Abstract

The molecular mechanisms determining magnitude and duration of inflammatory pain are still unclear. We assessed the contribution of G protein-coupled receptor kinase (GRK)-6 to inflammatory hyperalgesia in mice. We showed that GRK6 is a critical regulator of severity and duration of cytokine-induced hyperalgesia. In GRK6⁻/⁻ mice, a significantly lower dose (100 times lower) of intraplantar interleukin (IL)-1β was sufficient to induce hyperalgesia compared with wild-type (WT) mice. In addition, IL-1β hyperalgesia lasted much longer in GRK6⁻/⁻ mice than in WT mice (8 d in GRK6⁻/⁻ versus 6 h in WT mice). Tumor necrosis factor (TNF)-α-induced hyperalgesia was also enhanced and prolonged in GRK6⁻/⁻ mice. In vitro, IL-1β-induced p38 phosphorylation in GRK6⁻/⁻ dorsal root ganglion (DRG) neurons was increased compared with WT neurons. In contrast, IL-1β only induced activation of the phosphatidylinositol (PI) 3-kinase/Akt pathway in WT neurons, but not in GRK6⁻/⁻ neurons. In vivo, p38 inhibition attenuated IL-1β- and TNF-α-induced hyperalgesia in both genotypes. Notably, however, whereas PI 3-kinase inhibition enhanced and prolonged hyperalgesia in WT mice, it did not have any effect in GRK6-deficient mice. The capacity of GRK6 to regulate pain responses was also apparent in carrageenan-induced hyperalgesia, since thermal and mechanical hypersensitivity was significantly prolonged in GRK6⁻/⁻ mice. Finally, GRK6 expression was reduced in DRGs of mice with chronic neuropathic or inflammatory pain. Collectively, these findings underline the potential role of GRK6 in pathological pain. We propose the novel concept that GRK6 acts as a kinase that constrains neuronal responsiveness to IL-1β and TNF-α and cytokine-induced hyperalgesia via biased cytokine-induced p38 and PI 3-kinase/Akt activation.

Highlights

  • Proper functioning of the nociceptive system is essential to protect the body from tissue damage

  • To investigate whether changes in GRK6 do occur in conditions of chronic pain, we investigated GRK6 mRNA expression levels in Dorsal root ganglions (DRGs) of mice with chronic neuropathic or inflammatory pain

  • The novelty and significance of these results is that GRK6 emerges here as a kinase that constrains neuronal responsiveness to IL-1β and tumor necrosis factor (TNF)-α and cytokine-induced hyperalgesia via biased cytokine-induced p38 and PI 3-kinase/Akt activation (Figure 6E)

Read more

Summary

Introduction

Proper functioning of the nociceptive system is essential to protect the body from tissue damage. Inflammation sensitizes the nociceptive system, leading to a lower threshold to painful stimuli (hyperalgesia) [1]. This process is thought to serve adaptive purposes, but becomes maladaptive when hyperalgesia persists after resolution of inflammation. The proinflammatory cytokine interleukin (IL)-1β directly sensitizes nociceptors, leading to transient hyperalgesia [2,3]. Tory mediators such as prostaglandin E2 (PGE2) increases the sensitivity of nociceptors and the response to painful stimuli [4]. There is evidence that proinflammatory cytokines such as IL-1β and tumor necrosis factor (TNF)-α contribute to the genesis of neuropathic pain [5,6]

Objectives
Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.