Abstract

BackgroundPerturbation of endothelial function in people with cystic fibrosis (CF) has been reported, which may be associated with endothelial cell expression of the cystic fibrosis transmembrane conductance regulator (CFTR). Previous reports indicate that CFTR activity upregulates endothelial barrier function, endothelial nitric oxide synthase (eNOS) expression and NO release, while limiting interleukin-8 (IL-8) release, in human umbilical vein endothelial cells (HUVECs) in cell culture. In view of reported microvascular dysfunction in people with CF we investigated the role of CFTR expression and activity in the regulation of oxidative stress, cell signaling and inflammation in human lung microvascular endothelial cells (HLMVECs) in cell culture.MethodsHLMVECs were cultured in the absence and presence of the CFTR inhibitor GlyH-101 and CFTR siRNA. CFTR expression was analyzed using qRT-PCR, immunocytochemistry (IHC) and western blot, and function by membrane potential assay. IL-8 expression was analyzed using qRT-PCR and ELISA. Nrf2 expression, and NF-κB and AP-1 activation were determined using IHC and western blot. The role of the epidermal growth factor receptor (EGFR) in CFTR signaling was investigated using the EGFR tyrosine kinase inhibitor AG1478. Oxidative stress was measured as intracellular ROS and hydrogen peroxide (H2O2) concentration. VEGF and SOD-2 were measured in culture supernatants by ELISA.ResultsHLMVECs express low levels of CFTR that increase following inhibition of CFTR activity. Inhibition of CFTR, significantly increased intracellular ROS and H2O2 levels over 30 min and significantly decreased Nrf2 expression by 70% while increasing SOD-2 expression over 24 h. CFTR siRNA significantly increased constitutive expression of IL-8 by HLMVECs. CFTR inhibition activated the AP-1 pathway and increased IL-8 expression, without effect on NF-κB activity. Conversely, TNF-α activated the NF-κB pathway and increased IL-8 expression. The effects of TNF-α and GlyH-101 on IL-8 expression were additive and inhibited by AG1478. Inhibition of both CFTR and EGFR in HLMVECs significantly increased VEGF expression. The antioxidant N-acetyl cysteine significantly reduced ROS production and the increase in IL-8 and VEGF expression following CFTR inhibition.ConclusionFunctional endothelial CFTR limits oxidative stress and contributes to the normal anti-inflammatory state of HLMVECs. Therapeutic strategies to restore endothelial CFTR function in CF are warranted.

Highlights

  • Cystic fibrosis (CF) lung disease is associated with neutrophilic airway inflammation, bronchiectasis, respiratory failure, and early mortality (Nichols and Chmiel, 2015)

  • SOD-2 expression is not reported to be regulated by Nrf2 (Türei et al, 2013) and, considering the loss of Nrf2 function over 16–24 h (Figure 5), we propose that SOD-2 expression is upregulated in response to H2O2 mediated activation of other redox sensitive transcription factors (Yoshioka et al, 1994)

  • The findings in the present study clearly indicate that such an approach further enhances the increased vascular endothelial growth factor (VEGF) expression observed following CFTR inhibition in human lung microvascular endothelial cells (HLMVECs), and in this respect these endothelial cells differ from epithelial cells

Read more

Summary

Introduction

Cystic fibrosis (CF) lung disease is associated with neutrophilic airway inflammation, bronchiectasis, respiratory failure, and early mortality (Nichols and Chmiel, 2015). Increased circulating levels of von Willebrand factor (vWF) and tissue plasminogen activator (tPA), indicative of endothelial damage and altered hemostasis (Romano et al, 2001), and reduced flow-mediated dilation of the brachial artery, which was associated with more severe airway disease and symptomatic of reduced NO availability (Poore et al, 2013), were reported in CF. In view of reported microvascular dysfunction in people with CF we investigated the role of CFTR expression and activity in the regulation of oxidative stress, cell signaling and inflammation in human lung microvascular endothelial cells (HLMVECs) in cell culture

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