Neutrophil L-Plastin Controls Ocular Paucibacteriality and Susceptibility to Keratitis

Citation:

Lu X, Kugadas A, Smith-Page K, Lamb J, Lin T, Ru Y, Morley SC, Fichorova R, Mittal SK, Chauhan SK, Littleton S, Saban D, Gadjeva M. Neutrophil L-Plastin Controls Ocular Paucibacteriality and Susceptibility to Keratitis. Front Immunol 2020;11:547.

Date Published:

2020

Abstract:

Why ocular mucosa is paucibacterial is unknown. Many different mechanisms have been suggested but the comprehensive experimental studies are sparse. We found that a deficiency in L-plastin (LCP1), an actin bundling protein, resulted in an ocular commensal overgrowth, characterized with increased presence of conjunctival spp. The commensal overgrowth correlated with susceptibility to -induced keratitis. L-plastin knock-out (KO) mice displayed elevated bacterial burden in the -infected corneas, altered inflammatory responses, and compromised bactericidal activity. Mice with ablation of LPL under the LysM Cre ( ) and S100A8 Cre ( ) promoters had a similar phenotype to the LPL KOs mice. In contrast, infected mice did not display elevated susceptibility to infection, implicating the myeloid L-plastin-sufficient cells (e.g., macrophages and neutrophils) in maintaining ocular homeostasis. Mechanistically, the elevated commensal burden and the susceptibility to infection were linked to defects in neutrophil frequencies at steady state and during infection and compromised bactericidal activities upon priming. Macrophage exposure to commensal organisms primed neutrophil responses to , augmenting PMN bactericidal capacity in an L-plastin dependent manner. Cumulatively, our data highlight the importance of neutrophils in controlling ocular paucibacteriality, reveal molecular and cellular events involved in the process, and suggest a link between commensal exposure and resistance to infection.

See also: Cornea, April 2020, All, 2020
Last updated on 04/30/2020