Antimicrobial Peptide LL-37 Ameliorates Murine Sepsis Through the Induction of Microvesicle (Ectosome) Release from Neutrophils

Authors

    Yumi Kumagai, Taisuke Murakami, Kyoko Kuwahara, Johannes Reich, Hiroshi Tamura, Isao Nagaoka Department of Host Defense and Biochemical Research, School of Medicine, Juntendo University, Bunkyo, Tokyo 113-8421, Japan Department of Host Defense and Biochemical Research, School of Medicine, Juntendo University, Bunkyo, Tokyo 113-8421, Japan Department of Bacteriology, School of Medicine, Juntendo University, Bunkyo, Tokyo 113-8421, Japan Microcoat Biotechnologie GmbH, 82347 Bernried, Germany Laboratory Program Support (LPS) Consulting Office, Tokyo 160-0023, Japan Department of Host Defense and Biochemical Research, School of Medicine, Juntendo University, Bunkyo, Tokyo 113-8421, Japan

Keywords:

Ectosomes, Antimicrobial peptides, Neutrophils, Sepsis

Abstract

Neutrophils release microvesicles (ectosomes) upon stimulation. Interestingly, ectosome level is elevated in sepsis survivors. Previously, we revealed that LL-37, a human cathelicidin antimicrobial peptide, improves the survival of a murine cecal ligation and puncture (CLP) sepsis model. Thus, in this study, we elucidated the action of LL-37 on sepsis, by focusing on the effect of LL-37 on ectosome release in the CLP model. The results demonstrated that the ectosome level was elevated in CLP mice, and the level was further enhanced by the administration of LL-37, accompanied by reduced bacterial load. More importantly, ectosome-containing microvesicles isolated from LL-37-injected CLP mice contained a higher amount of antimicrobial proteins/peptides (such as lactoferrin and murine cathelicidin-related antimicrobial peptide), and exhibited higher antibacterial activity, compared with those from PBS-injected CLP mice, suggesting that LL-37 induces the release of ectosomes with antibacterial potential in vivo. In fact, LL-37 stimulated mouse bone marrow neutrophils to release ectosomes ex vivo, and the LL-37-induced ectosomes possessed antibacterial activity. Furthermore, the administration of LL-37-induced ectosomes reduced the bacterial load and improved the survival of CLP mice. These observations suggest that LL- 37 induces the release of ectosome-containing antimicrobial microvesicles in CLP mice, thereby reducing the bacterial load and protecting the mice from lethal septic
conditions.

References

Cecconi M, Evans L, Levy M, et al., 2018, Sepsis and Septic Shock. Lancet, 2018(392): 75–87.

Hosoda H, Nakamura K, Hu Z, et al., 2017, Antimicrobial Cathelicidin Peptide LL-37 Induces NET Formation and Suppresses the Inflammatory Response in a Mouse Septic Model. Mol Med Rep, 2017(16): 5618–5626.

Hu Z, Murakami T, Suzuki K, et al., 2016, Antimicrobial Cathelicidin Peptide LL-37 Inhibits the Pyroptosis of Macrophages and Improves the Survival of Polybacterial Septic Mice. Int Immunol, 2016(28): 245–253.

Kalra H, Drummen GP, Mathivanan S, 2016, Focus on Extracellular Vesicles: Introducing the Next Small Big Thing. Int J Mol Sci, 2016(17): 170.

Nauseef WM, Borregaard N, 2014, Neutrophils at Work. Nat Immunol, 2014(15): 602–611.

Dalli J, Norling LV, Montero-Melendez T, et al., 2014, Microparticle Alpha-2-Macroglobulin Enhances Pro- Resolving Responses and Promotes Survival in Sepsis. EMBO Mol Med, 2014(6): 27–42.

Lashin HMS, Nadkarni S, Oggero S, et al., 2018, Microvesicle Subsets in Sepsis due to Community Acquired Pneumonia Compared to Faecal Peritonitis. Shock, 2018(49): 393–401.

Hu Z, Murakami T, Tamura H, et al., 2017, Neutrophil Extracellular Traps Induce IL-1β Production by Macrophages in Combination with Lipopolysaccharide. Int J Mol Med, 2017(39): 549–558.

Downloads

Published

2021-12-31