Abstract

Proteasome inhibition has been shown to prevent development of fibrosis in several organs including the lung. However, effects of proteasome inhibitors on lung fibrosis are controversial and cytotoxic side effects of the overall inhibition of proteasomal protein degradation cannot be excluded. Therefore, we hypothesized that local lung-specific application of a novel, selective proteasome inhibitor, oprozomib (OZ), provides antifibrotic effects without systemic toxicity in a mouse model of lung fibrosis. Oprozomib was first tested on the human alveolar epithelial cancer cell line A549 and in primary mouse alveolar epithelial type II cells regarding its cytotoxic effects on alveolar epithelial cells and compared to the FDA approved proteasome inhibitor bortezomib (BZ). OZ was less toxic than BZ and provided high selectivity for the chymotrypsin-like active site of the proteasome. In primary mouse lung fibroblasts, OZ showed significant anti-fibrotic effects, i.e. reduction of collagen I and α smooth muscle actin expression, in the absence of cytotoxicity. When applied locally into the lungs of healthy mice via instillation, OZ was well tolerated and effectively reduced proteasome activity in the lungs. In bleomycin challenged mice, however, locally applied OZ resulted in accelerated weight loss and increased mortality of treated mice. Further, OZ failed to reduce fibrosis in these mice. While upon systemic application OZ was well tolerated in healthy mice, it rather augmented instead of attenuated fibrotic remodelling of the lung in bleomycin challenged mice. To conclude, low toxicity and antifibrotic effects of OZ in pulmonary fibroblasts could not be confirmed for pulmonary fibrosis of bleomycin-treated mice. In light of these data, the use of proteasome inhibitors as therapeutic agents for the treatment of fibrotic lung diseases should thus be considered with caution.

Highlights

  • Idiopathic pulmonary fibrosis (IPF) is an irreversible, lethal fibrotic disease of the lungs

  • The pathomechanism of IPF is not fully understood yet, but it is proposed that repeated microinjuries of epithelial cells induce a wound healing response during which fibroblasts differentiate into myofibroblasts

  • Specific inhibition of the CT-L active site by oprozomib is less toxic compared to bortezomib in alveolar epithelial cells

Read more

Summary

Introduction

Idiopathic pulmonary fibrosis (IPF) is an irreversible, lethal fibrotic disease of the lungs. The pathomechanism of IPF is not fully understood yet, but it is proposed that repeated microinjuries of epithelial cells induce a wound healing response during which fibroblasts differentiate into myofibroblasts. These activated myofibroblasts express α smooth muscle actin (αSMA) and release ECM proteins like collagens and fibronectin to promote matrix deposition and tissue remodelling. In lungs of IPF patients, myofibroblasts remain active and deposit excessive amounts of ECM. This leads to a destruction of alveolar organisation, loss of elastic recoil of the lung and the rapid decrease of lung function in patients. TGF-β has been identified as a main profibrotic cytokine involved in myofibroblast differentiation and as a driving factor for pathogenic pulmonary fibrosis [3,7]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call