Abstract

α7 Nicotinic acetylcholine receptors (nAChRs) reportedly reduce inflammation by blocking effects of the important pro-inflammatory transcription factor, nuclear factor kappa-light chain-enhancer of B cells (NFκB). The α7 nAChR partial agonist GTS-21 reduces secretion of pro-inflammatory cytokines including interleukin-6 (IL6) and tumor-necrosis factor (TNF) in models of endotoxemia and sepsis, and its anti-inflammatory effects are widely ascribed to α7 nAChR activation. However, mechanistic details of α7 nAChR involvement in GTS-21 effects on inflammatory pathways remain unclear. Here, we investigate how GTS-21 acts in two cell systems including the non-immune rat pituitary cell line GH4C1 expressing an NFκB-driven reporter gene and cytokine secretion by ex vivo cultures of primary mouse macrophages activated by lipopolysaccharide (LPS). GTS-21 does not change TNF-stimulated NFκB signaling in GH4C1 cells expressing rat α7 nAChRs, suggesting that GTS-21 requires additional unidentified factors besides α7 nAChR expression to allow anti-inflammatory effects in these cells. In contrast, GTS-21 dose-dependently suppresses LPS-induced IL6 and TNF secretion in primary mouse macrophages endogenously expressing α7 nAChRs. GTS-21 also blocks TNF-induced phosphorylation of NFκB inhibitor alpha (IκBα), an important intermediary in NFκB signaling. However, α7 antagonists methyllycaconitine and α-bungarotoxin only partially reverse GTS-21 blockade of IL6 and TNF secretion. Further, GTS-21 significantly inhibited LPS-induced IL6 and TNF secretion in macrophages isolated from knockout mice lacking α7 nAChRs. These data indicate that even though a discrete component of the anti-inflammatory effects of GTS-21 requires expression of α7 nAChRs in macrophages, GTS-21 also has anti-inflammatory effects independent of these receptors depending on the cellular context.

Highlights

  • We investigated whether GTS-21 causes anti-inflammatory signaling in rat GH4C1 cells, a pituitary-derived cell line transfected with α7 nicotinic acetylcholine receptors (nAChRs)

  • GH4C1 cells transfected with α7 nAChR, but not WT cells, exhibit surface 125I-αBGT binding (Fig 1A)

  • A 1 h treatment of cells transfected with both α7 nAChR and an NFκB-driven Secreted alkaline phosphatase (SEAP)

Read more

Summary

Introduction

Follow-up studies have since established that during macrophage activation, the afferent vagus nerve senses pro-inflammatory mediators in the periphery to relay the message to the brain which releases acetylcholine via efferent vagus activity to inhibit inflammation locally through α7 nAChRs activation [9]. This pathway is known as the ‘cholinergic antiinflammatory pathway’ [9, 10] and has increased interest in α7 nAChR agonists, such as GTS21, as potential anti-inflammatory drugs

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