Vδ2 T cell receptor-mediated killing. This AMPK-dependent mechanism of metabolic stress-induced ligand upregulation deepens our understanding of γδ T cell stress surveillance and suggests new avenues available to enhance γδ T cell anticancer activity.

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: Vγ9Vδ2 T cell coculture with genome-wide KO library of Daudi cells shows killing-evasion and -enhancement KO.
Fig. 2: Systematic discovery of BTN3A surface expression regulators.
Fig. 3: Metabolic regulation of BTN3A.
Fig. 4: Regulation of BTN2A1 and Vγ9Vδ2 TCR-mediated killing of cancer cells.
Fig. 5: Schematic of regulation of the BTN2A1–BTN3A1–BTN3A2 complex.

Similar content being viewed by others

Data availability

Sequencing datasets for the two screens and CUT&RUN are available in the NCBI Gene Expression Omnibus repository (coculture screen, GSE192828; BTN3A screen, GSE192827; CUT&RUN, GSE226931). Publicly available paired-end IRF1 ChIP–seq fastq files were downloaded from ENCODE68,69: IRF1 K562 IP rep 1 (ENCFF031RWN, ENCFF031WIT), IRF1 K562 IP rep 2 (ENCFF602NSL, ENCFF071PWE) and IRF1 K562 input (ENCFF285JYI, ENCFF420KFM). Also, publicly available TCGA data were utilized for this study (https://www.cancer.gov/tcga). Source data are provided with this paper.

Code availability

Computer code used to analyse the TCGA dataset has been deposited at Zenodo (https://doi.org/10.5281/zenodo.8011891).

References

  1. Silva-Santos, B., Serre, K. & Norell, H. γδ T cells in cancer. Nat. Rev. Immunol. 15, 683–691 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Sebestyen, Z., Prinz, I., Déchanet-Merville, J., Silva-Santos, B. & Kuball, J. Translating gammadelta (γδ) T cells and their receptors into cancer cell therapies. Nat. Rev. Drug Discov. 19, 169–184 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Raverdeau, M., Cunningham, S. P., Harmon, C. & Lynch, L. γδ T cells in cancer: a small population of lymphocytes with big implications. Clin. Transl. Immunol. 8, e01080 (2019).

    Article  Google Scholar 

  4. Silva-Santos, B., Mensurado, S. & Coffelt, S. B. γδ T cells: pleiotropic immune effectors with therapeutic potential in cancer. Nat. Rev. Cancer 19, 392–404 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Mensurado, S., Blanco-Domínguez, R. & Silva-Santos, B. The emerging roles of γδ T cells in cancer immunotherapy. Nat. Rev. Clin. Oncol. 20, 178–191 (2023).

    Article  CAS  PubMed  Google Scholar 

  6. Harly, C. et al. Key implication of CD277/butyrophilin-3 (BTN3A) in cellular stress sensing by a major human γδ T-cell subset. Blood 120, 2269–2279 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Rigau, M. et al. Butyrophilin 2A1 is essential for phosphoantigen reactivity by γδ T cells. Science 367, eaay5516 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Karunakaran, M. M. et al. Butyrophilin-2A1 directly binds germline-encoded regions of the Vγ9Vδ2 TCR and is essential for phosphoantigen sensing. Immunity 52, 487–498 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Groh, V., Steinle, A., Bauer, S. & Spies, T. Recognition of stress-induced MHC molecules by intestinal epithelial γδ T cells. Science 279, 1737–1740 (1998).

    Article  ADS  CAS  PubMed  Google Scholar 

  10. Strid, J., Sobolev, O., Zafirova, B., Polic, B. & Hayday, A. The intraepithelial T cell response to NKG2D-ligands links lymphoid stress surveillance to atopy. Science 334, 1293–1297 (2011).

    Article  ADS  CAS  PubMed  Google Scholar 

  11. Girardi, M. et al. Regulation of cutaneous malignancy by γδ T cells. Science 294, 605–609 (2001).

    Article  ADS  CAS  PubMed  Google Scholar 

  12. Harly, C. et al. Human γδ T cell sensing of AMPK-dependent metabolic tumor reprogramming through TCR recognition of EphA2. Sci. Immunol. 6, eaba9010 (2021).

    Article  CAS  PubMed  Google Scholar 

  13. Marlin, R. et al. Sensing of cell stress by human γδ TCR-dependent recognition of annexin A2. Proc. Natl Acad. Sci. USA 114, 3163–3168 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Chien, Y., Meyer, C. & Bonneville, M. γδ T cells: first line of defense and beyond. Annu. Rev. Immunol. 32, 121–155 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Uhlén, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  PubMed  Google Scholar 

  16. Payne, K. K. et al. BTN3A1 governs antitumor responses by coordinating αβ and γδ T cells. Science 369, 942–949 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sandstrom, A. et al. The intracellular B30.2 domain of butyrophilin 3A1 binds phosphoantigens to mediate activation of human Vγ9Vδ2 T cells. Immunity 40, 490–500 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Mullen, P. J., Yu, R., Longo, J., Archer, M. C. & Penn, L. Z. The interplay between cell signalling and the mevalonate pathway in cancer. Nat. Rev. Cancer 16, 718–731 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Yu, Z. et al. Identification of a transporter complex responsible for the cytosolic entry of nitrogen-containing bisphosphonates. eLife 7, e36620 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Dang, A. T. et al. NLRC5 promotes transcription of BTN3A1-3 genes and Vγ9Vδ2 T cell-mediated killing. iScience 24, 101900 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  21. Corvaisier, M. et al. Vγ9Vδ2 T cell response to colon carcinoma cells. J. Immunol. 175, 5481–5488 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Palakodeti, A. et al. The molecular basis for modulation of human Vγ9Vδ2 T cell responses by CD277/butyrophilin-3 (BTN3A)-specific antibodies. J. Biol. Chem. 287, 32780–32790 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sheftel, A., Stehling, O. & Lill, R. Iron–sulfur proteins in health and disease. Trends Endocrinol. Metab. 21, 302–314 (2010).

    Article  CAS  PubMed  Google Scholar 

  24. Voss, M. et al. Shedding of glycan‐modifying enzymes by signal peptide peptidase‐like 3 (SPPL3) regulates cellular N‐glycosylation. EMBO J. 33, 2890–2905 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Park, J. H. et al. Tumor hypoxia represses γδ T cell-mediated antitumor immunity against brain tumors. Nat. Immunol. 22, 336–346 (2021).

    Article  CAS  PubMed  Google Scholar 

  26. Blevins, M. A., Huang, M. & Zhao, R. The role of CtBP1 in oncogenic processes and its potential as a therapeutic target. Mol. Cancer Ther. 16, 981–990 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Annaert, W. & Kaether, C. Bring it back, bring it back, don’t take it away from me – the sorting receptor RER1. J. Cell Sci. 133, jcs231423 (2020).

    Article  PubMed  Google Scholar 

  28. Cano, C. E. et al. BTN2A1, an immune checkpoint targeting Vγ9Vδ2 T cell cytotoxicity against malignant cells. Cell Rep. 36, 109359 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Vantourout, P. et al. Heteromeric interactions regulate butyrophilin (BTN) and BTN-like molecules governing γδ T cell biology. Proc. Natl Acad. Sci. USA 115, 1039–1044 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Quinlan, K. G. R. et al. Specific recognition of ZNF217 and other zinc finger proteins at a surface groove of C-terminal binding proteins. Mol. Cell. Biol. 26, 8159–8172 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mick, E. et al. Distinct mitochondrial defects trigger the integrated stress response depending on the metabolic state of the cell. eLife 9, e49178 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Herzig, S. & Shaw, R. J. AMPK: guardian of metabolism and mitochondrial homeostasis. Nat. Rev. Mol. Cell Biol. 19, 121–135 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Marcu-Malina, V. et al. Redirecting αβ T cells against cancer cells by transfer of a broadly tumor-reactive γδT-cell receptor. Blood 118, 50–59 (2011).

    Article  CAS  PubMed  Google Scholar 

  34. Gründer, C. et al. γ9 and δ2CDR3 domains regulate functional avidity of T cells harboring γ9δ2TCRs. Blood 120, 5153–5162 (2012).

    Article  PubMed  Google Scholar 

  35. Straetemans, T. et al. GMP-grade manufacturing of T cells engineered to express a defined γδTCR. Front. Immunol. 9, 1062 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Dekkers, J. F. et al. Uncovering the mode of action of engineered T cells in patient cancer organoids. Nat. Biotechnol. 41, 60–69 (2023).

    Article  CAS  PubMed  Google Scholar 

  37. Gassart, A. D. et al. Development of ICT01, a first-in-class, anti-BTN3A antibody for activating Vγ9Vδ2 T cell-mediated antitumor immune response. Sci. Transl. Med. 13, eabj0835 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. Mu, X. et al. Glucose metabolism controls human γδ T-cell-mediated tumor immunosurveillance in diabetes. Cell. Mol. Immunol. 19, 944–956 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Meizlish, M. L., Franklin, R. A., Zhou, X. & Medzhitov, R. Tissue homeostasis and inflammation. Annu. Rev. Immunol. 39, 557–581 (2021).

    Article  CAS  PubMed  Google Scholar 

  40. Warburg, O., Posener, K. & Negelein, E. Über den Stoffwechsel der Tumoren. Biochem. Z. 152, 319–344 (1924).

    Google Scholar 

  41. Heiden, M. G. V., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Article  ADS  Google Scholar 

  42. Tzelepis, K. et al. A CRISPR dropout screen identifies genetic vulnerabilities and therapeutic targets in acute myeloid leukemia. Cell Rep. 17, 1193–1205 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Shifrut, E. et al. Genome-wide CRISPR screens in primary human T cells reveal key regulators of immune function. Cell 175, 1958–1971 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 15, 554 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Ma, Y. et al. CRISPR/Cas9 screens reveal Epstein-Barr virus-transformed B cell host dependency factors. Cell Host Microbe 21, 580–591 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Jiang, S. et al. CRISPR/Cas9‐mediated genome editing in Epstein‐Barr virus‐transformed lymphoblastoid B‐cell lines. Curr. Protoc. Mol. Biol. 121, 31.12.1–31.12.23 (2018).

    CAS  PubMed  Google Scholar 

  47. Mootha, V. K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  48. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  49. Miller, H. E. & Bishop, A. J. R. Correlation AnalyzeR: functional predictions from gene co-expression correlations. BMC Bioinformatics 22, 206 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Lachmann, A. et al. Massive mining of publicly available RNA-seq data from human and mouse. Nat. Commun. 9, 1366 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  51. Concordet, J.-P. & Haeussler, M. CRISPOR: intuitive guide selection for CRISPR/Cas9 genome editing experiments and screens. Nucleic Acids Res. 46, W242–W245 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 10–12 (2011).

    Article  Google Scholar 

  53. Clement, K. et al. CRISPResso2 provides accurate and rapid genome editing sequence analysis. Nat. Biotechnol. 37, 224–226 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Brinkman, E. K., Chen, T., Amendola, M. & van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res. 42, e168 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Davodeau, F. et al. Close correlation between Daudi and mycobacterial antigen recognition by human gamma delta T cells and expression of V9JPC1γ/V2DJCδ-encoded T cell receptors. J. Immunol. 151, 1214–1223 (1993).

    Article  CAS  PubMed  Google Scholar 

  56. Bai, L. et al. The majority of CD1d‐sulfatide‐specific T cells in human blood use a semiinvariant Vδ1 TCR. Eur. J. Immunol. 42, 2505–2510 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sachs, N. et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell 172, 373–386 (2018).

    Article  CAS  PubMed  Google Scholar 

  58. Pleguezuelos‐Manzano, C. et al. Establishment and culture of human intestinal organoids derived from adult stem cells. Curr. Protoc. Immunol. 130, e106 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Zheng, Y., Ahmad, K. & Henikoff, S. CUT&Tag data processing and analysis tutorial. Protocol.io https://doi.org/10.17504/protocols.io.bjk2kkye (2020).

  60. Meers, M. P., Tenenbaum, D. & Henikoff, S. Peak calling by Sparse Enrichment Analysis for CUT&RUN chromatin profiling. Epigenetics Chromatin 12, 42 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kutmon, M., Lotia, S., Evelo, C. T. & Pico, A. R. WikiPathways app for Cytoscape: making biological pathways amenable to network analysis and visualization. F1000Res. 3, 152 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Tsuchiya, S. et al. Implementation of GlycanBuilder to draw a wide variety of ambiguous glycans. Carbohydr. Res. 445, 104–116 (2017).

    Article  CAS  PubMed  Google Scholar 

  64. Varki, A. et al. Symbol nomenclature for graphical representations of glycans. Glycobiology 25, 1323–1324 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Gustavsen, J. A., Pai, S., Isserlin, R., Demchak, B. & Pico, A. R. RCy3: network biology using Cytoscape from within R. F1000Res. 8, 1774 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Martens, M. et al. WikiPathways: connecting communities. Nucleic Acids Res. 49, D613–D621 (2021).

    Article  CAS  PubMed  Google Scholar 

  67. Jiang, P. et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat. Med. 24, 1550–1558 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Dunham, I. et al. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  ADS  CAS  Google Scholar 

  69. Luo, Y. et al. New developments on the Encyclopedia of DNA Elements (ENCODE) data portal. Nucleic Acids Res. 48, D882–D889 (2019).

    Article  PubMed Central  Google Scholar 

  70. Ewels, P. A. et al. The nf-core framework for community-curated bioinformatics pipelines. Nat. Biotechnol. 38, 276–278 (2020).

    Article  CAS  PubMed  Google Scholar 

  71. Lopez-Delisle, L. et al. pyGenomeTracks: reproducible plots for multivariate genomic datasets. Bioinformatics 37, 422–423 (2020).

    Article  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Marson laboratory, I. Jain, S. Dodgson, S. Pyle, T. Tolpa and the Gladstone Flow Cytometry Core for providing valuable input and technical expertise. We also thank B. Gewurz (Harvard Medical School) for sharing Daudi-Cas9 cells. M.R.M. was a Cancer Research Institute (CRI) Irvington Fellow supported by CRI and was supported by the Human Vaccines Project Michelson Prizes for Human Immunology and Vaccine Research funded by the Michelson Medical Research Foundation. J.W.F. was funded by an NIH grant (no. R01HG008140). A.R. was supported by an NIH training grant (no. T32GM007281). M.M.A. is supported by an NSF GRFP grant (no. 2038436). M.O. was supported by Astellas Foundation for Research on Metabolic Disorder, Chugai Foundation for Innovative Drug Discovery Science and Mochida Memorial Foundation for Medicine and Pharmaceutical Research. K.A.T. was supported by the Gladstone PUMAS programme, funded by an NIH grant (no. 5R25HL121037). J.K. and Z.S. were supported by Oncode-PACT and Dutch Cancer Society grant nos. KWF 11393, 12586 and 13043. E.J.A. is funded by an NIH grant (no. R01AI155984). The Marson laboratory has received funds from the CRI Lloyd J. Old STAR grant, The Cancer League, the Innovative Genomics Institute, the Simons Foundation and the Parker Institute for Cancer Immunotherapy. We thank the Hubrecht Organoid Technology for providing patient-derived breast cancer organoids, and J. M. L. Roodhart (Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands) for providing the patient-derived colon cancer organoid. The Gladstone Flow Cytometry Core is supported by the James B. Pendleton Charitable Trust. The schematic in Fig. 3a was adapted from the BioRender ‘Electron Transport Chain’ template. Some of the results shown here are based on data generated by the TCGA Research Network: https://www.cancer.gov/tcga.

Author information

Authors and Affiliations

Authors

Contributions

M.R.M. and A.M. designed the study. M.R.M., S.V., A.R., M.M.A., A.D.M., P.A.C., V.Q.N. and K.A.T. performed experiments and generated essential reagents. J.W.F. performed computational analysis and data visualization. A.D.S. and M.O. performed TCGA data analysis and visualization. K.H. generated pathway enrichment visualization. A.C.R. helped in obtaining patient-derived tumour organoids. E.J.A., J.K., Z.S. and J.K.P. helped design assays and interpret results. M.R.M. and A.M. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Murad R. Mamedov or Alexander Marson.

Ethics declarations

Competing interests

A.M. is a cofounder of Arsenal Biosciences, Spotlight Therapeutics and Survey Genomics; serves on the boards of directors at Spotlight Therapeutics and Survey Genomics; is a board observer (and former member of the board of directors) at Arsenal Biosciences; is a member of the scientific advisory boards of Arsenal Biosciences, Spotlight Therapeutics, Survey Genomics, NewLimit, Amgen, Lightcast and Tenaya; owns stock in Arsenal Biosciences, Spotlight Therapeutics, NewLimit, Survey Genomics, PACT Pharma, Lightcast, and Tenaya; and has received fees from Arsenal Biosciences, Spotlight Therapeutics, NewLimit, Survey Genomics, Tenaya, Lightcast, 23andMe, PACT Pharma, Juno Therapeutics, Trizell, Vertex, Merck, Amgen, Genentech, AlphaSights, Rupert Case Management, Bernstein and ALDA. A.M. is an investor in, and informal advisor to, Offline Ventures and a client of EPIQ. J.W.F. was a consultant for NewLimit, is an employee of Genentech and has equity in Roche. The Marson laboratory has received research support from Juno Therapeutics, Epinomics, Sanofi, GlaxoSmithKline, Gilead and Anthem. J.K. is a shareholder of Gadeta B.V. J.K. and Z.S. are inventors on patents with γδTCR-related topics. A.M. and M.R.M. are inventors on patent applications that have been filed based on the findings described here.

Peer review

Peer review information

Nature thanks Dmitry Gabrilovich, Thomas Herrmann 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 Mevalonate pathway effects, co-culture screen consistency, and gene set enrichment analysis (GSEA).

(a) Schematic of the mevalonate pathway, adapted from WikiPathways. Phosphoantigens highlighted in blue. (b) Survival of eGFP+ Daudi cells co-cultured with primary Vγ9Vδ2 T cells at different effector-to-target (E:T) ratios with or without zoledronate (ZOL). Cells were quantified using real-time quantitative live-cell imaging (Incucyte). Survival was normalized to Daudi cells cultured without T cells. Mean ± SD. n = 3 per condition. (ce) Pairwise comparisons of log2(fold change [FC]) of screen results among the three healthy human PBMC donors. (f) Number of genes contained within each negatively enriched KEGG gene set after filtering out genes that were not in the screen dataset, FDR q-values, and (g) number of genes found in two KEGG gene sets.

Source data

Extended Data Fig. 2 Gene, gene set, and signature TCGA analysis.

(a) Heatmap of the hazard ratios (natural log-transformed) associated with the co-culture screen gene signature in TCGA patients for 33 cancer types (a positive log-ratio indicates a worse prognosis and a negative one indicates a protective effect of the gene signature). The co-culture screen gene signature was scaled to mean = 0, SD = 1. Values shown only for cancer types with significant survival and signature association in patient tumours, as determined by a Wald test with Benjamini-Hochberg multiple comparisons correction (two-sided padj < 0.05). (b, d) Correlation of tumour gene expression and survival in the low-grade glioma (LGG) patient cohort, (b) with the entire cohort and (d) with the cohort split according to TRGV9/TRDV2 tumour transcript abundance. Patients with high and low expression of every given gene were compared across the 1040 genes in the co-culture screen signature. Positive Wald test Z score indicates a positive correlation with survival, and negative Z score indicates a negative correlation with survival. (c, e) Correlation of KEGG pathway-derived and type I interferon response pathway-derived signature scores and survival in the low-grade glioma (LGG) patient cohort, (c) with the entire cohort and (e) with the cohort split according to TRGV9/TRDV2 tumour transcript abundance. Patients with high and low pathway signature scores were compared. (f, g) Survival of (f) all LGG patients and (g) TRGV9/TRDV2-high or TRGV9/TRDV2-low LGG patients split by high and low expression of the TCA cycle pathway signature. (h, i) Survival of TRAC/TRBC-high/low (h) LGG and (i) BLCA patients split by high and low expression of the co-culture screen gene signature. (b-e) Significance was determined by a Wald test (Cox regression) with Bonferroni multiple comparisons correction (two-sided padj < 0.05). (f-i) Log-rank test (Kaplan-Meier survival analysis), adjusted (padj) with Benjamini-Hochberg multiple comparisons correction. (c, e-g) Pathway signature levels were estimated by limiting the comparison to genes that overlapped between the co-culture screen hits signature and the pathway.

Extended Data Fig. 3 Correlation of expression between BTN3A1 and screen hits across thousands of samples.

(a) BTN3A1 expression correlation with gene ontology (GO) pathways across thousands of healthy samples (all tissues combined) collated by Correlation AnalyzeR. To determine pathways that correlate with BTN3A1, genome-wide Pearson correlations for BTN3A1 are used as a ranking metric in the GSEA algorithm, which determines the padj-value. (b) Pairwise correlations between expression of BTN3A1 and shown genes across thousands of healthy samples (all tissues combined) (Correlation AnalyzeR). (c) Each vertical line indicates the correlation between expression of BTN3A1 and one of the genes in the KEGG oxidative phosphorylation (OXPHOS) gene set, overlaid over a density plot of the BTN3A1 pairwise correlations with genes across the entire human genome. Data from healthy immune tissues (Correlation AnalyzeR).

Extended Data Fig. 4 BTN3A expression screen consistency, cross-screen correlation, FDPS validation, and GSEA.

(ac) Pairwise comparisons of significant (FDR < 0.01) and not significant results among the three replicates (Rep) of Daudi-Cas9 cell populations used for the BTN3A expression screen. (d) Correlation of screen effect sizes (LFC) among concordant hits separated into positive and negative regulators of BTN3A surface expression. Linear regression line with a 95% confidence interval is shown. (e) Surface BTN3A staining on live Daudi-Cas9 cells treated for 72 hours with zoledronate (ZOL), an inhibitor of FDPS. n = 3 per ZOL dose, representative data from one of three independent experiments. One-way ANOVA comparison to no treatment with Dunnett’s multiple comparisons test. (f) Surface BTN3A staining on live Daudi-Cas9 FDPS KO or control AAVS1 KO cells at indicated days after lentiviral sgRNA transduction. n = 4 for each KO. One-way ANOVA comparison to Daudi-Cas9 AAVS1 (#5) KO cells with Dunnett’s multiple comparisons test. Data from one experiment. (e, f) Mean ± SD. p < 0.0001 (****), p < 0.001 (***), p < 0.01 (**). (g) GSEA of KEGG gene sets that positively or negatively regulate surface BTN3A expression. Number of genes contained within each KEGG gene set after filtering out genes that were not in the screen dataset and FDR q-values are shown.

Source data

Extended Data Fig. 5 Pathway Enrichment Visualization.

(ad) Schematics of the depletion and enrichment of KOs within the (a) oxidative phosphorylation, (b) iron-sulphur (Fe-S) cluster biogenesis, (c) N-glycan biosynthesis, (d) and sialylation pathways across both screens. Shading indicating log2FC shown only for significant hits (FDR < 0.05). All pathways were adapted from WikiPathways. (a) OXPHOS subunits are shown with abridged names without accompanying prefixes (C I: NDUF; C II: SDH; C III: UQCR, C IV: COX; C V: ATP5) (e.g., B4 in CI is NDUFB4). Subunits encoded by mitochondrial genes are not included in the visualization.

Extended Data Fig. 6 BTN3A expression screen gene signature.

(a) Heatmap of the hazard ratios (natural log-transformed) associated with the BTN3A screen gene signature in TCGA patients for 33 cancer types (a positive log-ratio indicates a worse prognosis and a negative one indicates a protective effect of the gene signature). The BTN3A screen gene signature was scaled to mean = 0, SD = 1. Values shown only for cancer types with significant survival and signature association in patient tumours, as determined by a Wald test with Benjamini-Hochberg multiple comparisons correction (two-sided padj < 0.05). (b-d) Survival of (b) total, (c) TRGV9/TRDV2-high/low, or (d) TRAC/TRBC-high/low LGG patients split by high and low expression of the BTN3A expression screen gene signature. Log-rank test (Kaplan-Meier survival analysis) and Wald test (Cox regression), adjusted (padj) with Benjamini-Hochberg multiple comparisons correction.

Extended Data Fig. 7 Effects of screen hit KOs on Vγ9Vδ2 TCR tetramer staining and BTN3A2 and BTN2A1 expression.

(a) Representative histograms of surface BTN3A fluorescence for a subset of single gene Daudi-Cas9 KOs and the AAVS1 control. (b) G115 clone Vγ9Vδ2 TCR tetramer staining fluorescence (MFI) at 14 days after lentiviral sgRNA transduction. Data from one experiment. AAVS1 KO n = 12, BTN3A1 KO n = 3, all other deletions n = 6. (c, d) qPCR data for (c) BTN3A2 and (d) BTN2A1 transcripts normalized to ACTB transcripts. n = 5-6 (except RER (#1) KO n = 4 for BTN2A1), AAVS1 KO n = 12, data combined from two independent experiments. (b-d) One-way ANOVA with Dunnett’s multiple comparisons test. Mean ± SD. p < 0.0001 (****), p < 0.001 (***), p < 0.01 (**), p < 0.05 (*).

Source data

Extended Data Fig. 8 IRF1 and ZNF217 CUT&RUN, ChIP-Seq, and Vγ9Vδ2 T cell killing.

(a) Publicly available IRF1 ChIP-Seq for the butyrophilin locus in K562 cells stably expressing C-terminal eGFP-tagged IRF1 (ENCODE). (bd) CUT&RUN data for IRF1 and ZNF217 binding at promoters in (b) BTN3A1, (c) BTN3A2, and (d) BTN3A3 loci in WT Daudi-Cas9 cells. n = 3 per condition. The algorithm SEACR calls peaks and verifies them above a stringent background signal threshold. (e) Daudi-Cas9 KO survival after 24-hour co-culture with expanded Vγ9Vδ2 T cells in the presence of ZOL at an E:T ratio of 2:1. For each γδ T cell donor, Daudi survival is calculated relative to Daudi cells cultured without T cells and normalized to the Daudi-Cas9 AAVS1 KO control cell survival. Combined data from three donors and two independent experiments. AAVS1 KO n = 6, IRF1 KO n = 3. One-way ANOVA comparison to AAVS1 KO cells with Dunnett’s multiple comparisons test. Mean ± SD. p < 0.01 (**).

Source data

Extended Data Fig. 9 Metabolic effects on surface BTN3A expression.

(a) Surface BTN3A MFI in Daudi-Cas9 KOs cultured in different pyruvate concentrations for 3 days in RPMI (no glucose, no pyruvate). Normalized to cells grown without pyruvate (0 mM). (b-d, f) Surface BTN3A MFI in Daudi-Cas9 cells treated for 72 h with (b) an mTOR inhibitor (rapamycin), an ISR inhibitor (ISRIB), ISR agonists (guanabenz, Sal003, salubrinal, raphin1, sephin1), and DMSO (vehicle) (KO cells); (c) metformin (WT cells); (d) A-769662 compared to equivalent amounts of DMSO (vehicle) (WT cells); or (f) the shown compounds (KO cells). (e) Surface BTN3A MFI in WT Daudi-Cas9 cells co-treated with AICAR and increasing amounts of Compound C (AMPK inhibitor) or DMSO (vehicle). (a) n = 4 per condition (n = 3, TIMMDC1 (#2) at 0 mM), data combined from two independent experiments, each individually normalized. (b) n = 6 per condition (except n = 5 for AAVS1 (#5) with guanabenz and for PPAT (#1) with salubrinal), data combined from two independent experiments, each individually normalized to DMSO (vehicle)-treated cells. (c) n = 8 per condition, data combined from two independent experiments. One-way ANOVA comparison to cells that received no treatment with Dunnett’s multiple comparisons test. (d) n = 3 per condition, representative data from one of two independent experiments. (e) n = 3 per conditions, representative data from one of two independent experiments. Two-tailed unpaired Student’s t test with Bonferroni correction. (f) n = 3 per condition (n = 2 for AMPKα1 (#1) treated with DMSO), representative data from one of two independent experiments. One-way ANOVA comparison to AAVS1 (#5) KO cells with Dunnett’s multiple comparisons test. (a-f) Mean ± SD. p < 0.0001 (****), p < 0.001 (***), p < 0.01 (**), p < 0.05 (*), p > 0.05 (N.S.).

Source data

Extended Data Fig. 10 Isotype Control Staining, qPCR, and BTN3A blocking.

(a) G115 clone Vγ9Vδ2 TCR tetramer staining MFI of WT Daudi-Cas9 cells treated with 80 µM C991 (DMSO), DMSO (vehicle), 0.5 mM AICAR (aqueous), or without treatment for 72 h. Two-tailed unpaired Student’s t test. (b) Vγ4Vδ1 TCR (clone DP10.7) tetramer staining fluorescence (MFI) of Daudi-Cas9 KO cells treated with 80 µM C991 (DMSO), DMSO (vehicle), 0.5 mM AICAR (aqueous), or water for 72 hours. This staining with a tetramer of an irrelevant γδTCR clone defines the background for Vγ9Vδ2 TCR tetramer staining in Fig. 4a. (c) qPCR data for BTN2A1, BTN3A1, and BTN3A2 transcripts in Daudi-Cas9 cells treated with C991, internally normalized to ACTB transcripts and normalized to DMSO (vehicle)-treated cells. Two-tailed unpaired Student’s t test. (d) IgG1κ isotype control staining in Daudi-Cas9 KO cells treated with 80 µM Compound 991 (DMSO), DMSO (vehicle), 0.5 mM AICAR (aqueous), or water (vehicle) treatment for 72 h. (e) Survival of eGFP+ Daudi cells treated for 3 days with AICAR or water prior to co-culture (E:T 2:1) with primary Vγ9Vδ2 T cells in the presence of an anti-BTN3A antibody (clone 103.2). Cells were quantified using real-time quantitative live-cell imaging (Incucyte). Survival was normalized to Daudi cells cultured without T cells. (a) n = 4 per condition, representative data from one of two independent experiments. (b) n = 3 per condition, representative data from one of two independent experiments. (c) n = 4 per condition, representative data from one of three independent experiments. (d) n = 3, representative data from one of two independent experiments. (e) n = 4 per condition. (a-e) Mean ± SD. p < 0.0001 (****).

Source data

Supplementary information

Supplementary Information

Table of contents and Supplementary Fig. 1.

Reporting Summary

Supplementary Table 1

Coculture screen results by individual genes. Statistical significance was assessed by the MAGeCK RRA test followed by adjustment for multiple comparisons. FDR values were used to determine significance.

Supplementary Table 2

Coculture screen results by individual sgRNAs. Statistical significance was assessed by the MAGeCK RRA test followed by adjustment for multiple comparisons. FDR values were used to determine significance.

Supplementary Table 3

Negatively enriched coculture screen KEGG pathway gene sets.

Supplementary Table 4

LGG TCGA analysis with individual genes from the coculture screen signature. Wald test z-scores and unadjusted two-sided P values.

Supplementary Table 5

LGG TCGA analysis with pathways from the coculture screen signature. Wald test z-scores and unadjusted two-sided P values.

Supplementary Table 6

BTN3A expression screen results by individual genes. Statistical significance was assessed by the MAGeCK RRA test followed by adjustment for multiple comparisons. FDR values were used to determine significance.

Supplementary Table 7

BTN3A expression screen results by individual sgRNAs. Statistical significance was assessed by the MAGeCK RRA test followed by adjustment for multiple comparisons. FDR values were used to determine significance.

Supplementary Table 8

Primers relevant to the methods.

Supplementary Table 9

Validation sgRNAs.

Supplementary Table 10

qPCR probe assay details.

Supplementary Table 11

DP10.7 gdTCR tetramer sequences.

Supplementary Table 12

Additional gene signatures.

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

Mamedov, M.R., Vedova, S., Freimer, J.W. et al. CRISPR screens decode cancer cell pathways that trigger γδ T cell detection. Nature 621, 188–195 (2023). https://doi.org/10.1038/s41586-023-06482-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-06482-x

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