Immunopharmacology and Inflammation
CXCR2 antagonists block the N-Ac-PGP-induced neutrophil influx in the airways of mice, but not the production of the chemokine CXCL1

https://doi.org/10.1016/j.ejphar.2011.03.025Get rights and content

Abstract

Neutrophils are innate immune cells in chronic inflammatory diseases including chronic obstructive pulmonary disease (COPD) and can be attracted to the site of inflammation via the collagen breakdown product N-acetyl Proline-Glycine-Proline (N-Ac-PGP). To elucidate whether CXCR2 is involved in N-Ac-PGP-induced neutrophil migration and activation, studies using specific antagonists were performed in vivo. N-Ac-PGP and keratinocyte cell-derived chemokine (KC; CXCL1) were administered in C57Bl/6 mice via oropharyngeal aspiration. Intraperitoneal applications of CXCR2 antagonist SB225002 or SB332235 were administered 1 h prior and 1 h after oropharyngeal aspiration. Six hours after oropharyngeal aspiration mice were sacrificed. Neutrophil counts and CXCL1 levels were determined in bronchoalveolar lavage fluid, myleoperoxidase (MPO) levels were measured in lung tissue homogenates and an immunohistological staining for neutrophils was performed on lung tissue. N-Ac-PGP and CXCL1 induced a neutrophil influx in the bronchoalveolar lavage fluid and lung tissue, which was also reflected by increased MPO levels in lung tissue. The N-Ac-PGP- and CXCL1-induced neutrophil influx and the increased pulmonary tissue MPO levels were inhibited by the CXCR2 antagonists SB225002 and SB332235. Moreover, N-Ac-PGP administration enhanced the CXCL1 levels in bronchoalveolar lavage fluid, which could not be attenuated by both CXCR2 antagonists. In conclusion, neutrophil migration induced by N-Ac-PGP is mediated via direct CXCR2 interaction. The N-Ac-PGP-induced release of CXCL1 is independent of CXCR2. Related to the maximal effect of CXCL1, N-Ac-PGP is more potent at inducing neutrophil migration in the pulmonary tissue than into the bronchoalveolar lavage fluid, or N-ac-PGP may be more potent at inducing MPO levels in the lung tissue.

Introduction

Neutrophils are innate inflammatory cells in chronic inflammatory diseases including chronic obstructive pulmonary disease (COPD) and are attracted to the site of inflammation via chemoattractants, such as interleukin-8 (CXCL8) in humans and keratinocyte cell-derived chemokine (KC; CXCL1) or macrophage inflammatory protein-2 (MIP-2; CXCL2) in mice (Kobayashi, 2008). CXCL8, CXCL1 and CXCL2 bind to the CXCR2 that is predominantly expressed by neutrophils. Activated neutrophils release proteases, which contribute to collagen breakdown leading to lung emphysema. In our hypothesis, the increased protease activity leads not only to alveolar wall destruction but also to the formation of N-acetyl Pro-Gly-Pro (N-Ac-PGP) from collagen (Folkerts et al., 2008). N-Ac-PGP is chemotactic for neutrophils in vitro as well as in vivo (Weathington et al., 2006) and activates human neutrophils to release CXCL8 (Overbeek et al., in press). The importance of N-Ac-PGP in inflammatory diseases, such as COPD, is reinforced by several studies. Clinical data demonstrated that N-Ac-PGP can be detected in the bronchoalveolar lavage fluid, sputum and serum of COPD patients (O'Reilly et al., 2009, Weathington et al., 2006). Chronic airway exposure to N-Ac-PGP causes neutrophil infiltration and lung emphysema in mice (van Houwelingen et al., 2008, Weathington et al., 2006).

It has been suggested that the basis for N-Ac-PGP effects lies in its structural homology with the GP motif present in all ELR+ CXC chemokines, such as CXCL8. This GP motif is essential for cell activation and ligand binding to CXCR1/2 receptors on neutrophils (Clark-Lewis et al., 1994, Weathington et al., 2006). Overbeek et al., in press, Weathington et al., 2006 reported that N-Ac-PGP activity is mediated via the G-protein coupled receptor CXCR2, since CXCR2 antibodies suppressed the N-Ac-PGP-induced neutrophil chemotaxis in vitro. Furthermore, the accumulation of neutrophils seen in mice upon intratracheal administration of N-Ac-PGP, was not detected in CXCR2−/− mice (Weathington et al., 2006).

The aim of this study was to investigate whether the N-Ac-PGP-induced neutrophil influx was mediated via CXCR2 in vivo. We investigated the effects of two different selective nonpeptide CXCR2 antagonists, SB225002 and SB332235, in a murine model where a neutrophil influx was induced via oropharyngeal administration of N-Ac-PGP or CXCL1. In this report, we demonstrate that N-Ac-PGP and CXCL1 can induce a neutrophil influx in lung tissue and bronchoalveolar lavage fluid of mice, which can be inhibited by CXCR2 antagonists. The neutrophil migration induced by N-Ac-PGP is mediated via direct interaction with CXCR2 and by a small, if any, release of CXCL1. The N-Ac-PGP-induced release of CXCL1 is independent of the CXCR2.

Section snippets

Animals

Male C57Bl/6 mice, 6–7 weeks old were obtained from Charles River Laboratories and housed under controlled conditions in standard laboratory cages in the animal facility. They were provided free access to water and food. All in vivo experimental protocols were approved by the local Ethics Committee and were performed under strict governmental and international guidelines on animal experimentation.

Oropharyngeal aspiration

Oropharyngeal aspiration of CXCL1 (0.5 μg in 70 μl PBS) (R&D systems, Minneapolis, USA), N-Ac-PGP (500 

N-Ac-PGP-induced neutrophil infiltration in lung tissue is more pronounced than the influx into the bronchoalveolar lavage fluid

Oropharyngeal aspiration of N-Ac-PGP and CXCL1 resulted in a neutrophilic airway inflammation, since an increased number of neutrophils was observed in the bronchoalveolar lavage fluid of N-Ac-PGP- and CXCL1-treated mice compared to control mice (Fig. 1, Fig. 2). However, N-Ac-PGP at a dose of 500 μg/mouse only induced 5% of the amount of neutrophils in bronchoalveolar lavage fluid compared to that observed after treatment with 0.5 μg CXCL1 per mouse.

Next, we examined whether the appearance of

Discussion

The aim of this study was to investigate the role of CXCR2 in neutrophilic airway inflammation induced by N-Ac-PGP. Neutrophils are the first cells to be recruited to the site of inflammation in response to chemo-attractants, such as CXCL8 in humans and CXCL1 and CXCL2 in mice (Kobayashi, 2008). In chronically inflamed tissues, neutrophilic mediator release leads to extracellular matrix breakdown and subsequently to the formation of collagen fragments with chemotactic properties, such as

Acknowledgements

The authors would like to thank Kim Verheijden en Marije Kleinjan for their excellent technical assistance.

This work was performed within the framework of the Dutch Top Institute Pharma Project T1-103.

References (30)

  • C. Chang et al.

    Demonstration of the chemotactic properties of collagen

    Proc. Soc. Exp. Biol. Med.

    (1970)
  • P. de Kruijf et al.

    Nonpeptidergic allosteric antagonists differentially bind to the CXCR2 chemokine receptor

    J. Pharmacol. Exp. Ther.

    (2009)
  • C.W. Frevert et al.

    Functional characterization of the rat chemokine KC and its importance in neutrophil recruitment in a rat model of pulmonary inflammation

    J. Immunol.

    (1995)
  • J.L. Haddox et al.

    Bioactivity of peptide analogs of the neutrophil chemoattractant, N-acetyl-proline-glycine-proline

    Invest. Ophthalmol. Vis. Sci.

    (1999)
  • W. Herbold et al.

    Importance of CXC chemokine receptor 2 in alveolar neutrophil and exudate macrophage recruitment in response to pneumococcal lung infection

    Infect. Immun.

    (2010)
  • Cited by (0)

    1

    These authors contributed equally to this work.

    View full text