[HTML][HTML] IL-33 acts as a costimulatory signal to generate alloreactive Th1 cells in graft-versus-host disease

GK Dwyer, LR Mathews, JA Villegas… - The Journal of …, 2022 - Am Soc Clin Investig
GK Dwyer, LR Mathews, JA Villegas, A Lucas, AG de Peredo, BR Blazar, JP Girard…
The Journal of clinical investigation, 2022Am Soc Clin Investig
Antigen-presenting cells (APCs) integrate signals emanating from local pathology and
program appropriate T cell responses. In allogeneic hematopoietic stem cell transplantation
(alloHCT), recipient conditioning releases damage-associated molecular patterns (DAMPs)
that generate proinflammatory APCs that secrete IL-12, which is a driver of donor Th1
responses, causing graft-versus-host disease (GVHD). Nevertheless, other mechanisms
exist to initiate alloreactive T cell responses, as recipients with disrupted DAMP signaling or …
Antigen-presenting cells (APCs) integrate signals emanating from local pathology and program appropriate T cell responses. In allogeneic hematopoietic stem cell transplantation (alloHCT), recipient conditioning releases damage-associated molecular patterns (DAMPs) that generate proinflammatory APCs that secrete IL-12, which is a driver of donor Th1 responses, causing graft-versus-host disease (GVHD). Nevertheless, other mechanisms exist to initiate alloreactive T cell responses, as recipients with disrupted DAMP signaling or lacking IL-12 develop GVHD. We established that tissue damage signals are perceived directly by donor CD4+ T cells and promoted T cell expansion and differentiation. Specifically, the fibroblastic reticular cell–derived DAMP IL-33 is increased by recipient conditioning and is critical for the initial activation, proliferation, and differentiation of alloreactive Th1 cells. IL-33 stimulation of CD4+ T cells was not required for lymphopenia-induced expansion, however. IL-33 promoted IL-12–independent expression of Tbet and generation of Th1 cells that infiltrated GVHD target tissues. Mechanistically, IL-33 augmented CD4+ T cell TCR-associated signaling pathways in response to alloantigen. This enhanced T cell expansion and Th1 polarization, but inhibited the expression of regulatory molecules such as IL-10 and Foxp3. These data establish an unappreciated role for IL-33 as a costimulatory signal for donor Th1 generation after alloHCT.
The Journal of Clinical Investigation