Genome-wide CRISPR screens of T cell exhaustion identify chromatin remodeling factors that limit T cell persistence. Academic Article uri icon

Overview

abstract

  • T cell exhaustion limits antitumor immunity, but the molecular determinants of this process remain poorly understood. Using a chronic stimulation assay, we performed genome-wide CRISPR-Cas9 screens to systematically discover regulators of T cell exhaustion, which identified an enrichment of epigenetic factors. In vivo CRISPR screens in murine and human tumor models demonstrated that perturbation of the INO80 and BAF chromatin remodeling complexes improved T cell persistence in tumors. In vivo Perturb-seq revealed distinct transcriptional roles of each complex and that depletion of canonical BAF complex members, including Arid1a, resulted in the maintenance of an effector program and downregulation of exhaustion-related genes in tumor-infiltrating T cells. Finally, Arid1a depletion limited the acquisition of exhaustion-associated chromatin accessibility and led to improved antitumor immunity. In summary, we provide an atlas of the genetic regulators of T cell exhaustion and demonstrate that modulation of epigenetic state can improve T cell responses in cancer immunotherapy.

publication date

  • June 23, 2022

Research

keywords

  • Chromatin Assembly and Disassembly
  • Neoplasms

Identity

PubMed Central ID

  • PMC9949532

Scopus Document Identifier

  • 85133872187

Digital Object Identifier (DOI)

  • 10.1016/j.ccell.2022.06.001

PubMed ID

  • 35750052

Additional Document Info

volume

  • 40

issue

  • 7