Interleukin-1 and interferon-γ orchestrate β-glucan-activated human dendritic cell programming via IκB-ζ modulation

Recognition of microbial components via innate receptors including the C-type lectin receptor Dectin-1, together with the inflammatory environment, programs dendritic cells (DCs) to orchestrate the magnitude and type of adaptive immune responses. The exposure to β-glucan, a known Dectin-1 agonist an...

Full description

Saved in:
Bibliographic Details
Published inPloS one Vol. 9; no. 12; p. e114516
Main Authors Cardone, Marco, Dzutsev, Amiran K, Li, Hongchuan, Riteau, Nicolas, Gerosa, Franca, Shenderov, Kevin, Winkler-Pickett, Robin, Provezza, Lisa, Riboldi, Elena, Leighty, Robert M, Orr, Selinda J, Steinhagen, Folkert, Wewers, Mark D, Sher, Alan, Anderson, Stephen K, Goldszmid, Romina, McVicar, Daniel W, Lyakh, Lyudmila, Trinchieri, Giorgio
Format Journal Article
LanguageEnglish
Published United States Public Library of Science 04.12.2014
Public Library of Science (PLoS)
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Recognition of microbial components via innate receptors including the C-type lectin receptor Dectin-1, together with the inflammatory environment, programs dendritic cells (DCs) to orchestrate the magnitude and type of adaptive immune responses. The exposure to β-glucan, a known Dectin-1 agonist and component of fungi, yeasts, and certain immune support supplements, activates DCs to induce T helper (Th)17 cells that are essential against fungal pathogens and extracellular bacteria but may trigger inflammatory pathology or autoimmune diseases. However, the exact mechanisms of DC programming by β-glucan have not yet been fully elucidated. Using a gene expression/perturbation approach, we demonstrate that in human DCs β-glucan transcriptionally activates via an interleukin (IL)-1- and inflammasome-mediated positive feedback late-induced genes that bridge innate and adaptive immunity. We report that in addition to its known ability to directly prime T cells toward the Th17 lineage, IL-1 by promoting the transcriptional cofactor inhibitor of κB-ζ (IκB-ζ) also programs β-glucan-exposed DCs to express cell adhesion and migration mediators, antimicrobial molecules, and Th17-polarizing factors. Interferon (IFN)-γ interferes with the IL-1/IκB-ζ axis in β-glucan-activated DCs and promotes T cell-mediated immune responses with increased release of IFN-γ and IL-22, and diminished production of IL-17. Thus, our results identify IL-1 and IFN-γ as regulators of DC programming by β-glucan. These molecular networks provide new insights into the regulation of the Th17 response as well as new targets for the modulation of immune responses to β-glucan-containing microorganisms.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
Current address: Division of Therapeutic Proteins, Office of Pharmaceutical Science, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
Current address: Department for Anaesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
Competing Interests: All of the authors have declared that no competing interests exist. Coauthors Amiran K. Dzutsev and Hongchuan Li are employed by Leidos Biomedical Research, Inc. and Robert M. Leighty by Data Management Services, Inc. They work under contract from the Cancer Research Center, NCI, NIH. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.
Conceived and designed the experiments: MC GT. Performed the experiments: MC FG LL. Analyzed the data: MC AKD RML. Contributed reagents/materials/analysis tools: HL MDW SKA DWM. Wrote the paper: MC GT. Provided technical help and/or made suggestions regarding the project: RWP HL NR KS LP ER SJO FS RG AS SKA DWM.
ISSN:1932-6203
1932-6203
DOI:10.1371/journal.pone.0114516