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.

  • Letter
  • Published:

Inhibition of mechanical allodynia in neuropathic pain by TLR5-mediated A-fiber blockade

Abstract

Mechanical allodynia, induced by normally innocuous low-threshold mechanical stimulation, represents a cardinal feature of neuropathic pain. Blockade or ablation of high-threshold, small-diameter unmyelinated group C nerve fibers (C-fibers) has limited effects on mechanical allodynia1,2,3,4. Although large, myelinated group A fibers, in particular Aβ-fibers, have previously been implicated in mechanical allodynia5,6,7, an A-fiber–selective pharmacological blocker is still lacking. Here we report a new method for targeted silencing of A-fibers in neuropathic pain. We found that Toll-like receptor 5 (TLR5) is co-expressed with neurofilament-200 in large-diameter A-fiber neurons in the dorsal root ganglion (DRG). Activation of TLR5 with its ligand flagellin results in neuronal entry of the membrane-impermeable lidocaine derivative QX-314, leading to TLR5-dependent blockade of sodium currents, predominantly in A-fiber neurons of mouse DRGs. Intraplantar co-application of flagellin and QX-314 (flagellin/QX-314) dose-dependently suppresses mechanical allodynia after chemotherapy, nerve injury, and diabetic neuropathy, but this blockade is abrogated in Tlr5-deficient mice. In vivo electrophysiology demonstrated that co-application of flagellin/QX-314 selectively suppressed Aβ-fiber conduction in naive and chemotherapy-treated mice. TLR5-mediated Aβ-fiber blockade, but not capsaicin-mediated C-fiber blockade, also reduced chemotherapy-induced ongoing pain without impairing motor function. Finally, flagellin/QX-314 co-application suppressed sodium currents in large-diameter human DRG neurons. Thus, our findings provide a new tool for targeted silencing of Aβ-fibers and neuropathic pain treatment.

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

Access options

Buy this article

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

Figure 1: TLR5 is co-localized with the A-fiber marker NF200 in DRG neurons, skin nerve fibers, and spinal cord axonal terminals in mice.
Figure 2: Co-application of flagellin and QX-314 blocks sodium currents in large-diameter A-fiber neurons of mouse and human DRGs.
Figure 3: Co-application of flagellin and QX-314 selectively inhibits Aβ-fiber conduction in sciatic nerves of naive and chemotherapy-treated mice.
Figure 4: A-fiber blockade by co-application of flagellin/QX-314 inhibits mechanical allodynia and ongoing pain in different neuropathic pain conditions.

Similar content being viewed by others

References

  1. Ossipov, M.H., Bian, D., Malan, T.P. Jr., Lai, J. & Porreca, F. Lack of involvement of capsaicin-sensitive primary afferents in nerve-ligation injury induced tactile allodynia in rats. Pain 79, 127–133 (1999).

    Article  CAS  PubMed  Google Scholar 

  2. Minett, M.S. et al. Pain without nociceptors? Nav1.7-independent pain mechanisms. Cell Rep. 6, 301–312 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Liu, C.N. et al. Tactile allodynia in the absence of C-fiber activation: altered firing properties of DRG neurons following spinal nerve injury. Pain 85, 503–521 (2000).

    Article  CAS  PubMed  Google Scholar 

  4. Brenneis, C. et al. Phenotyping the function of TRPV1-expressing sensory neurons by targeted axonal silencing. J. Neurosci. 33, 315–326 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Koltzenburg, M., Lundberg, L.E. & Torebjork, H.E. Dynamic and static components of mechanical hyperalgesia in human hairy skin. Pain 51, 207–219 (1992).

    Article  CAS  PubMed  Google Scholar 

  6. Ossipov, M.H. et al. Selective mediation of nerve injury-induced tactile hypersensitivity by neuropeptide Y. J. Neurosci. 22, 9858–9867 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Campbell, J.N., Raja, S.N., Meyer, R.A. & Mackinnon, S.E. Myelinated afferents signal the hyperalgesia associated with nerve injury. Pain 32, 89–94 (1988).

    Article  CAS  PubMed  Google Scholar 

  8. Hayashi, F. et al. The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature 410, 1099–1103 (2001).

    Article  CAS  PubMed  Google Scholar 

  9. Akira, S., Uematsu, S. & Takeuchi, O. Pathogen recognition and innate immunity. Cell 124, 783–801 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Liu, T., Gao, Y.J. & Ji, R.R. Emerging role of Toll-like receptors in the control of pain and itch. Neurosci. Bull. 28, 131–144 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Liu, T. et al. TLR3 deficiency impairs spinal cord synaptic transmission, central sensitization, and pruritus in mice. J. Clin. Invest. 122, 2195–2207 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lawson, S.N. & Waddell, P.J. Soma neurofilament immunoreactivity is related to cell size and fibre conduction velocity in rat primary sensory neurons. J. Physiol. (Lond.) 435, 41–63 (1991).

    Article  CAS  Google Scholar 

  13. McCoy, E.S. et al. Peptidergic CGRPα primary sensory neurons encode heat and itch and tonically suppress sensitivity to cold. Neuron 78, 138–151 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Li, L. et al. The functional organization of cutaneous low-threshold mechanosensory neurons. Cell 147, 1615–1627 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Todd, A.J. Neuronal circuitry for pain processing in the dorsal horn. Nat. Rev. Neurosci. 11, 823–836 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Binshtok, A.M., Bean, B.P. & Woolf, C.J. Inhibition of nociceptors by TRPV1-mediated entry of impermeant sodium channel blockers. Nature 449, 607–610 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. Roberson, D.P. et al. Activity-dependent silencing reveals functionally distinct itch-generating sensory neurons. Nat. Neurosci. 16, 910–918 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Park, C.K. et al. Extracellular microRNAs activate nociceptor neurons to elicit pain via TLR7 and TRPA1. Neuron 82, 47–54 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Smith, K.D. et al. Toll-like receptor 5 recognizes a conserved site on flagellin required for protofilament formation and bacterial motility. Nat. Immunol. 4, 1247–1253 (2003).

    Article  CAS  PubMed  Google Scholar 

  20. Liu, X.J. et al. Nociceptive neurons regulate innate and adaptive immunity and neuropathic pain through MyD88 adapter. Cell Res. 24, 1374–1377 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Cavaletti, G. & Marmiroli, P. Chemotherapy-induced peripheral neurotoxicity. Nat. Rev. Neurol. 6, 657–666 (2010).

    Article  PubMed  Google Scholar 

  22. Binshtok, A.M. et al. Nociceptors are interleukin-1β sensors. J. Neurosci. 28, 14062–14073 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. King, T. et al. Contribution of afferent pathways to nerve injury-induced spontaneous pain and evoked hypersensitivity. Pain 152, 1997–2005 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Xu, Z.Z. et al. Neuroprotectin/protectin D1 protects neuropathic pain in mice after nerve trauma. Ann. Neurol. 74, 490–495 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Khan, G.M., Chen, S.R. & Pan, H.L. Role of primary afferent nerves in allodynia caused by diabetic neuropathy in rats. Neuroscience 114, 291–299 (2002).

    Article  CAS  PubMed  Google Scholar 

  26. Bennett, G.J. & Xie, Y.K. A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 33, 87–107 (1988).

    Article  CAS  PubMed  Google Scholar 

  27. Melzack, R. & Wall, P.D. Pain mechanisms: a new theory. Science 150, 971–979 (1965).

    Article  CAS  PubMed  Google Scholar 

  28. Usoskin, D. et al. Unbiased classification of sensory neuron types by large-scale single-cell RNA sequencing. Nat. Neurosci. 18, 145–153 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Devor, M. Neuropathic pain and injured nerve: peripheral mechanisms. Br. Med. Bull. 47, 619–630 (1991).

    Article  CAS  PubMed  Google Scholar 

  30. Ma, C. et al. Similar electrophysiological changes in axotomized and neighboring intact dorsal root ganglion neurons. J. Neurophysiol. 89, 1588–1602 (2003).

    Article  PubMed  Google Scholar 

  31. Haroutounian, S. et al. Primary afferent input critical for maintaining spontaneous pain in peripheral neuropathy. Pain 155, 1272–1279 (2014).

    Article  PubMed  Google Scholar 

  32. Kim, Y.H. et al. TRPV1 in GABAergic interneurons mediates neuropathic mechanical allodynia and disinhibition of the nociceptive circuitry in the spinal cord. Neuron 74, 640–647 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Seal, R.P. et al. Injury-induced mechanical hypersensitivity requires C-low threshold mechanoreceptors. Nature 462, 651–655 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Boada, M.D. et al. Fast-conducting mechanoreceptors contribute to withdrawal behavior in normal and nerve injured rats. Pain 155, 2646–2655 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Smith, A.K., O'Hara, C.L. & Stucky, C.L. Mechanical sensitization of cutaneous sensory fibers in the spared nerve injury mouse model. Mol. Pain 9, 61 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Lu, Y. et al. A feed-forward spinal cord glycinergic neural circuit gates mechanical allodynia. J. Clin. Invest. 123, 4050–4062 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Duan, B. et al. Identification of spinal circuits transmitting and gating mechanical pain. Cell 159, 1417–1432 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Vijay-Kumar, M. et al. Flagellin treatment protects against chemicals, bacteria, viruses, and radiation. J. Immunol. 180, 8280–8285 (2008).

    Article  CAS  PubMed  Google Scholar 

  39. Sfondrini, L. et al. Antitumor activity of the TLR-5 ligand flagellin in mouse models of cancer. J. Immunol. 176, 6624–6630 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Alessandri-Haber, N., Dina, O.A., Joseph, E.K., Reichling, D.B. & Levine, J.D. Interaction of transient receptor potential vanilloid 4, integrin, and SRC tyrosine kinase in mechanical hyperalgesia. J. Neurosci. 28, 1046–1057 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Berta, T. et al. Extracellular caspase-6 drives murine inflammatory pain via microglial TNF-α secretion. J. Clin. Invest. 124, 1173–1186 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Pagadala, P. et al. Loss of NR1 subunit of NMDARs in primary sensory neurons leads to hyperexcitability and pain hypersensitivity: involvement of Ca2+-activated small conductance potassium channels. J. Neurosci. 33, 13425–13430 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Liu, T., Xu, Z.Z., Park, C.K., Berta, T. & Ji, R.R. Toll-like receptor 7 mediates pruritus. Nat. Neurosci. 13, 1460–1462 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Xie, R.G. et al. Blockade of persistent sodium currents contributes to the riluzole-induced inhibition of spontaneous activity and oscillations in injured DRG neurons. PLoS ONE 6, e18681 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Lee, J.H. et al. A monoclonal antibody that targets a NaV1.7 channel voltage sensor for pain and itch relief. Cell 157, 1393–1404 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Baumann, T.K., Chaudhary, P. & Martenson, M.E. Background potassium channel block and TRPV1 activation contribute to proton depolarization of sensory neurons from humans with neuropathic pain. Eur. J. Neurosci. 19, 1343–1351 (2004).

    Article  PubMed  Google Scholar 

  47. Davidson, S. et al. Human sensory neurons: Membrane properties and sensitization by inflammatory mediators. Pain 155, 1861–1870 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Pinto, V., Derkach, V.A. & Safronov, B.V. Role of TTX-sensitive and TTX-resistant sodium channels in Adelta- and C-fiber conduction and synaptic transmission. J. Neurophysiol. 99, 617–628 (2008).

    Article  CAS  PubMed  Google Scholar 

  49. Tsuchimochi, H., McCord, J.L., Leal, A.K. & Kaufman, M.P. Dorsal root tetrodotoxin-resistant sodium channels do not contribute to the augmented exercise pressor reflex in rats with chronic femoral artery occlusion. Am. J. Physiol. Heart Circ. Physiol. 300, H652–H663 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  51. Hargreaves, K., Dubner, R., Brown, F., Flores, C. & Joris, J. A new and sensitive method for measuring thermal nociception in cutaneous hyperalgesia. Pain 32, 77–88 (1988).

    Article  CAS  PubMed  Google Scholar 

  52. King, T. et al. Unmasking the tonic-aversive state in neuropathic pain. Nat. Neurosci. 12, 1364–1366 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Park, C.K. et al. Resolving TRPV1- and TNF-α-mediated spinal cord synaptic plasticity and inflammatory pain with neuroprotectin D1. J. Neurosci. 31, 15072–15085 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study is supported by US National Institutes of Health (NIH) R01 grants NS67686 (R.-R.J.), NS87988 (R.-R.J.), NS89479 (R.-R.J.), DE17794 (R.-R.J.), and DE22743 (R.-R.J.); NIH R21 grants NS82985 (Z.-Z.X.) and NS91779 (Z.-Z.X.); NIH R01 grant DE19440 (F.W.); a Korea government grant 2012R1A3A2048834 (S.B.O.); and a Korea National Research Foundation grant 2013R1A6A3A04065858 (Y.H.K.).

Author information

Authors and Affiliations

Authors

Contributions

Z.-Z.X. developed the behavioral part of the project and designed and performed immunohistochemical and behavioral experiments. Y.H.K. recorded action potentials in mouse DRG neurons and compound potentials in intact mice, and recorded sodium currents in human DRG neurons. S.B. initially tested the idea of flagellin/QX-314 blockade of sodium currents in mouse DRG neurons and examined dye (phallodin-rhodamine) uptake in DRGs. Y.Z. performed in situ hybridization under the guidance of F.W. T.B. performed the PCR experiments. R.-R.J. conceived and supervised the project. S.B.O. and F.W. discussed the project; and R.-R.J., Z.-Z.X., S.B.O., and F.W. wrote the paper.

Corresponding author

Correspondence to Ru-Rong Ji.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Table 1 and Supplementary Figures 1–12 (PDF 5037 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, ZZ., Kim, Y., Bang, S. et al. Inhibition of mechanical allodynia in neuropathic pain by TLR5-mediated A-fiber blockade. Nat Med 21, 1326–1331 (2015). https://doi.org/10.1038/nm.3978

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3978

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