Article Text

Download PDFPDF

1049 Reconstruction of gene regulatory networks dissects transcriptional control of intratumoral regulatory T cells
Free
  1. Feng Shan1,
  2. Anthony Cillo1,
  3. Carly Cardello1,
  4. Daniel Yuan1,
  5. Sheryl Kunning1,
  6. Jian Cui1,
  7. Robert Ferris1,
  8. Tullia Bruno1,
  9. Creg Workman1,
  10. Panayiotis Benos2 and
  11. Dario Vignali1
  1. 1University of Pittsburgh, Pittsburgh, PA, USA
  2. 2University of Florida, Gainesville, FL, USA

Abstract

Background Regulatory T cells (Treg) -targeted therapy exhibit clinical benefit and has been reported as a promising strategy. However, many gaps remain in our understanding of Treg biology within the context of tumor microenvironment (TME). The autoimmune toxicity and restricted efficacy are major limitations of Treg therapies in the clinic, when Treg depletion occurred not only in the tumor but in other organ systems, or concurrent downregulation of antitumor effector T cells.1, 2

Methods We profiled 51,195 single-cell transcriptomes of CD4+ T cells in tumors and peripheral blood from patients with head and neck squamous cell carcinomas (HNSCC)3, in inflamed tonsil tissues and in healthy peripheral blood. Canonical genes, gene sets and RNA Velocity4 were used to define cell states of Treg. Cibersortx5 and bulk RNA sequencing data in The Cancer Genome Atlas were used to infer the association between the enrichment of Treg subpopulations and progression-free survival of patients with solid tumors. SCENIC6 and Causal mixed graphical modeling7 were used to reconstruct the gene regulatory network (GRN). Knockout of BATF with CRISPR-Cas98 in conjunction with bulk RNA sequencing, immunophenotyping and in vitro functional assays were used to interrogate the roles of BATF in human activated Treg.

Results We identified an activated subpopulation of Treg expressing multiple tumor necrosis factor receptor (TNFR) genes, including OX40 and 4-1BB, which is highly enriched in solid TME compared with non-tumor tissues. These TNFR-activated Treg were associated with worse prognosis across solid tumors. Notably, we found BATF is a central component of a GRN that controls the transcriptional signature of TNFR-activated Treg. Consistent with single-cell analyses, BATF was co-expressed with 4-1BB, OX40, CD96 and CD39 that highly enriched in HNSCC intratumoral Treg at protein level. CRISPR-editing results revealed an enhancement of immunosuppression in BATF KO Treg and activation in BATF KO Treg accompanied with increased expression of genes including 4-1BB, OX40, ICOS, LAG3 and neuropilin-1, indicating that BATF functions as a transcriptional nexus in human activated Treg that essential for Treg activation, function and stability.

Conclusions We identify a unique intertumoral subpopulation of Treg characterized by BATF-driven expression of tumor necrosis factor receptor family expression and associated with survival across solid tumors, suggesting a possibility to target suppressive intratumoral Treg without causing overt autoimmunity in normal tissues. A deeper understanding of transcriptional network in Treg biology will provide novel mechanisms for immunotherapies in cancer, but also for Treg engineering in autoimmunity.

Acknowledgements We thank the Vignali, Bruno and Benos Labs for all their constructive comments and feedback.

References

  1. Zappasodi R, Sirard C, Li Y, Budhu S, Abu-Akeel M, Liu C, et al. Rational design of anti-GITR-based combination immunotherapy. Nature Medicine 2019;25(5):759–66.

  2. Jacobs JF, Punt CJ, Lesterhuis WJ, Sutmuller RP, Brouwer HM, Scharenborg NM, et al. Dendritic cell vaccination in combination with anti-CD25 monoclonal antibody treatment: a phase I/II study in metastatic melanoma patients. Clin Cancer Res 2010;16(20):5067–78.

  3. Cillo AR, Kürten CHL, Tabib T, Qi Z, Onkar S, Wang T, et al. Immune Landscape of Viral- and Carcinogen-Driven Head and Neck Cancer. Immunity 2020;52(1):183–99.

  4. Bergen V, Lange M, Peidli S, Wolf FA, Theis FJ. Generalizing RNA velocity to transient cell states through dynamical modeling. Nature Biotechnology 2020;38(12):1408–14.

  5. Newman AM, Steen CB, Liu CL, Gentles AJ, Chaudhuri AA, Scherer F, et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nature Biotechnology 2019;37(7):773–82.

  6. Van de Sande B, Flerin C, Davie K, De Waegeneer M, Hulselmans G, Aibar S, et al. A scalable SCENIC workflow for single-cell gene regulatory network analysis. Nature Protocols 2020;15(7):2247–76.

  7. Sedgewick AJ, Buschur K, Shi I, Ramsey JD, Raghu VK, Manatakis DV, et al. Mixed graphical models for integrative causal analysis with application to chronic lung disease diagnosis and prognosis. Bioinformatics 2019;35(7):1204–12.

  8. Schumann K, Raju SS, Lauber M, Kolb S, Shifrut E, Cortez JT, et al. Functional CRISPR dissection of gene networks controlling human regulatory T cell identity. Nature Immunology 2020;21(11):1456–66.

Ethics Approval All patients provided informed written consent, and this study was approved by our Institutional Review Board (University of Pittsburgh Cancer Institute, Tissue Collection Protocol 99-069).

Statistics from Altmetric.com

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.