Your browser doesn't support javascript.
loading
Apoptotic cells induce CD103 expression and immunoregulatory function in myeloid dendritic cell precursors through integrin αv and TGF-ß activation.
Zhang, Ailiang; Paidassi, Helena; Lacy-Hulbert, Adam; Savill, John.
Affiliation
  • Zhang A; Medical Research Council Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, Scotland, United Kingdom.
  • Paidassi H; Inserm U1111, Lyon, France.
  • Lacy-Hulbert A; Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States of America.
  • Savill J; Medical Research Council Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, Scotland, United Kingdom.
PLoS One ; 15(7): e0232307, 2020.
Article in En | MEDLINE | ID: mdl-32667911
ABSTRACT
In the mammalian gut CD103+ve myeloid DCs are known to suppress inflammation threatened by luminal bacteria, but stimuli driving DC precursor maturation towards this beneficial phenotype are incompletely understood. We isolated CD11+ve DCs from mesenteric lymph nodes (MLNs) of healthy mice; CD103+ve DCs were 8-24 fold more likely than CD103-ve DCs to exhibit extensive of prior phagocytosis of apoptotic intestinal epithelial cells. However, CD103+ve and CD103-ve MLN DCs exhibited similar ex vivo capacity to ingest apoptotic cells, indicating that apoptotic cells might drive immature DC maturation towards the CD103+ve phenotype. When cultured with apoptotic cells, myeloid DC precursors isolated from murine bone marrow and characterised as lineage-ve CD103-ve, displayed enhanced expression of CD103 and ß8 integrin and acquired increased capacity to induce T regulatory lymphocytes (Tregs) after 7d in vitro. However, DC precursors isolated from αv-tie2 mice lacking αv integrins in the myeloid line exhibited reduced binding of apoptotic cells and complete deficiency in the capacity of apoptotic cells and/or latent TGF-ß1 to enhance CD103 expression in culture, whereas active TGF-ß1 increased DC precursor CD103 expression irrespective of αv expression. Fluorescence microscopy revealed clustering of αv integrin chains and latent TGF-ß1 at points of contact between DC precursors and apoptotic cells. We conclude that myeloid DC precursors can deploy αv integrin to orchestrate binding of apoptotic cells, activation of latent TGF-ß1 and acquisition of the immunoregulatory CD103+ve ß8+ve DC phenotype. This implies that a hitherto unrecognised consequence of apoptotic cell interaction with myeloid phagocytes is programming that prevents inflammation.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Dendritic Cells / Antigens, CD / Gene Expression Regulation / Apoptosis / Integrin alpha Chains / Integrin alphaV / Transforming Growth Factor beta1 / Immunomodulation Limits: Animals Language: En Journal: PLoS One Journal subject: CIENCIA / MEDICINA Year: 2020 Document type: Article Affiliation country: Reino Unido

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Dendritic Cells / Antigens, CD / Gene Expression Regulation / Apoptosis / Integrin alpha Chains / Integrin alphaV / Transforming Growth Factor beta1 / Immunomodulation Limits: Animals Language: En Journal: PLoS One Journal subject: CIENCIA / MEDICINA Year: 2020 Document type: Article Affiliation country: Reino Unido