4.7 Article

Divergent clonal differentiation trajectories of T cell exhaustion

Journal

NATURE IMMUNOLOGY
Volume 23, Issue 11, Pages 1614-+

Publisher

NATURE PORTFOLIO
DOI: 10.1038/s41590-022-01337-5

Keywords

-

Categories

Funding

  1. National Institutes of Health (NIH) [K08CA230188, U01CA260852, RM1-HG007735, R01AI130152, R21AI161040]
  2. Career Award for Medical Scientists from the Burroughs Wellcome Fund
  3. Lloyd J. Old STAR Award from the Cancer Research Institute
  4. American Society of Hematology
  5. Parker Institute for Cancer Immunotherapy
  6. Pew-Stewart Scholars for Cancer Research Award
  7. Baxter Foundation Faculty Scholar Award
  8. Leukemia and Lymphoma Society Scholar Award
  9. Scleroderma Research Foundation
  10. National Science Foundation Graduate Research Fellowship Program [NSF DGE-1656518]
  11. Stanford Graduate Fellowship
  12. NCI Predoctoral to Postdoctoral Fellow Transition Award [NIH F99CA253729]
  13. Stanford School of Medicine Propel Postdoctoral Scholarship
  14. National Science Foundation Graduate Research Fellowship [DGE-1656518]
  15. NIH training grant through the Stanford Immunology Program [5T32AI007290-38]
  16. NIH training grant [T32CA009547]
  17. Cancer Research Institute-Mark Foundation Fellowship [T32CA009140]
  18. Stanford Innovative Medicines Accelerator (IMA)
  19. NIH [S10OD018220, 1S10OD021763]

Ask authors/readers for more resources

In this study, the authors generate a single-cell multiomic atlas of T cell exhaustion in chronic viral infection, revealing molecular programs and clonal differentiation trajectories of exhausted T cell subsets. The results show that T cell receptor signal strength is a driver of clonal fate in T cell exhaustion.
Chronic antigen exposure during viral infection or cancer promotes an exhausted T cell (Tex) state with reduced effector function. However, whether all antigen-specific T cell clones follow the same Tex differentiation trajectory remains unclear. Here, we generate a single-cell multiomic atlas of T cell exhaustion in murine chronic viral infection that redefines Tex phenotypic diversity, including two late-stage Tex subsets with either a terminal exhaustion (Tex(term)) or a killer cell lectin-like receptor-expressing cytotoxic (Tex(KLR)) phenotype. We use paired single-cell RNA and T cell receptor sequencing to uncover clonal differentiation trajectories of Tex(term)-biased, Tex(KLR)-biased or divergent clones that acquire both phenotypes. We show that high T cell receptor signaling avidity correlates with Tex(term), whereas low avidity correlates with effector-like Tex(KLR) fate. Finally, we identify similar clonal differentiation trajectories in human tumor-infiltrating lymphocytes. These findings reveal clonal heterogeneity in the T cell response to chronic antigen that influences Tex fates and persistence. Daniel, Yost, Hsiung, et al. generate a single-cell multiomic atlas of T cell exhaustion in chronic viral infection, which reveals molecular programs of exhausted T cell subsets, identifies divergent clonal exhausted T cell differentiation trajectories and nominates TCR signal strength as a driver of clonal fate.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available