Abstract

BackgroundThe aryl hydrocarbon receptor (AhR) has gradually emerged as a regulator of inflammation in the lung and other tissues. AhR may interact with the p65-subunit of the nuclear factor (NF)-κB transcription factors, but reported outcomes of AhR/NF-κB-interactions are conflicting. Some studies suggest that AhR possess pro-inflammatory activities while others suggest that AhR may be anti-inflammatory. The present study explored the impact of AhR and its binding partner AhR nuclear translocator (Arnt) on p65-activation and two differentially regulated chemokines, CXCL8 (IL-8) and CCL5 (RANTES), in human bronchial epithelial cells (BEAS-2B).ResultsCells were exposed to CXCL8- and CCL5-inducing chemicals, 1-nitropyrene (1-NP) and 1-aminopyrene (1-AP) respectively, or the synthetic double-stranded RNA analogue, polyinosinic-polycytidylic acid (Poly I:C) which induced both chemokines. Only CXCL8, and not CCL5, appeared to be p65-dependent. Yet, constitutively active unligated AhR suppressed both CXCL8 and CCL5, as shown by siRNA knock-down and the AhR antagonist α-naphthoflavone. Moreover, AhR suppressed activation of p65 by TNF-α and Poly I:C as assessed by luciferase-assay and p65-phosphorylation at serine 536, without affecting basal p65-activity. In contrast, Arnt suppressed only CXCL8, but did not prevent the p65-activation directly. However, Arnt suppressed expression of the NF-κB-subunit RelB which is under transcriptional regulation by p65. Furthermore, AhR-ligands alone at high concentrations induced a moderate CXCL8-response, without affecting CCL5, but suppressed both CXCL8 and CCL5-responses by Poly I:C.ConclusionAhR and Arnt may differentially and independently regulate chemokine-responses induced by both inhaled pollutants and pulmonary infections. Constitutively active, unligated AhR suppressed the activation of p65, while Arnt may possibly interfere with the action of activated p65. Moreover, ligand-activated AhR suppressed CXCL8 and CCL5 responses by other agents, but AhR ligands alone induced CXCL8 responses when given at sufficiently high concentrations, thus underscoring the duality of AhR in regulation of inflammation. We propose that AhR-signaling may be a weak activator of p65-signaling that suppresses p65-activity induced by strong activators of NF-κB, but that its anti-inflammatory properties also are due to interference with additional pathways.

Highlights

  • The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor originally discovered as a receptor for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and other environmental pollutants, including polycyclic aromatic hydrocarbons (PAHs)

  • Active, unligated AhR suppressed the activation of p65, while AhR nuclear translocator (Arnt) may possibly interfere with the action of activated p65

  • We propose that AhR-signaling may be a weak activator of p65-signaling that suppresses p65-activity induced by strong activators of nuclear factor (NF)-κB, but that its anti-inflammatory properties are due to interference with additional pathways

Read more

Summary

Introduction

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor originally discovered as a receptor for 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and other environmental pollutants, including polycyclic aromatic hydrocarbons (PAHs). In its classical mode of action, cytosolic AhR activated by hydrocarbons rapidly translocates to the nucleus where it dimerizes with the AhR nuclear translocator (Arnt). It has been suggested that increases in such chronic disorders at least partly may be related to inhalation of environmental pollutants, contributing to development or exacerbation of inflammation [4,5,6]. The aryl hydrocarbon receptor (AhR) has gradually emerged as a regulator of inflammation in the lung and other tissues. The present study explored the impact of AhR and its binding partner AhR nuclear translocator (Arnt) on p65-activation and two differentially regulated chemokines, CXCL8 (IL-8) and CCL5 (RANTES), in human bronchial epithelial cells (BEAS-2B)

Methods
Results
Discussion
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