{Reference Type}: Journal Article {Title}: Distinct epigenomic landscapes underlie tissue-specific memory T cell differentiation. {Author}: Buquicchio FA;Fonseca R;Yan PK;Wang F;Evrard M;Obers A;Gutierrez JC;Raposo CJ;Belk JA;Daniel B;Zareie P;Yost KE;Qi Y;Yin Y;Nico KF;Tierney FM;Howitt MR;Lareau CA;Satpathy AT;Mackay LK; {Journal}: Immunity {Volume}: 0 {Issue}: 0 {Year}: 2024 Jul 16 {Factor}: 43.474 {DOI}: 10.1016/j.immuni.2024.06.014 {Abstract}: The memory CD8+ T cell pool contains phenotypically and transcriptionally heterogeneous subsets with specialized functions and recirculation patterns. Here, we examined the epigenetic landscape of CD8+ T cells isolated from seven non-lymphoid organs across four distinct infection models, alongside their circulating T cell counterparts. Using single-cell transposase-accessible chromatin sequencing (scATAC-seq), we found that tissue-resident memory T (TRM) cells and circulating memory T (TCIRC) cells develop along distinct epigenetic trajectories. We identified organ-specific transcriptional regulators of TRM cell development, including FOSB, FOS, FOSL1, and BACH2, and defined an epigenetic signature common to TRM cells across organs. Finally, we found that although terminal TEX cells share accessible regulatory elements with TRM cells, they are defined by TEX-specific epigenetic features absent from TRM cells. Together, this comprehensive data resource shows that TRM cell development is accompanied by dynamic transcriptome alterations and chromatin accessibility changes that direct tissue-adapted and functionally distinct T cell states.