A Novel Role for C5a in B-1 Cell Homeostasis

Front Immunol. 2018 Feb 19:9:258. doi: 10.3389/fimmu.2018.00258. eCollection 2018.

Abstract

B-1 cells constitute a unique subpopulation of lymphocytes residing mainly in body cavities like the peritoneal cavity (PerC) but are also found in spleen and bone marrow (BM). As innate-like B cells, they mediate first line immune defense through low-affinity natural IgM (nIgM) antibodies. PerC B-1 cells can egress to the spleen and differentiate into nIgM antibody-secreting plasma cells that recognize conserved exogenous and endogenous cellular structures. Homing to and homeostasis within the PerC are regulated by the chemokine CXCL13 released by PerC macrophages and stroma cells. However, the exact mechanisms underlying the regulation of CXCL13 and B-1 homeostasis are not fully explored. B-1 cells play important roles in the inflammatory response to infection, autoimmunity, ischemia/reperfusion injury, obesity, and atherosclerosis. Remarkably, this list of inflammatory entities has a strong overlap with diseases that are regulated by complement suggesting a link between B-1 cells and the complement system. Interestingly, up to now, no data exist regarding the role of complement in B-1 cell biology. Here, we demonstrate for the first time that C5a regulates B-1 cell steady-state dynamics within the peritoneum, the spleen, and the BM. We found decreased B-1a cell numbers in the peritoneum and the spleen of C5aR1-/- mice associated with increased B1-a and B1-b numbers in the spleen and high serum titers of nIgM antibodies directed against phosphorylcholine and several pneumococcal polysaccharides. Similarly, peritoneal B-1a cells were decreased in the peritoneum and splenic B-1a and B-1b cells were increased in C5aR2-/- mice. The decrease in peritoneal B-1 cell numbers was associated with decreased peritoneal CXCL13 levels in C5aR1-/- and C5aR2-/- mice. In search for mechanisms, we found that combined TLR2 and IL-10 receptor activation in PerC macrophages induced strong CXCL13 production, which was significantly reduced in cells from C5aR1- and C5aR2-deficient mice and after combined C5aR-targeting. Such stimulation also induced marked local C5 production by PerC macrophages and C5a generation. Importantly, peritoneal in vivo administration of C5a increased CXCL13 production. Taken together, our findings suggest that local non-canonical C5 activation in PerC macrophages fuels CXCL13 production as a novel mechanism to control B-1 cell homeostasis.

Keywords: B-1 cells; C5; C5a; CXCL13; complement; natural antibodies.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • B-Lymphocyte Subsets / immunology*
  • B-Lymphocytes / immunology*
  • Cells, Cultured
  • Chemokine CXCL13 / metabolism*
  • Complement C5a / metabolism*
  • Cytokines / metabolism
  • Homeostasis
  • Humans
  • Immunity, Innate
  • Immunoglobulin M / blood
  • Lymphocyte Activation
  • Macrophages / immunology*
  • Mice
  • Mice, Inbred BALB C
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Peritoneum / immunology*
  • Receptor, Anaphylatoxin C5a / genetics
  • Receptors, Interleukin-10 / metabolism
  • Spleen / immunology*
  • Th1 Cells / immunology

Substances

  • C5ar1 protein, mouse
  • C5ar2 protein, mouse
  • Chemokine CXCL13
  • Cxcl13 protein, mouse
  • Cytokines
  • Immunoglobulin M
  • Receptor, Anaphylatoxin C5a
  • Receptors, Interleukin-10
  • Complement C5a