TY - JOUR T1 - Divergent clonal differentiation trajectories of T cell exhaustion JF - bioRxiv DO - 10.1101/2021.12.16.472900 SP - 2021.12.16.472900 AU - Bence Daniel AU - Kathryn E. Yost AU - Katalin Sandor AU - Yu Xia AU - Yanyan Qi AU - Kamir J. Hiam-Galvez AU - Stefanie L. Meier AU - Julia A. Belk AU - Josephine R. Giles AU - E. John Wherry AU - Howard Y. Chang AU - Takeshi Egawa AU - Ansuman T. Satpathy Y1 - 2021/01/01 UR - http://biorxiv.org/content/early/2021/12/17/2021.12.16.472900.abstract N2 - T cells activated by chronic antigen exposure in the setting of viral infections or cancer can adopt an exhausted T cell (Tex) state, characterized by reduced effector function and proliferative capacity, and the upregulation of inhibitory receptors. However, whether all antigen-specific T cell clones follow the same molecular and cellular Tex differentiation trajectory remains unclear. Here, we generate a single-cell multi-omic atlas of T cell exhaustion that redefines the phenotypic diversity and molecular regulation of Tex phenotypes. Longitudinal analysis during chronic viral infection identifies an early effector phenotype that is epigenetically primed for Tex differentiation and two late-stage Tex cell states with either a terminal exhaustion or a killer cell lectin-like receptor (KLR)-expressing cytotoxic gene signature. We define clonal trajectories of antigen-specific T cells using paired single-cell RNA and T cell receptor sequencing and reveal distinct differentiation trajectories resulting in terminal Tex-biased, KLR Tex-biased, or divergent clones that differentiate into both phenotypes. Comparison of Tex phenotypes among shared T cell clones that traffic to multiple organs reveals that clonal differentiation trajectories are maintained across tissues. Finally, we show that differences in clonal differentiation trajectory are driven by TCR signal strength, whereby high-affinity T cell clones preferentially adopt a terminal Tex fate, while low-affinity clones adopt an effector-like KLR Tex fate that is detectable long-term but depleted in high antigen settings. These findings reveal clonal heterogeneity in the T cell response to chronic antigen and genomic programs that underlie Tex fates and persistence.HighlightsA single-cell atlas of T cell exhaustion identifies novel early effector and KLR Tex states.Clonal T cell analysis defines divergent differentiation trajectories during chronic viral infection leading to terminal and KLR Tex fates.The heterogeneity of the Tex pool arises from three primary differentiation patterns and are differentially persistent in the setting of high antigen.Clonal Tex differentiation patterns are conserved across organ sites and driven by TCR signal strength.Competing Interest StatementA.T.S. is a founder of Immunai and Cartography Biosciences and receives research funding from Allogene Therapeutics, Merck Research Laboratories, and 10x Genomics. H.Y.C. is a co-founder of Accent Therapeutics, Boundless Bio and Cartography Biosciences, and an advisor to 10x Genomics, Arsenal Biosciences, and Spring Discovery. K.E.Y. is a consultant for Cartography Biosciences. J.A.B. is a consultant for Immunai. ER -