Fibrocyte CXCR4 regulation as a therapeutic target in pulmonary fibrosis

https://doi.org/10.1016/j.biocel.2009.02.020Get rights and content

Abstract

Fibrotic interstitial lung diseases are characterized by progressive decline in lung function and premature death from respiratory failure. Fibrocytes are circulating bone marrow-derived progenitor cells that traffic to the lungs and contribute to fibrosis and may represent novel therapeutic targets in these diseases. We have previously found the recruitment of fibrocytes to the lung to be dependent on the chemokine ligand CXCL12. Given that the expression of the CXCL12 receptor, CXCR4, can be modulated pharmacologically in other cell types, we tested the hypotheses that the regulation of CXCR4 expression on fibrocytes mediates their influx to the lung in the context of pulmonary fibrosis and that pharmacologic inhibition of this process results in attenuated disease severity. CXCR4 was the predominant chemokine receptor on human fibrocytes, and its expression on fibrocytes was enhanced by hypoxia and by growth factors including platelet-derived growth factor. Both hypoxia-induced and growth factor-induced CXCR4 expressions were attenuated by specific inhibition of PI3-kinase and mTOR. Finally, in the mouse model of bleomycin-induced pulmonary fibrosis, treatment with the mTOR inhibitor rapamycin resulted in reduced numbers of CXCR4-expressing fibrocytes in the peripheral blood and lung as well as reduced lung collagen deposition. Taken together, these experiments support the notion that pharmacologic inhibition of the CXCR4/CXCL12 biological axis is achievable in human fibrocytes and reduces the magnitude of pulmonary fibrosis in an animal model. This approach may hold promise in human fibrotic lung diseases.

Introduction

Fibrotic interstitial lung diseases are illnesses characterized by tissue remodeling, fibroproliferation, and deposition of extra-cellular matrix in the lung parenchyma. Some of these illnesses occur in response to known insults – such as inorganic dusts, inhaled antigens, or autoantigens – whereas others are of unknown cause. The prototypical fibrotic lung disease is idiopathic pulmonary fibrosis (IPF), a progressive disorder that culminates in premature death from respiratory failure and in which no treatment intervention has been effective to date. There is thus an urgent clinical need for new therapeutic approaches these diseases.

A fundamental question in the pathogenesis of fibrotic interstitial lung diseases is the cellular precursors of the lung fibroblasts and myofibroblasts. In this context, we have previously examined the contribution of a population of bone marrow-derived circulating progenitor cells, the fibrocytes (Phillips et al., 2004). Specifically, we found fibrocytes to accumulate in the lungs of mice with bleomycin-induced pulmonary fibrosis in associated with deposition of collagen and that human fibrocytes given to SCID mice challenged with bleomycin specifically trafficked to the lungs (Phillips et al., 2004). Consistent with this, we also found an expanded pool of fibrocytes in the peripheral blood of patients with fibrotic interstitial lung diseases (Mehrad et al., 2007) findings that have been subsequently reproduced by another group (Andersson-Sjoland et al., 2008). We have recently found that the expanded pool of circulating fibrocytes in patients with IPF is an independent predictor of mortality (Moeller et al., 2009), lending further support to the relevance of these cells in the pathogenesis of pulmonary fibrosis.

Identification of the chemokine ligand–receptor pair that mediates the mobilization of human fibrocytes from the bone marrow to the lung in the context of pulmonary fibrosis has the potential to open new avenues for therapy for these illnesses, but the issue remains unsettled in the literature. This is, at least in part, due to differences between human and mouse fibrocyte chemokine receptor repertoire: human fibrocytes express CXCR4, CCR3, CCR5 and CCR7 (Phillips et al., 2004, Quan et al., 2004) whereas mouse fibrocytes express CXCR4, CCR2 and CCR7 (Phillips et al., 2004, Abe et al., 2001, Moore et al., 2005, Quan et al., 2004). CXCR4 is a critical chemokine receptor in haematopoietic and non-haematopoietic stem cell homing in both mice and humans, and the differential expression of CXCL12 in tissue creates a gradient essential for traffic of CXCR4-expressing cells (Murdoch, 2000). Consistent with their progenitor cell phenotype, human fibrocytes express CXCR4 and migrate in response to CXCL12 in vitro (Phillips et al., 2004). In the context of the mouse model, there was a marked induction in lung CXCL12 after bleomycin challenge and the neutralization of this ligand resulted in reduced accumulation of fibrocytes in the lungs and attenuated lung fibrosis (Phillips et al., 2004). In human interstitial lung disease, we found elevated plasma and lung levels of CXCL12 protein associated with ∼90% CXCR4 expression in circulating fibrocytes (Mehrad et al., 2007, Phillips et al., 2004). Other groups have shown mouse fibrocytes to traffic to tissue via the CCR2/CCL12 axis in the FITC-induced model of pulmonary fibrosis (Moore et al., 2005, Moore et al., 2006), via the CCL3–CCR5 axis in bleomycin-induced pulmonary fibrosis (Ishida et al., 2007), and via CCR7 in models of renal fibrosis and wound healing (Abe et al., 2001, Sakai et al., 2006).

Prior work has demonstrated the expression of CXCR4 on fibrocytes in the context of mouse and human pulmonary fibrosis and implicated CXCL12-mediated traffic of fibrocytes to the lungs in the pathogenesis of pulmonary fibrosis (Phillips et al., 2004, Mehrad et al., 2007, Andersson-Sjoland et al., 2008). The purpose of the current work was to provide proof-of-principle that this mechanism can be manipulated pharmacologically as a therapeutic strategy in fibrotic lung disease. We therefore tested the hypotheses that the regulation of CXCR4 expression on fibrocytes mediates their influx to the lung in the context of pulmonary fibrosis and that pharmacologic inhibition of this process results in attenuated disease severity.

Section snippets

Human fibrocyte isolation

Human fibrocytes were quantified in fresh blood samples collected in EDTA tubes or were isolated from leukophoresis packs (HemaCare, Woodland Hills, CA) as previously described (Phillips et al., 2004). Briefly, peripheral blood mononuclear cells were plated on fibronectin-coated flasks and cultured in complete media [Dulbecco's Modified Eagle Media (DMEM, Invitrogen, Carlsbad, CA) supplemented with 20% FCS, 25 mM HEPES, 100 units/mL penicillin, and 100 ng/mL streptomycin]. Non-adherent cells were

Fibrocytes traffic to the lungs and differentiate into myofibroblasts

We have previously shown that human fibrocytes transferred to SCID mice challenged with bleomycin traffic to the lungs, and that neutralization of CXCL12 in bleomycin-induced pulmonary fibrosis results in reduced accumulation murine fibrocytes in the lungs that is associated with reduced lung collagen deposition (Phillips et al., 2004). On the other hand, the magnitude of the contribution of bone marrow-derived fibrocytes to total lung fibroblasts in the context of pulmonary fibrosis is not

Discussion

We have previously shown that CD45+ Col1+ CXCR4+ circulating fibrocytes accumulate in the lung in response to CXCL12 and mediate fibrosis (Phillips et al., 2004). In the context of human pulmonary fibrosis, there is a correlation between lung and plasma levels of CXCL12 and circulating and lung fibrocyte numbers (Mehrad et al., 2007, Andersson-Sjoland et al., 2008). To provide the proof-of-principle that the CXCL12–CXCR4 axis can be targeted therapeutically in pulmonary fibrosis, the present

Acknowledgment

Sources of funding: This work was supported by NIH grants HL73848 and an American Lung Association Career Investigator Award (Mehrad) and CA87879 and HL66027 (Strieter).

References (59)

  • T.E. Quan et al.

    Circulating fibrocytes: collagen-secreting cells of the peripheral blood

    Int J Biochem Cell Biol

    (2004)
  • A.B. Rice et al.

    Specific inhibitors of platelet-derived growth factor or epidermal growth factor receptor tyrosine kinase reduce pulmonary fibrosis in rats

    Am J Pathol

    (1999)
  • D.E. Richard et al.

    Nonhypoxic pathway mediates the induction of hypoxia-inducible factor 1alpha in vascular smooth muscle cells

    J Biol Chem

    (2000)
  • J. Schlessinger

    Cell signaling by receptor tyrosine kinases

    Cell

    (2000)
  • C.L. Semerad et al.

    G-CSF potently inhibits osteoblast activity and CXCL12 mRNA expression in the bone marrow

    Blood

    (2005)
  • T. Sugiyama et al.

    Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches

    Immunity

    (2006)
  • H. Yasuoka et al.

    Insulin-like growth factor-binding protein-5 induces pulmonary fibrosis and triggers mononuclear cellular infiltration

    Am J Pathol

    (2006)
  • A. Abdollahi et al.

    Inhibition of platelet-derived growth factor signaling attenuates pulmonary fibrosis

    J Exp Med

    (2005)
  • R. Abe et al.

    Peripheral blood fibrocytes: differentiation pathway and migration to wound sites

    J Immunol

    (2001)
  • A. Andersson-Sjoland et al.

    Fibrocytes are a potential source of lung fibroblasts in idiopathic pulmonary fibrosis

    Int J Biochem Cell Biol

    (2008)
  • H.N. Antoniades et al.

    Platelet-derived growth factor in idiopathic pulmonary fibrosis

    J Clin Invest

    (1990)
  • C. Aston et al.

    Enhanced insulin-like growth factor molecules in idiopathic pulmonary fibrosis

    Am J Respir Crit Care Med

    (1995)
  • A.F. Cashen et al.

    Mobilizing stem cells from normal donors: is it possible to improve upon G-CSF?

    Bone Marrow Transplant

    (2007)
  • D.J. Ceradini et al.

    Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1

    Nat Med

    (2004)
  • M.J. Christopher et al.

    Suppression of CXCL12 production by bone marrow osteoblasts is a common and critical pathway for cytokine-induced mobilization

    Blood

    (2009)
  • A. Fasolo et al.

    mTOR inhibitors in the treatment of cancer

    Expert Opin Investig Drugs

    (2008)
  • B.N. Gomperts et al.

    Circulating progenitor epithelial cells traffic via CXCR4/CXCL12 in response to airway injury

    J Immunol

    (2006)
  • N. Hashimoto et al.

    Bone marrow-derived progenitor cells in pulmonary fibrosis

    J Clin Invest

    (2004)
  • C.H. Heldin et al.

    Mechanism of action and in vivo role of platelet-derived growth factor

    Physiol Rev

    (1999)
  • Cited by (154)

    View all citing articles on Scopus
    View full text