Your browser doesn't support javascript.
loading
Targeting transitioning lung monocytes/macrophages as treatment strategies in lung disease related to environmental exposures.
Schwab, Aaron D; Wyatt, Todd A; Moravec, Grace; Thiele, Geoffrey M; Nelson, Amy J; Gleason, Angela; Schanze, Oliver; Duryee, Michael J; Romberger, Debra J; Mikuls, Ted R; Poole, Jill A.
Affiliation
  • Schwab AD; Division of Allergy & Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
  • Wyatt TA; Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE, USA.
  • Moravec G; Division of Pulmonary, Critical Care & Sleep, University of Nebraska Medical Center, Omaha, NE, USA.
  • Thiele GM; Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA.
  • Nelson AJ; Division of Allergy & Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
  • Gleason A; Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE, USA.
  • Schanze O; Division of Rheumatology and Immunology, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
  • Duryee MJ; Division of Allergy & Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
  • Romberger DJ; Division of Allergy & Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
  • Mikuls TR; Division of Allergy & Immunology, University of Nebraska Medical Center, Omaha, NE, USA.
  • Poole JA; Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE, USA.
Respir Res ; 25(1): 157, 2024 Apr 09.
Article in En | MEDLINE | ID: mdl-38594676
ABSTRACT

BACKGROUND:

Environmental/occupational exposures cause significant lung diseases. Agricultural organic dust extracts (ODE) and bacterial component lipopolysaccharide (LPS) induce recruited, transitioning murine lung monocytes/macrophages, yet their cellular role remains unclear.

METHODS:

CCR2 RFP+ mice were intratracheally instilled with high concentration ODE (25%), LPS (10 µg), or gram-positive peptidoglycan (PGN, 100 µg) for monocyte/macrophage cell-trafficking studies. CCR2 knockout (KO) mice and administration of intravenous clodronate liposomes strategies were employed to reduce circulating monocytes available for lung recruitment following LPS exposure. Lung tissues and bronchoalveolar lavage fluid (BALF) were collected. Pro-inflammatory and/or pro-fibrotic cytokines, chemokines, and lung extracellular matrix mediators were quantitated by ELISA. Infiltrating lung cells including monocyte/macrophage subpopulations, neutrophils, and lymphocytes were characterized by flow cytometry. Lung histopathology, collagen content, vimentin, and post-translational protein citrullination and malondialdehyde acetaldehyde (MAA) modification were quantitated. Parametric statistical tests (one-way ANOVA, Tukey'smultiple comparison) and nonparametric statistical (Kruskal-Wallis, Dunn's multiple comparison) tests were used following Shapiro-Wilk testing for normality.

RESULTS:

Intratracheal instillation of ODE, LPS, or PGN robustly induced the recruitment of inflammatory CCR2+ CD11cintCD11bhi monocytes/macrophages and both CCR2+ and CCR2- CD11c-CD11bhi monocytes at 48 h. There were also increases in CCR2+ CD4+ and CD8+ T cells and NK cells. Despite reductions in LPS-induced lung infiltrating CD11cintCD11bhi cells (54% reduction), CCR2 knockout (KO) mice were not protected against LPS-induced inflammatory and pro-fibrotic consequences. Instead, compensatory increases in lung neutrophils and CCL2 and CCL7 release occurred. In contrast, the depletion of circulating monocytes through the administration of intravenous clodronate (vs. vehicle) liposomes 24 h prior to LPS exposure reduced LPS-induced infiltrating CD11cintCD11bhi monocyte-macrophage subpopulation by 59% without compensatory changes in other cell populations. Clodronate liposome pre-treatment significantly reduced LPS-induced IL-6 (66% reduction), matrix metalloproteinases (MMP)-3 (36%), MMP-8 (57%), tissue inhibitor of metalloproteinases (61%), fibronectin (38%), collagen content (22%), and vimentin (40%). LPS-induced lung protein citrullination and MAA modification, post-translational modifications implicated in lung disease, were reduced (39% and 48%) with clodronate vs. vehicle liposome.

CONCLUSION:

Highly concentrated environmental/occupational exposures induced the recruitment of CCR2+ and CCR2- transitioning monocyte-macrophage and monocyte subpopulations and targeting peripheral monocytes may reduce the adverse lung consequences resulting from exposures to LPS-enriched inhalants.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Monocytes / Lung Diseases Limits: Animals Language: En Journal: Respir Res Year: 2024 Document type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Monocytes / Lung Diseases Limits: Animals Language: En Journal: Respir Res Year: 2024 Document type: Article Affiliation country: United States