Back to Search Start Over

TLR4 is required for macrophage efferocytosis during resolution of ventilator-induced lung injury.

Authors :
Kai Su
Lulong Bo
Chunling Jiang
Xiaoming Deng
You-Yang Zhao
Minshall, Richard D.
Guochang Hu
Source :
American Journal of Physiology: Lung Cellular & Molecular Physiology. Oct2021, Vol. 321 Issue 4, pL787-L801. 15p.
Publication Year :
2021

Abstract

Mechanical ventilation is a life-sustaining therapy for patients with respiratory failure but can cause further lung damage known as ventilator-induced lung injury (VILI). However, the intrinsic molecular mechanisms underlying recovery of VILI remain unknown. Phagocytosis of apoptotic cells (also known as efferocytosis) is a key mechanism orchestrating successful resolution of inflammation. Here we show the positive regulation of macrophage Toll-like receptor (TLR) 4 in efferocytosis and resolution of VILI. Mice were depleted of alveolar macrophages and then subjected to injurious ventilation (tidal volume, 20 mL/kg) for 4 h. On day 1 after mechanical ventilation, Tlr4+/+ or Tlr4-/- bone marrow-derived macrophages (BMDMs) were intratracheally administered to alveolar macrophage-depleted mice. We observed that mice depleted of alveolar macrophages exhibited defective resolution of neutrophilic inflammation, exuded protein, lung edema, and lung tissue injury after ventilation, whereas these delayed responses were reversed by administration of Tlr4+/+ BMDMs. Importantly, these proresolving effects by Tlr4+/+ BMDMs were abolished in mice receiving Tlr4-/- BMDMs. The number of macrophages containing apoptotic cells or bodies in bronchoalveolar lavage fluid was much less in mice receiving Tlr4-/- BMDMs than that in those receiving Tlr4+/+ BMDMs. Macrophage TLR4 deletion facilitated a disintegrin and metalloprotease 17 maturation and enhanced Mer cleavage in response to mechanical ventilation. Heat shock protein 70 dramatically increased Mer tyrosine kinase surface expression, phagocytosis of apoptotic neutrophils, and rescued the inflammatory phenotype in alveolar macrophage-depleted mice receiving Tlr4+/+ BMDMs, but not Tlr4-/- BMDMs. Our results suggest that macrophage TLR4 promotes resolution of VILI via modulation of Mer-mediated efferocytosis. [ABSTRACT FROM AUTHOR]

Details

Language :
English
ISSN :
10400605
Volume :
321
Issue :
4
Database :
Academic Search Index
Journal :
American Journal of Physiology: Lung Cellular & Molecular Physiology
Publication Type :
Academic Journal
Accession number :
153150888
Full Text :
https://doi.org/10.1152/ajplung.00226.2021