Your browser doesn't support javascript.
loading
Innate immune stimulation of whole blood reveals IFN-1 hyper-responsiveness in type 1 diabetes.
Rodrigues, Kameron B; Dufort, Matthew J; Llibre, Alba; Speake, Cate; Rahman, M Jubayer; Bondet, Vincent; Quiel, Juan; Linsley, Peter S; Greenbaum, Carla J; Duffy, Darragh; Tarbell, Kristin V.
Affiliation
  • Rodrigues KB; Immune Tolerance Section, Diabetes Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA.
  • Dufort MJ; Pathology Department, Stanford University School of Medicine, Palo Alto, CA, USA.
  • Llibre A; Systems Immunology Division, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
  • Speake C; Immunobiology of Dendritic Cells/Inserm U1223, Département d'Immunologie, Institut Pasteur, 25 rue de Dr. Roux, 75724, Paris, France.
  • Rahman MJ; Diabetes Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
  • Bondet V; Immune Tolerance Section, Diabetes Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA.
  • Quiel J; Immunobiology of Dendritic Cells/Inserm U1223, Département d'Immunologie, Institut Pasteur, 25 rue de Dr. Roux, 75724, Paris, France.
  • Linsley PS; Immune Tolerance Section, Diabetes Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA.
  • Greenbaum CJ; Systems Immunology Division, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
  • Duffy D; Diabetes Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
  • Tarbell KV; Immunobiology of Dendritic Cells/Inserm U1223, Département d'Immunologie, Institut Pasteur, 25 rue de Dr. Roux, 75724, Paris, France. darragh.duffy@pasteur.fr.
Diabetologia ; 63(8): 1576-1587, 2020 08.
Article in En | MEDLINE | ID: mdl-32500289
ABSTRACT
AIMS/

HYPOTHESIS:

Self-antigen-specific T cell responses drive type 1 diabetes pathogenesis, but alterations in innate immune responses are also critical and not as well understood. Innate immunity in human type 1 diabetes has primarily been assessed via gene-expression analysis of unstimulated peripheral blood mononuclear cells, without the immune activation that could amplify disease-associated signals. Increased responsiveness in each of the two main innate immune pathways, driven by either type 1 IFN (IFN-1) or IL-1, have been detected in type 1 diabetes, but the dominant innate pathway is still unclear. This study aimed to determine the key innate pathway in type 1 diabetes and assess the whole blood immune stimulation assay as a tool to investigate this.

METHODS:

The TruCulture whole blood ex vivo stimulation assay, paired with gene expression and cytokine measurements, was used to characterise changes in the stimulated innate immune response in type 1 diabetes. We applied specific cytokine-induced signatures to our data, pre-defined from the same assays measured in a separate cohort of healthy individuals. In addition, NOD mice were stimulated with CpG and monocyte gene expression was measured.

RESULTS:

Monocytes from NOD mice showed lower baseline vs diabetes-resistant B6.g7 mice, but higher induced IFN-1-associated gene expression. In human participants, ex vivo whole blood stimulation revealed higher induced IFN-1 responses in type 1 diabetes, as compared with healthy control participants. In contrast, neither the IL-1-induced gene signature nor response to the adaptive immune stimulant Staphylococcal enterotoxin B were significantly altered in type 1 diabetes samples vs healthy control participants. Targeted gene-expression analysis showed that this enhanced IFN response was specific to IFN-1, as IFN-γ-driven responses were not significantly different. CONCLUSIONS/

INTERPRETATION:

Our study identifies increased responsiveness to IFN-1 as a feature of both the NOD mouse model of autoimmune diabetes and human established type 1 diabetes. A stimulated IFN-1 gene signature may be a potential biomarker for type 1 diabetes and used to evaluate the effects of therapies targeting this pathway. DATA

AVAILABILITY:

Mouse gene expression data are found in the gene expression omnibus (GEO) repository, accession GSE146452 ( www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE146452 ). Nanostring count data from the human experiments were deposited in the GEO repository, accession GSE146338 ( www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE146338 ). Data files and R code for all analyses are available at https//github.com/rodriguesk/T1D_truculture_diabetologia . Graphical abstract.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Leukocytes, Mononuclear / Monocytes / Diabetes Mellitus, Type 1 / Immunity, Innate Type of study: Prognostic_studies Limits: Animals / Female / Humans Language: En Journal: Diabetologia Year: 2020 Document type: Article Affiliation country: Estados Unidos

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Leukocytes, Mononuclear / Monocytes / Diabetes Mellitus, Type 1 / Immunity, Innate Type of study: Prognostic_studies Limits: Animals / Female / Humans Language: En Journal: Diabetologia Year: 2020 Document type: Article Affiliation country: Estados Unidos