Abstract

Idiopathic pulmonary fibrosis (IPF) is a progressive clinical syndrome of fatal outcome. The lack of information about the signaling pathways that sustain fibrosis and the myofibroblast phenotype has prevented the development of targeted therapies for IPF. Our previous study showed that isolated fibrogenic lung fibroblasts have high endogenous levels of the hyaluronan receptor, CD44V6 (CD44 variant containing exon 6), which enhances the TGFβ1 autocrine signaling and induces fibroblasts to transdifferentiate into myofibroblasts. NADPH oxidase 4 (NOX4) enzyme, which catalyzes the reduction of O2 to hydrogen peroxide (H2O2), has been implicated in the cardiac and lung myofibroblast phenotype. However, whether CD44V6 regulates NOX4 to mediate tissue repair and fibrogenesis is not well-defined. The present study assessed the mechanism of how TGF-β-1-induced CD44V6 regulates the NOX4/reactive oxygen species (ROS) signaling that mediates the myofibroblast differentiation. Specifically, we found that NOX4/ROS regulates hyaluronan synthesis and the transcription of CD44V6 via an effect upon AP-1 activity. Further, CD44V6 is part of a positive-feedback loop with TGFβ1/TGFβRI signaling that acts to increase NOX4/ROS production, which is required for myofibroblast differentiation, myofibroblast differentiation, myofibroblast extracellular matrix production, myofibroblast invasion, and myofibroblast contractility. Both NOX4 and CD44v6 are up-regulated in the lungs of mice subjected to experimental lung injury and in cases of human IPF. Genetic (CD44v6 shRNA) or a small molecule inhibitor (CD44v6 peptide) targeting of CD44v6 abrogates fibrogenesis in murine models of lung injury. These studies support a function for CD44V6 in lung fibrosis and offer proof of concept for therapeutic targeting of CD44V6 in lung fibrosis disorders.

Highlights

  • Idiopathic pulmonary fibrosis (IPF) is a progressive clinical syndrome of fatal outcome

  • The ability of TGF␤1 to stimulate cellular production of reactive oxygen species (H2O2) through NOX in human and mouse lung fibroblasts has been established [16], and NOX is activated through CD44 in an atherosclerosis model [90]

  • Because recent studies from our laboratory and other show that (i) proinflammatory TGF␤1 up-regulates CD44V6 [1], (ii) TGF␤1 up-regulates NADPH oxidase [16], and (iii) NOX-derived reactive oxygen species (ROS) regulates Ap-1 [101], we investigated whether Nox4derived ROS, CD44v6, and Ap-1 activation are interrelated during lung fibrosis

Read more

Summary

To whom correspondence may be addressed

Broblast differentiation, myofibroblast differentiation, myofibroblast extracellular matrix production, myofibroblast invasion, and myofibroblast contractility. Our recent study showed that a unique functional activity of CD44V6 includes stimulation of ERK activation, increased collagen-1 (COL1A1) synthesis, and induction of ␣-smooth-muscle actin (␣-SMA) in isolated lung myofibroblasts through TGF␤1 autocrine signaling in fibrogenic lung disease of human subjects [1]. Studies using IPF lung-derived fibroblasts (IPFFbs) demonstrated that TGF␤1-dependent profibrotic responses, including resistance to apoptosis, up-regulation of ␣-SMA gene expression, and the secretion of the ECM components fibronectin and COL1A1, are NOX4-dependent and contribute to persistent fibrosis in IPF disease [6, 15, 16]. We hypothesize that CD44V6 may be a critical determinant of the fate of fibrosis by promoting myofibroblast activation through its interaction with NOX4 in the fibroblasts from IPF patients and that this leads to the progressive fibrosis Proinflammatory cytokines, such as tumor necrosis factor ␣ (TNF␣), are key features of inflammatory processes and may. Our proof-of-concept data suggest that non-toxic/anti-inflammatory CD44V6 shRNA (V6 shRNA)/transferrin (Tf)-PEG-PEI (nanoparticles) or endogenous V6-PEP/nanoparticles abrogate fibrogenesis in a murine model of lung injury by reducing NOX4/ROS activity, reducing TNF-␣ production, and stimulating IL-10 production, with subsequent abrogation of transendothelial migration of leukocytes

Results
Discussion
Experimental procedures
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