Abstract

Laryngotracheal stenosis (LTS) is a life-threatening disease that commonly results in airway obstruction in children. Traditional treatments such as laryngotracheal reconstruction and balloon dilation all have the risk of laryngotracheal restenosis. It is of great importance to spare patients the morbidity of LTS and risks of restenosis associated with these treatments. Laboratory and clinical trials have focused on fibrosis, the crucial pathological process of LTS. This study was undertaken to investigate the function of CXC chemokine receptor-7 (CXCR7) in the fibroblasts derived from LTS. RNA sequencing was performed on acquired human LTS and normal trachea tissues to analyze differentially expressed genes. Fibroblasts from LTS and normal trachea tissues were isolated and cultured. CXCR7 knockdown was performed using specific small interfering RNAs (siRNAs) and activated by CXCR7 agonist VUF11207. The assessment of cell proliferation and migration was conducted using EdU proliferation, wound healing, and transwell assays. The assessment of cell proliferation and migration was conducted using EdU proliferation, wound healing, and transwell assays. The expressions of CXCR7, E-cadherin and NF-κB signaling pathway were analyzed by quantitative polymerase chain reaction (qPCR), western blotting, immunohistochemistry, and immunofluorescence. RNA sequencing showed that CXCR7 was among the most differentially expressed genes. LTS had an increased CXCR7 expression but decreased E-cadherin expression in vivo. CXCR7 agonist stimulated the migration of LTS derived fibroblasts significantly in vitro, with no significant influence on the cell proliferation and apoptosis. CXCR7 agonist inhibited the expression of E-cadherin by activating the NF-κB signaling pathway. The effects of CXCR7 on cell migration and E-cadherin expression were blocked by CXCR7 siRNA. LTS had an increased CXCR7 expression but decreased E-cadherin expression. CXCR7 activation inhibited E-cadherin expression by NF-κB signaling pathway and thereby promoted the migration of LTS derived fibroblasts.

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