Abstract

In inflammation, pain is regulated by a balance of pro- and analgesic mediators. Analgesic mediators include opioid peptides which are secreted by neutrophils at the site of inflammation, leading to activation of opioid receptors on peripheral sensory neurons. In humans, local opioids and opioid peptides significantly downregulate postoperative as well as arthritic pain. In rats, inflammatory pain is induced by intraplantar injection of heat inactivated Mycobacterium butyricum, a component of complete Freund's adjuvant. We hypothesized that mycobacterially derived formyl peptide receptor (FPR) and/or toll like receptor (TLR) agonists could activate neutrophils, leading to opioid peptide release and inhibition of inflammatory pain. In complete Freund's adjuvant-induced inflammation, thermal and mechanical nociceptive thresholds of the paw were quantified (Hargreaves and Randall-Selitto methods, respectively). Withdrawal time to heat was decreased following systemic neutrophil depletion as well as local injection of opioid receptor antagonists or anti-opioid peptide (i.e. Met-enkephalin, β-endorphin) antibodies indicating an increase in pain. In vitro, opioid peptide release from human and rat neutrophils was measured by radioimmunoassay. Met-enkephalin release was triggered by Mycobacterium butyricum and formyl peptides but not by TLR-2 or TLR-4 agonists. Mycobacterium butyricum induced a rise in intracellular calcium as determined by FURA loading and calcium imaging. Opioid peptide release was blocked by intracellular calcium chelation as well as phosphoinositol-3-kinase inhibition. The FPR antagonists Boc-FLFLF and cyclosporine H reduced opioid peptide release in vitro and increased inflammatory pain in vivo while TLR 2/4 did not appear to be involved. In summary, mycobacteria activate FPR on neutrophils, resulting in tonic secretion of opioid peptides from neutrophils and in a decrease in inflammatory pain. Future therapeutic strategies may aim at selective FPR agonists to boost endogenous analgesia.

Highlights

  • The four cardinal signs of inflammation are rubor, calor, dolor and functio laesa

  • Previous research indicated that mycobacteria enhance nociceptive responses via toll like receptors-2 and -4

  • We demonstrate that mycobacteria activate formyl peptide receptors on neutrophils leading to opioid peptide release and the inhibition of such responses

Read more

Summary

Introduction

The four cardinal signs of inflammation are rubor (redness), calor (hyperthermia), dolor (pain/hyperalgesia) and functio laesa (impaired function). Bacteria and their components play a critical role in eliciting pain since inflammatory pain is significantly decreased in animals raised under germ free conditions [1]. Pain is elicited by proalgesic mediators including proinflammatory cytokines (tumor necrosis factor-a, interleukin-1b), bradykinin, and protons [2,4]. Bacteria and their components are recognized by pattern recognition receptors including toll like receptors (TLR) as well as formyl peptide receptors (FPR). In contrast to these pronociceptive effects of TLR agonists, FPR agonists were shown to decrease pain induced by formalin, but the underlying mechanism remained unclear [11]

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