scholarly journals The isoprenoid end product N6-isopentenyladenosine reduces inflammatory response through the inhibition of the NFκB and STAT3 pathways in cystic fibrosis cells

2017 ◽  
Vol 67 (4) ◽  
pp. 315-326 ◽  
Author(s):  
Antonietta Santoro ◽  
Elena Ciaglia ◽  
Vanessa Nicolin ◽  
Alessandra Pescatore ◽  
Lucia Prota ◽  
...  
2018 ◽  
Vol 314 (4) ◽  
pp. L635-L641 ◽  
Author(s):  
Manon Ruffin ◽  
Lucie Roussel ◽  
Émilie Maillé ◽  
Simon Rousseau ◽  
Emmanuelle Brochiero

Cystic fibrosis patients exhibit chronic Pseudomonas aeruginosa respiratory infections and sustained proinflammatory state favoring lung tissue damage and remodeling, ultimately leading to respiratory failure. Loss of cystic fibrosis transmembrane conductance regulator (CFTR) function is associated with MAPK hyperactivation and increased cytokines expression, such as interleukin-8 [chemoattractant chemokine (C-X-C motif) ligand 8 (CXCL8)]. Recently, new therapeutic strategies directly targeting the basic CFTR defect have been developed, and ORKAMBI (Vx-809/Vx-770 combination) is the only Food and Drug Administration-approved treatment for CF patients homozygous for the F508del mutation. Here we aimed to determine the effect of the Vx-809/Vx-770 combination on the induction of the inflammatory response by fully differentiated primary bronchial epithelial cell cultures from CF patients carrying F508del mutations, following exposure to P. aeruginosa exoproducts. Our data unveiled that CFTR functional rescue with Vx-809/Vx-770 drastically reduces CXCL8 (as well as CXCL1 and CXCL2) transcripts and p38 MAPK phosphorylation in response to P. aeruginosa exposure through a CFTR-dependent mechanism. These results suggest that ORKAMBI has anti-inflammatory properties that could decrease lung inflammation and contribute to the observed beneficial impact of this treatment in CF patients.


2011 ◽  
Vol 10 (6) ◽  
pp. 401-406 ◽  
Author(s):  
James A. Reihill ◽  
John E. Moore ◽  
J. Stuart Elborn ◽  
Madeleine Ennis

PEDIATRICS ◽  
1969 ◽  
Vol 44 (2) ◽  
pp. 305-306
Author(s):  
A. Myron Johnson

It appears that Dr. Strober and I disagree on the implications of the word "cause" (Pediatrics, 44:144, 1969). The paper in question did not even consider the possibility that some other factor(s) could result in increased blood volume and decreased albumin synthesis concurrently. One possible third factor is the acute inflammatory response. If such be the case, even normal albumin synthesis may represent a compensatory response to the hypervolemia. The arguments in Dr. Strober's reply are based, in part, on the speculation that the liver is incapable of producing albumin at a rate much above the normal, resting rate.


2016 ◽  
Vol 113 (26) ◽  
pp. E3725-E3734 ◽  
Author(s):  
Beth Malcomson ◽  
Hollie Wilson ◽  
Eleonora Veglia ◽  
Gayathri Thillaiyampalam ◽  
Ruth Barsden ◽  
...  

Cystic fibrosis (CF) lung disease is characterized by chronic and exaggerated inflammation in the airways. Despite recent developments to therapeutically overcome the underlying functional defect in the cystic fibrosis transmembrane conductance regulator, there is still an unmet need to also normalize the inflammatory response. The prolonged and heightened inflammatory response in CF is, in part, mediated by a lack of intrinsic down-regulation of the proinflammatory NF-κB pathway. We have previously identified reduced expression of the NF-κB down-regulator A20 in CF as a key target to normalize the inflammatory response. Here, we have used publicly available gene array expression data together with a statistically significant connections’ map (sscMap) to successfully predict drugs already licensed for the use in humans to induce A20 mRNA and protein expression and thereby reduce inflammation. The effect of the predicted drugs on A20 and NF-κB(p65) expression (mRNA) as well as proinflammatory cytokine release (IL-8) in the presence and absence of bacterial LPS was shown in bronchial epithelial cells lines (16HBE14o−, CFBE41o−) and in primary nasal epithelial cells from patients with CF (Phe508del homozygous) and non-CF controls. Additionally, the specificity of the drug action on A20 was confirmed using cell lines with tnfαip3 (A20) knockdown (siRNA). We also show that the A20-inducing effect of ikarugamycin and quercetin is lower in CF-derived airway epithelial cells than in non-CF cells.


Sign in / Sign up

Export Citation Format

Share Document