RT Journal Article SR Electronic T1 Prostaglandin E2 induction by cytosolic Listeria monocytogenes in phagocytes is necessary for optimal T-cell priming JF bioRxiv FD Cold Spring Harbor Laboratory SP 2021.03.23.436585 DO 10.1101/2021.03.23.436585 A1 Courtney E. McDougal A1 Zachary T. Morrow A1 Seonyoung Kim A1 Drake Carter A1 David M. Stevenson A1 Daniel Amador-Noguez A1 Mark J. Miller A1 John-Demian Sauer YR 2021 UL http://biorxiv.org/content/early/2021/03/23/2021.03.23.436585.abstract AB Listeria monocytogenes is an intracellular bacterium that elicits robust CD8+ T-cell responses. Despite the ongoing development of L. monocytogenes-based platforms as cancer vaccines, our understanding of how L. monocytogenes drives robust CD8+ T-cell responses remains incomplete. One overarching hypothesis is that activation of cytosolic innate pathways is critical for immunity, as strains of L. monocytogenes that are unable to access the cytosol fail to elicit robust CD8+ T-cell responses and in fact inhibit optimal T-cell priming. Counterintuitively, however, activation of known cytosolic pathways, such as the inflammasome and type I IFN, lead to impaired immunity. Here, we describe a cytosol-dependent response that is critical for immunity to L. monocytogenes, namely production of prostaglandin E2 (PGE2) downstream of cyclooxygenase-2 (COX-2). Vacuole-constrained L. monocytogenes elicit reduced PGE2 production compared to wild-type strains in macrophages and dendritic cells ex vivo. In vivo, infection with wild-type L. monocytogenes leads to 10-fold increases in PGE2 production early during infection whereas vacuole-constrained strains fail to induce PGE2 over mock-immunized controls. Mice deficient in COX-2 specifically in Lyz2+ or CD11c+ cells produce less PGE2, suggesting these cell subsets contribute to PGE2 levels in vivo, while depletion of phagocytes with clodronate abolishes PGE2 production completely. Taken together, this work identifies the first known cytosol-dependent innate immune response critical for generating CD8+ T-cell responses to L. monocytogenes, suggesting that one reason cytosolic access is required to prime CD8+ T-cell responses may be due to induction of PGE2.Author summary L. monocytogenes is an intracellular bacterial pathogen that generates robust cell-mediated immune responses. Due to this robust induction, L. monocytogenes is used as both a model to understand how CD8+ T-cells are primed, as well as a platform for cancer immunotherapy vaccines. L. monocytogenes must enter the cytosol of an infected host cell to stimulate robust T-cell responses, however, which cytosolic innate pathway(s) contribute to T-cell priming remains unclear. Here, we define COX-2 dependent PGE2 production as the first cytosol-dependent innate immune response critical for immunity to L. monocytogenes. We found that ex vivo PGE2 production by macrophages and dendritic cells is partially dependent on cytosolic access, as vacuole-constrained strains of L. monocytogenes elicit reduced PGE2. In vivo, cytosolic access is essential for PGE2 production. L. monocytogenes elicits a 10-fold increase in PGE2 production, whereas strains of L. monocytogenes that cannot access the cytosol fail to elicit PGE2 compared to mock immunized mice. Furthermore, CD11c+ and Lyz2+ cells contribute to PGE2 production in vivo, as mice deficient in COX-2 in these cell subsets have impaired PGE2 production. Taken together, our work identifies the first known cytosol-dependent pathway that is critical for generating immunity to L. monocytogenes.