24.02.2013 Views

The vagus nerve as a modulator of intestinal inflammation - TI Pharma

The vagus nerve as a modulator of intestinal inflammation - TI Pharma

The vagus nerve as a modulator of intestinal inflammation - TI Pharma

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

INTRODUC<strong>TI</strong>ON<br />

Vagal anti-inflammatory pathway mediated via JAK2-STAT3 activation<br />

<strong>The</strong> innate immune response h<strong>as</strong> been incre<strong>as</strong>ingly recognized <strong>as</strong> being under<br />

substantial neuronal control 1 . For example, acetylcholine or nicotine effectively<br />

attenuates the activation <strong>of</strong> macrophages 2 . This so-called ‘cholinergic antiinflammatory<br />

pathway’ is characterized by a nicotine dose−dependent decre<strong>as</strong>e in<br />

the production <strong>of</strong> proinflammatory mediators, including high-mobility group box 1<br />

proteins 3 , tumor necrosis factor (TNF), interleukin 1β (IL-1β), IL-6 and IL-18 (ref 2),<br />

by macrophages stimulated with endotoxin. Consistently, stimulation <strong>of</strong> the efferent<br />

<strong>vagus</strong> <strong>nerve</strong> dampens macrophage activation in rodent models <strong>of</strong> endotoxemia and<br />

shock 1,2 . Two nicotinic acetylcholine receptor (nAChR) subtypes are involved in the<br />

nicotine-induced decre<strong>as</strong>e in proinflammatory cytokine production by stimulated<br />

human and mouse macrophages: the α7 homopentamer expressed by monocytederived<br />

human and mouse macrophages 4 , and the α4β2 heteropentamer expressed<br />

by alveolar macrophages 5 . Activation <strong>of</strong> the α7 homopentamer nAChR inhibits<br />

transactivational activity <strong>of</strong> the transcription factor NF-ĸB p65 (ref 3). However,<br />

the subcellular mechanism explaining the deactivating effect <strong>of</strong> acetylcholine on<br />

macrophages h<strong>as</strong> remained unknown.<br />

Here we evaluated the involvement <strong>of</strong> the transcription factor STAT3 in<br />

this process, because STAT3 is a potential negative regulator <strong>of</strong> inflammatory<br />

responses 6,7 . STAT3 and the tyrosine kin<strong>as</strong>e Jak2, which phosphorylates STAT3, are<br />

required for both IL-6 receptor (IL-6R) and IL-10R signaling. IL-6 contributes to the<br />

progression <strong>of</strong> many inflammatory dise<strong>as</strong>es, where<strong>as</strong> IL-10 is an anti-inflammatory<br />

cytokine that suppresses the activation <strong>of</strong> macrophages. IL-6R signaling is inhibited<br />

by the Src homology 2 domain protein SOCS3, whose expression is induced by<br />

STAT3 activation 8,9 . SOCS3 binds to the glycoprotein 130 (gp130) subunit <strong>of</strong> the<br />

IL-6R, leading to inhibited activation <strong>of</strong> STAT3 by IL-6R ligands 8,9 . Consistent with<br />

that finding, in LPS-stimulated macrophages deficient in SOCS3, IL-6R ligands<br />

induce a sustained STAT3 activation, which leads to the reduced production <strong>of</strong><br />

proinflammatory cytokines such <strong>as</strong> TNF 10 .<br />

Here we demonstrate that nicotine exerts its anti-inflammatory effect on<br />

peritoneal macrophages via Jak2 and STAT3 signaling in vitro and in vivo. In isolated<br />

peritoneal macrophages, nicotine activated nAChRs, leading to phosphorylation<br />

<strong>of</strong> STAT3 via Jak2. Jak2 w<strong>as</strong> recruited to the α7 subunit <strong>of</strong> the nAChR and w<strong>as</strong><br />

phosphorylated after nicotine binding. We further studied the effect <strong>of</strong> cholinergic<br />

inhibition <strong>of</strong> macrophage activity in vivo on the occurrence <strong>of</strong> post-surgical <strong>intestinal</strong><br />

<strong>inflammation</strong> in a mouse model <strong>of</strong> postoperative ileus 11,12 . Postoperative ileus is<br />

characterized by general hypomotility <strong>of</strong> the g<strong>as</strong>tro<strong>intestinal</strong> tract and delayed g<strong>as</strong>tric<br />

emptying 13 and is a pathological condition commonly noted after abdominal surgery<br />

with <strong>intestinal</strong> manipulation. This condition is the result <strong>of</strong> <strong>inflammation</strong> <strong>of</strong> the<br />

<strong>intestinal</strong> muscularis due to activation <strong>of</strong> resident macrophages 14,15 that are triggered<br />

41

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!