Lysyl oxidase directly contributes to extracellular matrix production and fibrosis in systemic sclerosis

XX Nguyen, T Nishimoto, T Takihara… - … of Physiology-Lung …, 2021 - journals.physiology.org
XX Nguyen, T Nishimoto, T Takihara, L Mlakar, AD Bradshaw, C Feghali-Bostwick
American Journal of Physiology-Lung Cellular and Molecular …, 2021journals.physiology.org
Pulmonary fibrosis is one of the important causes of morbidity and mortality in
fibroproliferative disorders such as systemic sclerosis (SSc) and idiopathic pulmonary
fibrosis (IPF). Lysyl oxidase (LOX) is a copper-dependent amine oxidase whose primary
function is the covalent crosslinking of collagens in the extracellular matrix (ECM). We
investigated the role of LOX in the pathophysiology of SSc. LOX mRNA and protein levels
were increased in lung fibroblasts of SSc patients compared with healthy controls and IPF …
Pulmonary fibrosis is one of the important causes of morbidity and mortality in fibroproliferative disorders such as systemic sclerosis (SSc) and idiopathic pulmonary fibrosis (IPF). Lysyl oxidase (LOX) is a copper-dependent amine oxidase whose primary function is the covalent crosslinking of collagens in the extracellular matrix (ECM). We investigated the role of LOX in the pathophysiology of SSc. LOX mRNA and protein levels were increased in lung fibroblasts of SSc patients compared with healthy controls and IPF patients. In vivo, bleomycin induced LOX mRNA expression in lung tissues, and LOX activity increased in the circulation of mice with pulmonary fibrosis, suggesting that circulating LOX parallels levels in lung tissues. Circulating levels of LOX were reduced upon amelioration of fibrosis with an antifibrotic peptide. LOX induced ECM production at the transcriptional level in lung fibroblasts, human lungs, and human skin maintained in organ culture. In vivo, LOX synergistically exacerbated fibrosis in bleomycin-treated mice. Further, LOX increased the production of interleukin (IL)-6, and the increase was mediated by LOX-induced c-Fos expression, the nuclear localization of c-Fos, and its engagement with the IL-6 promoter region. Our findings demonstrate that LOX expression and activity correlate with fibrosis in vitro, ex vivo, and in vivo. LOX induced ECM production via upregulation of IL-6 and nuclear localization of c-Fos. Thus, LOX has a direct pathogenic role in SSc-associated fibrosis that is independent of its crosslinking function. Our findings also suggest that measuring circulating LOX levels and activity can be used for monitoring response to antifibrotic therapy.
American Physiological Society