IL-10 inhibits inflammation but does not affect fibrosis in the pulmonary response to bleomycin

https://doi.org/10.1016/j.yexmp.2003.12.010Get rights and content

Abstract

Bleomycin yields pulmonary injury characterized by inflammation that proceeds to fibrosis. The production of IL-10 by pulmonary macrophages is increased in the inflammation that accompanies bleomycin lung injury. In the present study, IL-10 deficient and wildtype mice received 0.075 units of bleomycin intratracheally at day 0 and were sacrificed at day 7 or day 14. At day 7, pulmonary inflammation was increased in IL-10-deficient mice as reflected by increased representation of CD3+ and CD4+ lymphocytes and GR-1+ pulmonary granulocytes in the bronchoalveolar lavage (BAL) fluid. Pulmonary interstitial CD80+ and CD86+ mononuclear cells were increased in situ. At day 14, mononuclear cell inflammation was comparable between groups but pulmonary eosinophils were increased in the wildtype. There was no difference in the degree of pulmonary fibrosis, as judged by histology or lung hydroxyproline content. Lung chemokine expression of MIP-1α/β, MIP-2, and eotaxin was increased at days 7 and 14 with a trend towards increased MCP-1 expression at day 14. The findings suggest an immunomodulatory role for IL-10 in the inflammatory response but not in the pulmonary fibrosis yielded by bleomycin.

Introduction

Bleomycin (BLM) yields a pulmonary fibroinflammatory response in mice (Adamson and Bowden, 1974) that mimics the histological changes of human chronic interstitial pneumonitis. IL-10 inhibits the activities of a variety of immunomodulatory Th1 cytokines (Andrea et al., 1993) and has been implicated in pathways leading to pulmonary fibrosis (Furuie et al., 1997).

IL-10 yields its anti-inflammatory effects by antagonizing the activities of IL-12 (Andrea et al., 1993) and the activities of certain Th1 cytokines, including TNF-α (Armstrong et al., 1996) and IFN-γ (Andrea et al., 1993). In addition, IL-10 down-regulates the accessory activities of dendritic cells (Qin et al., 1997) and macrophages (Bogdan et al., 1991), thereby limiting inflammation.

The role of IL-10 in vivo has previously been examined in murine models of pulmonary inflammation and fibrosis. Huaux et al. (2002) demonstrated that IL-10 decreased production of TNF-α in the pulmonary response to silica. They noted that pulmonary fibrosis was not inhibited by IL-10, suggesting that the proinflammatory and profibrotic pathways of fibroinflammatory pulmonary injury might be dissociated. Arai et al. (2000) showed that the introduction of human IL-10 gene into mice reduced inflammation and diminished hydroxyproline content in BLM-mediated lung injury.

Zhu et al. (2002) demonstrated decreased pulmonary inflammation in response to endotoxin challenge in mice that over-expressed IL-10. However, in the same model, IL-10 yielded increased inflammation in mice with null mutations for IL-13, IL-4, and STAT-6. In another study, when IL-10-deficient mice were challenged with Pseudomonas species (Chmiel et al., 2002a), they developed persistently increased pulmonary inflammation compared to wild-type controls. Considered together, these studies support an anti-inflammatory effect of IL-10 in pulmonary inflammation. However, it is uncertain whether exhibit IL-10 affects the development of pulmonary fibrosis in vivo.

When C57BL/6 mice are treated with BLM, pulmonary interstitial inflammation peaks at day 7 and pulmonary fibrosis is established at day 14 (Sharma et al., 1996). Following the intratracheal instillation of BLM, pulmonary macrophages show increased expression of IL-10 at day 5 that persists through the development of pulmonary fibrosis at day 14 (Sakamoto et al., 2002). To determine the effect of IL-10 in the pulmonary fibroinflammatory response, we investigated the immune phenotype of BLM lung injury in IL-10-deficient mice.

Section snippets

Mice

Female C57BL/6 was obtained from Charles River Laboratory (Wilmington, MA). IL-10−/− (deficient) mice developed on a genetic background of C57BL/6 mice were purchased from Jackson Laboratories. Mice were housed five per cage in the Massachusetts General Hospital (MGH) Animal Care Barrier Facility. As IL-10-deficient mice are prone to developing inflammatory bowel disease, all mice were examined at the time of sacrifice for evidence of colitis and one of the mice examined showed evidence of that

Histopathology

There were no differences in the histology of untreated lungs of IL-10-deficient and wildtype mice (not shown). The pulmonary inflammatory response to BLM was examined in IL-10-deficient mice at days 7 and 14, corresponding to the peaks of pulmonary inflammation and fibrosis, respectively, in wildtype mice. Lung sections at day 7 showed increased interstitial mononuclear cell inflammation (3.2 ± 1.1 vs. 1.6 ± 0.8, P = 0.04) in IL-10-deficient mice compared to controls (Figs. 1A, B). Mononuclear

Discussion

In a previous study, IL-10 expression by pulmonary macrophages was demonstrated to increase markedly between days 5 and 14 in C57BL/6 mice in the fibroinflammatory response to BLM (11). To examine the specific effects of IL-10 in BLM lung injury, cellular, and fibrotic responses were examined in IL-10-deficient mice. IL-10-deficient mice showed increased pulmonary inflammation compared to wildtype controls at day 7. The pulmonary inflammatory response in IL-10-deficient mice was characterized

Acknowledgements

This research was supported by NIH Grant RO139054. We wish to thank Carole Leary for her expert technical assistance.

References (38)

  • B Kozower et al.

    Am. J. Transplant.

    (2002)
  • R.L Kradin et al.

    Clin. Immunol. Immunopathol.

    (1986)
  • A Mizoguchi et al.

    Immunity

    (2002)
  • S Romagnani

    Immunol. Today

    (1997)
  • H Sakamoto et al.

    Exp. Mol. Pathol.

    (2002)
  • T Shanley et al.

    Cytokine

    (2000)
  • A Zlotnik et al.

    Immunity

    (2000)
  • I.Y.R Adamson et al.

    Am. J. Pathol.

    (1974)
  • A Andrea et al.

    J. Exp. Med.

    (1993)
  • T Arai et al.

    Introduction of the interleukin-10 gene into mice inhibited bleomycin-induced lung injury in vivo

    Am. J. Physiol., Lung Cell. Mol. Physiol.

    (2000)
  • L Armstrong et al.

    Thorax

    (1996)
  • C Bogdan et al.

    J. Exp. Med.

    (1991)
  • V Buelens et al.

    Eur. J. Immunol.

    (1995)
  • J Chmiel et al.

    Am. J. Respir. Crit. Care Med.

    (2002)
  • J Chmiel et al.

    Am. J. Respir. Crit. Care Med.

    (2002)
  • E Choi et al.

    Immunology

    (2003)
  • P.D Collins et al.

    J. Exp. Med.

    (1995)
  • J.E de Vries

    Ann. Med.

    (1995)
  • U Forssmann et al.

    J. Exp. Med.

    (1997)
  • Cited by (35)

    • Salt-inducible kinase 2 regulates fibrosis during bleomycin-induced lung injury

      2022, Journal of Biological Chemistry
      Citation Excerpt :

      The induction of TNF was however not affected by the loss of SIK2 or SIK1/2 activity (Fig. 4C). The levels of granulocyte–macrophage colony-stimulating factor and IL-10, cytokines implicated in limiting inflammation in the bleomycin model (52–54), were also increased following bleomycin treatment, but again, no significant differences were observed between wildtype, SIK2, or SIK1/2 knockin mice (Fig. 4, D and E). In addition to cytokines, several chemokines, including CCL2 and CXCL2, have been reported to be increased in mice following the administration of bleomycin and are required for the induction of bleomycin-induced pathology (14, 55, 56).

    • Pulmonary Macrophages: A New Therapeutic Pathway in Fibrosing Lung Disease?

      2016, Trends in Molecular Medicine
      Citation Excerpt :

      Global IL-10 knockout mice are also protected from silica or bleomycin-induced fibrosis [99]. However, the mechanism is unclear because Kradin et al. found that IL-10 controlled the inflammatory but not the fibrotic response to bleomycin [100]. Although originally described as a molecule secreted by activated T cells, IL-13 has also been implicated in pathogenesis of pulmonary fibrosis, and IL-13-producing AMs have been described in patients with pulmonary fibrosis [101].

    • The cytokines of pulmonary fibrosis: Much learned, much more to learn

      2015, Cytokine
      Citation Excerpt :

      Based on their findings, the authors reasoned that targeting IL-1 in chronic fibroses, such as those in human patients but not in predominantly acute mouse models of fibrosis, may have a deleterious profibrotic effect, by eliminating the protective anti-TGF-β action of IL-1β. Depending on specifics of the animal models tested, IL-10 may act profibrotically [229,230], antifibrotically [231,232], or with no effect on fibrosis [233]. Several recent reports, e.g., [168,234], have suggested a profibrotic role for IL-17 in association with inflammation.

    • Pulmonary Macrophages

      2015, Comparative Biology of the Normal Lung: Second Edition
    • Effect of tripterygium glycosides on pulmonary function in adjuvant arthritisrats

      2013, Journal of the Chinese Medical Association
      Citation Excerpt :

      Finally, the immunosuppression of CD4+ CD25+ Treg is exemplified exclusively, as our data implied. Our results showed that tripterygium can apparently improve the pulmonary function in AA rats, and the mechanism may be through the inhibition of the expression of TNF-α, ET-1, upregulation of the level of CD4+ CD25+ Treg, and promotion of the expression of IL-10 and Foxp3.11–15 In conclusion, it is suggested that tripterygium can upregulate CD4+ CD25+ Treg and Foxp3 expression, and this may explain why tripterygium is effective in the treatment of RA.16

    View all citing articles on Scopus
    View full text