Abstract

Abstract Immunoglobulin A (IgA) secretion by plasma cells (PCs), terminally differentiated B cells residing in the intestinal lamina propria, assures microbiome homeostasis and protects the host against enteric infections. However, whether exposure to diet-derived and commensal-derived signals instruct tissue resident PCs effector function and dynamically shape IgA immune responses at the mucosal barrier remain largely uninvestigated. Here, we demonstrated that intestinal epithelial cells (IECs) integrate luminal input to produce 7α,25-dihydroxycholesterol (7α,25-HC), a cholesterol metabolite (oxysterol) with immunomodulatory functions. In IECs, both cholesterol uptake via NPC1L1 and commensal recognition via MyD88 controlled oxysterol production. Mice lacking the oxysterol enzyme CH25H specifically in IECs abolished 7α,25-HC production and allowed to study oxysterol generation and activity in the small intestine. Inability of IECs to generate 7α,25-HC enhanced IgA secretion by PCs in the gut, suggesting that oxysterol negatively regulate humoral response at the mucosal barriers. Mechanistically, we showed that intestinal PCs sensed 7α,25-HC via the chemoattractant receptor GPR183 to position in the lamina propria tissue. This IEC-PC axis was rapidly modulated by cholesterol dietary content and tuned Salmonella-specific IgA response. Our finding revealed a new mechanism linking dietary cholesterol and humoral immune responses centered around PC localization for efficient mucosal protection. Supported by grants from Kenneth Rainin Foundation, Innovator Award, Charles H. Hood Foundation Child Health Research Awards Program, The Leukemia and Lymphoma Society New Idea Award and the Multiple Myeloma Research Fellowship, NIH ( AI40098 ) and The American Association of Immunologists Careers in Immunology Fellowship Program.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call