[HTML][HTML] Dysregulated cytokine production by dendritic cells modulates B cell responses in the NZM2410 mouse model of lupus

A Sang, YY Zheng, Y Yin, I Dozmorov, H Li, HC Hsu… - PloS one, 2014 - journals.plos.org
A Sang, YY Zheng, Y Yin, I Dozmorov, H Li, HC Hsu, JD Mountz, L Morel
PloS one, 2014journals.plos.org
The breakdown in tolerance of autoreactive B cells in the lupus-prone NZM2410-derived B6.
Sle1. Sle2. Sle3 (TC) mice results in the secretion of autoantibodies. TC dendritic cells (DCs)
enhance B cell proliferation and antibody secretion in a cytokine-dependent manner.
However, the specific cytokine milieu by which TC DCs activate B cells was not known. In
this study, we compared TC and C57BL/6 (B6) control for the distribution of DC subsets and
for their production of cytokines affecting B cell responses. We show that TC DCs enhanced …
The breakdown in tolerance of autoreactive B cells in the lupus-prone NZM2410-derived B6.Sle1.Sle2.Sle3 (TC) mice results in the secretion of autoantibodies. TC dendritic cells (DCs) enhance B cell proliferation and antibody secretion in a cytokine-dependent manner. However, the specific cytokine milieu by which TC DCs activate B cells was not known. In this study, we compared TC and C57BL/6 (B6) control for the distribution of DC subsets and for their production of cytokines affecting B cell responses. We show that TC DCs enhanced B cell proliferation through the production of IL-6 and IFN-γ, while antibody secretion was only dependent on IL-6. Pre-disease TC mice showed an expanded PDCA1+ cells prior to disease onset that was localized to the marginal zone and further expanded with age. The presence of PDCA1+ cells in the marginal zone correlated with a Type I Interferon (IFN) signature in marginal zone B cells, and this response was higher in TC than B6 mice. In vivo administration of anti-chromatin immune complexes upregulated IL-6 and IFN-γ production by splenic DCs from TC but not B6 mice. The production of BAFF and APRIL was decreased upon TC DC stimulation both in vitro and in vivo, indicating that these B cell survival factors do not play a role in B cell modulation by TC DCs. Finally, TC B cells were defective at downregulating IL-6 expression in response to anti-inflammatory apoptotic cell exposure. Overall, these results show that the TC autoimmune genetic background induces the production of B cell-modulating inflammatory cytokines by DCs, which are regulated by the microenvironment as well as the interplay between DC.
PLOS