IgG is elevated in obese white adipose tissue but does not induce glucose intolerance via Fcγ-receptor or complement

Background/Objectives: In obesity, B cells accumulate in white adipose tissue (WAT) and produce IgG, which may contribute to the development of glucose intolerance. IgG signals by binding to Fcγ receptors (FcγR) and by activating the complement system. The aim of our study was to investigate whether...

Full description

Saved in:
Bibliographic Details
Published inInternational Journal of Obesity Vol. 42; no. 2; pp. 260 - 269
Main Authors van Dam, A D, van Beek, L, Pronk, A C M, van den Berg, S M, Van den Bossche, J, de Winther, M P J, Koning, F, van Kooten, C, Rensen, P C N, Boon, M R, Verbeek, J S, van Dijk, K Willems, van Harmelen, V
Format Journal Article
LanguageEnglish
Published London Nature Publishing Group UK 01.02.2018
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Background/Objectives: In obesity, B cells accumulate in white adipose tissue (WAT) and produce IgG, which may contribute to the development of glucose intolerance. IgG signals by binding to Fcγ receptors (FcγR) and by activating the complement system. The aim of our study was to investigate whether activation of FcγR and/or complement C3 mediates the development of high-fat diet-induced glucose intolerance. Methods: We studied mice lacking all four FcγRs (FcγRI/II/III/IV −/− ), only the inhibitory FcγRIIb (FcγRIIb −/− ), only the central component of the complement system C3 (C3 −/− ), and mice lacking both FcγRs and C3 (FcγRI/II/III/IV/C3 −/− ). All mouse models and wild-type controls were fed a high-fat diet (HFD) for 15 weeks to induce obesity. Glucose metabolism was assessed and adipose tissue was characterized for inflammation and adipocyte functionality. Results: In obese WAT of wild-type mice, B cells (+142%, P <0.01) and IgG (+128% P <0.01) were increased compared to lean WAT. Macrophages of FcγRI/II/III/IV −/− mice released lower levels of cytokines compared to wild-type mice upon IgG stimulation. Only C3 −/− mice showed reduced HFD-induced weight gain as compared to controls (−18%, P <0.01). Surprisingly, FcγRI/II/III/IV −/− mice had deteriorated glucose tolerance (AUC +125%, P <0.001) despite reduced leukocyte number (−30%, P <0.05) in gonadal WAT (gWAT), whereas glucose tolerance and leukocytes within gWAT in the other models were unaffected compared to controls. Although IgG in gWAT was increased (+44 to +174%, P <0.05) in all mouse models lacking FcγRIIb, only FcγRI/II/III/IV/C3 −/− mice exhibited appreciable alterations in immune cells in gWAT, for example, increased macrophages (+36%, P <0.001). Conclusions: Lack of FcγRs reduces the activity of macrophages upon IgG stimulation, but neither FcγR nor C3 deficiency protects against HFD-induced glucose intolerance or reduces adipose tissue inflammation. This indicates that if obesity-induced IgG contributes to the development of glucose intolerance, this is not mediated by FcγR or complement activation.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:0307-0565
1476-5497
DOI:10.1038/ijo.2017.209