Inhibition of PIM1 kinase attenuates bleomycin-induced pulmonary fibrosis in mice by modulating the ZEB1/E-cadherin pathway in alveolar epithelial cells

X Zhang, Y Zou, Y Liu, Y Cao, J Zhu, J Zhang… - Molecular …, 2020 - Elsevier
X Zhang, Y Zou, Y Liu, Y Cao, J Zhu, J Zhang, X Chen, R Zhang, J Li
Molecular Immunology, 2020Elsevier
PIM1 is serine/threonine protein kinase that is involved in numerous biological processes.
Pulmonary fibrosis (PF) is a chronic pathological result of the dysfunctional repair of lung
injury without effective therapeutic treatments. In the current study, we investigated whether
PIM1 inhibition would improve bleomycin (BLM)-induced pulmonary fibrosis. In a BLM-
induced pulmonary fibrosis model, PIM1 was persistently upregulated in fibrotic lung tissues.
Furthermore, PIM1 inhibition by the PIM1-specific inhibitor SMI-4a showed protective effects …
Abstract
PIM1 is serine/threonine protein kinase that is involved in numerous biological processes. Pulmonary fibrosis (PF) is a chronic pathological result of the dysfunctional repair of lung injury without effective therapeutic treatments. In the current study, we investigated whether PIM1 inhibition would improve bleomycin (BLM)-induced pulmonary fibrosis. In a BLM-induced pulmonary fibrosis model, PIM1 was persistently upregulated in fibrotic lung tissues. Furthermore, PIM1 inhibition by the PIM1-specific inhibitor SMI-4a showed protective effects against BLM-induced mortality. Furthermore, SMI-4a suppressed hydroxyproline deposition and reversed epithelial-mesenchymal transition (EMT) formation, which was characterized by E-cadherin and α-SMA expression in vivo. More importantly, the ZEB1/E-cadherin pathway was found to be closely associated with BLM-induced pulmonary fibrosis. After the in vitro treatment of A549 cells, PIM1 regulated E-cadherin expression by dependently modulating the activity of the transcription factor ZEB1. These findings were verified in vivo after SMI-4a administration. Finally, an shPIM1-expressing adeno-associated virus was delivered via intratracheal injection to induce a long-term PIM1 deficiency in the alveolar epithelium. AAV-mediated PIM1 knockdown in the lung tissues alleviated BLM-induced pulmonary fibrosis, as indicated by collagen accumulation reduction, pulmonary histopathological mitigation and EMT reversion. These findings enhance our understanding of the roles of PIM1 in BLM-induced pulmonary fibrosis and suggest PIM1 inhibition as a potential therapeutic strategy in chronic pulmonary injuries.
Elsevier