Your browser doesn't support javascript.
loading
STAT3 Inhibits Autocrine IFN Signaling in Type I Conventional Dendritic Cells.
Chrisikos, Taylor T; Zhou, Yifan; Kahn, Laura M; Patel, Bhakti; Denne, Nina L; Brooks, Athena; Shen, Li; Wang, Jing; Watowich, Stephanie S.
Afiliação
  • Chrisikos TT; Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX.
  • Zhou Y; University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX; and.
  • Kahn LM; Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX.
  • Patel B; Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX.
  • Denne NL; University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX; and.
  • Brooks A; Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX.
  • Shen L; Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX.
  • Wang J; Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX.
  • Watowich SS; Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX.
J Immunol ; 209(7): 1286-1299, 2022 10 01.
Article em En | MEDLINE | ID: mdl-36038291
ABSTRACT
Type I conventional dendritic cells (cDC1s) are an essential Ag-presenting population required for generating adaptive immunity against intracellular pathogens and tumors. While the transcriptional control of cDC1 development is well understood, the mechanisms by which extracellular stimuli regulate cDC1 function remain unclear. We previously demonstrated that the cytokine-responsive transcriptional regulator STAT3 inhibits polyinosinicpolycytidylic acid [poly(IC)]-induced cDC1 maturation and cDC1-mediated antitumor immunity in murine breast cancer, indicating an intrinsic, suppressive role for STAT3 in cDC1s. To probe transcriptional mechanisms regulating cDC1 function, we generated novel RNA sequencing datasets representing poly(IC)-, IL-10-, and STAT3-mediated gene expression responses in murine cDC1s. Bioinformatics analyses indicated that poly(IC) stimulates multiple inflammatory pathways independent of STAT3, while IL-10-activated STAT3 uniquely inhibits the poly(IC)-induced type I IFN (IFN-I) transcriptional response. We validated this mechanism using purified cDC1s deficient for STAT3 or IFN signaling. Our data reveal IL-10-activated STAT3 suppresses production of IFN-ß and IFN-γ, accrual of tyrosine phosphorylated STAT1, and IFN-stimulated gene expression in cDC1s after poly(IC) exposure. Moreover, we found that maturation of cDC1s in response to poly(IC) is dependent on the IFN-I receptor, but not the type II IFN receptor, or IFN-λ. Taken together, we elucidate an essential role for STAT3 in restraining autocrine IFN-I signaling in cDC1s elicited by poly(IC) stimulation, and we provide novel RNA sequencing datasets that will aid in further delineating inflammatory and anti-inflammatory mechanisms in cDC1s.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Interleucina-10 / Fator de Transcrição STAT3 Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Interleucina-10 / Fator de Transcrição STAT3 Idioma: En Ano de publicação: 2022 Tipo de documento: Article