Tumor necrosis factor-α accelerates the resolution of established pulmonary fibrosis in mice by targeting profibrotic lung macrophages

EF Redente, RC Keith, W Janssen… - American journal of …, 2014 - atsjournals.org
EF Redente, RC Keith, W Janssen, PM Henson, LA Ortiz, GP Downey, DL Bratton…
American journal of respiratory cell and molecular biology, 2014atsjournals.org
Idiopathic pulmonary fibrosis (IPF) is a relentless, fibrotic parenchymal lung disease in which
alternatively programmed macrophages produce profibrotic molecules that promote
myofibroblast survival and collagen synthesis. Effective therapies to treat patients with IPF
are lacking, and conventional therapy may be harmful. We tested the hypothesis that
therapeutic lung delivery of the proinflammatory cytokine tumor necrosis factor (TNF)-α into
wild-type fibrotic mice would reduce the profibrotic milieu and accelerate the resolution of …
Idiopathic pulmonary fibrosis (IPF) is a relentless, fibrotic parenchymal lung disease in which alternatively programmed macrophages produce profibrotic molecules that promote myofibroblast survival and collagen synthesis. Effective therapies to treat patients with IPF are lacking, and conventional therapy may be harmful. We tested the hypothesis that therapeutic lung delivery of the proinflammatory cytokine tumor necrosis factor (TNF)-α into wild-type fibrotic mice would reduce the profibrotic milieu and accelerate the resolution of established pulmonary fibrosis. Fibrosis was assessed in bleomycin-instilled wild-type and TNF-α−/− mice by measuring hydroxyproline levels, static compliance, and Masson’s trichrome staining. Macrophage infiltration and programming status was assessed by flow cytometry of enzymatically digested lung and in situ immunostaining. Pulmonary delivery of TNF-α to wild-type mice with established pulmonary fibrosis was found to reduce their fibrotic burden, to improve lung function and architecture, and to reduce the number and programming status of profibrotic alternatively programmed macrophages. In contrast, fibrosis and alternative macrophage programming were prolonged in bleomycin-instilled TNF-α−/− mice. To address the role of the reduced numbers of alternatively programmed macrophages in the TNF-α–induced resolution of established pulmonary fibrosis, we conditionally depleted macrophages in MAFIA (MAcrophage Fas-Induced Apoptosis) mice. Conditional macrophage depletion phenocopied the resolution of established pulmonary fibrosis observed after therapeutic TNF-α delivery. Taken together, our results show for the first time that TNF-α is involved in the resolution of established pulmonary fibrosis via a mechanism involving reduced numbers and programming status of profibrotic macrophages. We speculate that pulmonary delivery of TNF-α or augmenting its signaling pathway represent a novel therapeutic strategy to resolve established pulmonary fibrosis.
ATS Journals