Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Resolvins RvE1 and RvD1 attenuate inflammatory pain via central and peripheral actions

Abstract

Inflammatory pain, such as arthritis pain, is a growing health problem1. Inflammatory pain is generally treated with opioids and cyclooxygenase (COX) inhibitors, but both are limited by side effects. Recently, resolvins, a unique family of lipid mediators, including RvE1 and RvD1 derived from omega-3 polyunsaturated fatty acid, have shown marked potency in treating disease conditions associated with inflammation2,3. Here we report that peripheral (intraplantar) or spinal (intrathecal) administration of RvE1 or RvD1 in mice potently reduces inflammatory pain behaviors induced by intraplantar injection of formalin, carrageenan or complete Freund's adjuvant (CFA), without affecting basal pain perception. Intrathecal RvE1 injection also inhibits spontaneous pain and heat and mechanical hypersensitivity evoked by intrathecal capsaicin and tumor necrosis factor-α (TNF-α). RvE1 has anti-inflammatory activity by reducing neutrophil infiltration, paw edema and proinflammatory cytokine expression. RvE1 also abolishes transient receptor potential vanilloid subtype-1 (TRPV1)- and TNF-α–induced excitatory postsynaptic current increases and TNF-α–evoked N-methyl-D-aspartic acid (NMDA) receptor hyperactivity in spinal dorsal horn neurons via inhibition of the extracellular signal–regulated kinase (ERK) signaling pathway. Thus, we show a previously unknown role for resolvins in normalizing the spinal synaptic plasticity that has been implicated in generating pain hypersensitivity. Given the potency of resolvins and the well-known side effects of opioids and COX inhibitors, resolvins may represent new analgesics for treating inflammatory pain.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Preemptive spinal (intrathecal) administration of RvE1 reduces the second phase of formalin-induced inflammatory pain.
Figure 2: Central and peripheral actions of resolvins on persistent inflammatory pain and inflammation.
Figure 3: Spinal administration of RvE1 reduces heat hyperalgesia and spontaneous pain by blocking TRPV1 and TNF-α signaling in DRG neurons and spinal presynaptic terminals.
Figure 4: Spinal RvE1 administration attenuates mechanical allodynia and blocks TNF-α signaling in postsynaptic dorsal horn neurons.

References

  1. Schnitzer, T.J. Update on guidelines for the treatment of chronic musculoskeletal pain. Clin. Rheumatol. 25 Suppl 1, S22–S29 (2006).

    Article  Google Scholar 

  2. Serhan, C.N., Chiang, N. & Van Dyke, T.E. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nat. Rev. Immunol. 8, 349–361 (2008).

    Article  CAS  Google Scholar 

  3. Connor, K.M. et al. Increased dietary intake of omega-3-polyunsaturated fatty acids reduces pathological retinal angiogenesis. Nat. Med. 13, 868–873 (2007).

    Article  CAS  Google Scholar 

  4. Gilroy, D.W. et al. Inducible cyclooxygenase may have anti-inflammatory properties. Nat. Med. 5, 698–701 (1999).

    Article  CAS  Google Scholar 

  5. Schwab, J.M., Chiang, N., Arita, M. & Serhan, C.N. Resolvin E1 and protectin D1 activate inflammation-resolution programmes. Nature 447, 869–874 (2007).

    Article  CAS  Google Scholar 

  6. Chiang, N. et al. Anesthetics impact the resolution of inflammation. PLoS One 3, e1879 (2008).

    Article  Google Scholar 

  7. Haworth, O., Cernadas, M., Yang, R., Serhan, C.N. & Levy, B.D. Resolvin E1 regulates interleukin 23, interferon-γ and lipoxin A4 to promote the resolution of allergic airway inflammation. Nat. Immunol. 9, 873–879 (2008).

    Article  CAS  Google Scholar 

  8. Julius, D. & Basbaum, A.I. Molecular mechanisms of nociception. Nature 413, 203–210 (2001).

    Article  CAS  Google Scholar 

  9. Hucho, T. & Levine, J.D. Signaling pathways in sensitization: toward a nociceptor cell biology. Neuron 55, 365–376 (2007).

    Article  CAS  Google Scholar 

  10. Woolf, C.J. & Salter, M.W. Neuronal plasticity: increasing the gain in pain. Science 288, 1765–1769 (2000).

    Article  CAS  Google Scholar 

  11. Ji, R.R., Kohno, T., Moore, K.A. & Woolf, C.J. Central sensitization and LTP: do pain and memory share similar mechanisms? Trends Neurosci. 26, 696–705 (2003).

    Article  CAS  Google Scholar 

  12. Yamamoto, T. & Yaksh, T.L. Comparison of the antinociceptive effects of pre- and posttreatment with intrathecal morphine and MK801, an NMDA antagonist, on the formalin test in the rat. Anesthesiology 77, 757–763 (1992).

    Article  CAS  Google Scholar 

  13. Ji, R.R., Baba, H., Brenner, G.J. & Woolf, C.J. Nociceptive-specific activation of ERK in spinal neurons contributes to pain hypersensitivity. Nat. Neurosci. 2, 1114–1119 (1999).

    Article  CAS  Google Scholar 

  14. Yaksh, T.L. Spinal systems and pain processing: development of novel analgesic drugs with mechanistically defined models. Trends Pharmacol. Sci. 20, 329–337 (1999).

    Article  CAS  Google Scholar 

  15. Kawasaki, Y. et al. Distinct roles of matrix metalloproteases in the early- and late-phase development of neuropathic pain. Nat. Med. 14, 331–336 (2008).

    Article  CAS  Google Scholar 

  16. Arita, M. et al. Stereochemical assignment, antiinflammatory properties, and receptor for the omega-3 lipid mediator resolvin E1. J. Exp. Med. 201, 713–722 (2005).

    Article  CAS  Google Scholar 

  17. Cash, J.L. et al. Synthetic chemerin-derived peptides suppress inflammation through ChemR23. J. Exp. Med. 205, 767–775 (2008).

    Article  CAS  Google Scholar 

  18. Wittamer, V. et al. Specific recruitment of antigen-presenting cells by chemerin, a novel processed ligand from human inflammatory fluids. J. Exp. Med. 198, 977–985 (2003).

    Article  CAS  Google Scholar 

  19. Liu, X.J. et al. Treatment of inflammatory and neuropathic pain by uncoupling Src from the NMDA receptor complex. Nat. Med. 14, 1325–1332 (2008).

    Article  CAS  Google Scholar 

  20. Goldberg, R.J. & Katz, J. A meta-analysis of the analgesic effects of omega-3 polyunsaturated fatty acid supplementation for inflammatory joint pain. Pain 129, 210–223 (2007).

    Article  CAS  Google Scholar 

  21. Arita, M. et al. Metabolic inactivation of resolvin E1 and stabilization of its anti-inflammatory actions. J. Biol. Chem. 281, 22847–22854 (2006).

    Article  CAS  Google Scholar 

  22. Schäfers, M., Svensson, C.I., Sommer, C. & Sorkin, L.S. Tumor necrosis factor-α induces mechanical allodynia after spinal nerve ligation by activation of p38 MAPK in primary sensory neurons. J. Neurosci. 23, 2517–2521 (2003).

    Article  Google Scholar 

  23. Junger, H. & Sorkin, L.S. Nociceptive and inflammatory effects of subcutaneous TNFα. Pain 85, 145–151 (2000).

    Article  CAS  Google Scholar 

  24. Kawasaki, Y., Zhang, L., Cheng, J.K. & Ji, R.R. Cytokine mechanisms of central sensitization: distinct and overlapping role of interleukin-1β, interleukin-6, and tumor necrosis factor-α in regulating synaptic and neuronal activity in the superficial spinal cord. J. Neurosci. 28, 5189–5194 (2008).

    Article  CAS  Google Scholar 

  25. Caterina, M.J. et al. Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science 288, 306–313 (2000).

    Article  CAS  Google Scholar 

  26. Nicol, G.D., Lopshire, J.C. & Pafford, C.M. Tumor necrosis factor enhances the capsaicin sensitivity of rat sensory neurons. J. Neurosci. 17, 975–982 (1997).

    Article  CAS  Google Scholar 

  27. Jin, X. & Gereau, R.W. Acute p38-mediated modulation of tetrodotoxin-resistant sodium channels in mouse sensory neurons by tumor necrosis factor-α. J. Neurosci. 26, 246–255 (2006).

    Article  CAS  Google Scholar 

  28. Zhuang, Z.Y., Xu, H., Clapham, D.E. & Ji, R.R. Phosphatidylinositol 3-kinase activates ERK in primary sensory neurons and mediates inflammatory heat hyperalgesia through TRPV1 sensitization. J. Neurosci. 24, 8300–8309 (2004).

    Article  CAS  Google Scholar 

  29. Vara, H., Onofri, F., Benfenati, F., Sassoe-Pognetto, M. & Giustetto, M. ERK activation in axonal varicosities modulates presynaptic plasticity in the CA3 region of the hippocampus through synapsin I. Proc. Natl. Acad. Sci. USA 106, 9872–9877 (2009).

    Article  CAS  Google Scholar 

  30. Karim, F., Wang, C.C. & Gereau, R.W. Metabotropic glutamate receptor subtypes 1 and 5 are activators of extracellular signal–regulated kinase signaling required for inflammatory pain in mice. J. Neurosci. 21, 3771–3779 (2001).

    Article  CAS  Google Scholar 

  31. Samad, T.A. et al. Interleukin-1β–mediated induction of Cox-2 in the CNS contributes to inflammatory pain hypersensitivity. Nature 410, 471–475 (2001).

    Article  CAS  Google Scholar 

  32. Woolf, C.J. μ and δ opioid receptors diverge. Cell 137, 987–988 (2009).

    Article  CAS  Google Scholar 

  33. Porreca, F. & Ossipov, M.H. Nausea and vomiting side effects with opioid analgesics during treatment of chronic pain: mechanisms, implications and management options. Pain Med. 10, 654–662 (2009).

    Article  Google Scholar 

  34. Funk, C.D. & FitzGerald, G.A. COX-2 inhibitors and cardiovascular risk. J. Cardiovasc. Pharmacol. 50, 470–479 (2007).

    Article  CAS  Google Scholar 

  35. Gavva, N.R. Body-temperature maintenance as the predominant function of the vanilloid receptor TRPV1. Trends Pharmacol. Sci. 29, 550–557 (2008).

    Article  CAS  Google Scholar 

  36. Brennan, T.J., Vandermeulen, E.P. & Gebhart, G.F. Characterization of a rat model of incisional pain. Pain 64, 493–501 (1996).

    Article  CAS  Google Scholar 

  37. Kim, S.H. & Chung, J.M. An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain 50, 355–363 (1992).

    Article  CAS  Google Scholar 

  38. Hylden, J.L. & Wilcox, G.L. Intrathecal morphine in mice: a new technique. Eur. J. Pharmacol. 67, 313–316 (1980).

    Article  CAS  Google Scholar 

  39. Gao, Y.J. et al. JNK-induced MCP-1 production in spinal cord astrocytes contributes to central sensitization and neuropathic pain. J. Neurosci. 29, 4096–4108 (2009).

    Article  CAS  Google Scholar 

  40. Dixon, W.J. Efficient analysis of experimental observations. Annu. Rev. Pharmacol. Toxicol. 20, 441–462 (1980).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported in part by US National Institutes of Health grants R01-DE17794 and R01-NS54362 to R.-R.J., R37 GM38765, R01-DE019938 and R01-DK074448 to C.N.S. and NS67686 to both R.-R.J. and C.N.S.

Author information

Authors and Affiliations

Authors

Contributions

R.-R.J. and C.N.S. formulated the hypotheses, designed and supervised the project and prepared the manuscript; R.-R.J. designed most experiments; Z.-Z.X., T.L. and J.Y.P. conducted behavioral studies; L.Z. performed electrophysiological studies; Z.-Z.X. and L.Z. performed immunohistochemistry; Z.-Z.X. performed siRNA-mediated knockdown, cytokine arrays and western blotting; T.B. performed in situ hybridization; R.Y. prepared resolvins and their analogs.

Corresponding authors

Correspondence to Charles N Serhan or Ru-Rong Ji.

Ethics declarations

Competing interests

Resolvins are biotemplates for stable analogs. Patents on these are awarded and assigned to the Brigham and Women's Hospital, and C.N.S. is the inventor. These patents are licensed for clinical development.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Methods (PDF 810 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Xu, ZZ., Zhang, L., Liu, T. et al. Resolvins RvE1 and RvD1 attenuate inflammatory pain via central and peripheral actions. Nat Med 16, 592–597 (2010). https://doi.org/10.1038/nm.2123

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2123

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing