DOI QR코드

DOI QR Code

Investigation of serum biomarkers for neuropathic pain in neuromyelitis optica spectrum disorder: a preliminary study

  • Hyun, Jae-Won (Department of Neurology, Division of Clinical Research, National Cancer Center) ;
  • Kim, Yeseul (Research Institute and Hospital of National Cancer Center) ;
  • Kim, Ho Jin (Department of Neurology, Division of Clinical Research, National Cancer Center)
  • Received : 2020.10.27
  • Accepted : 2021.02.25
  • Published : 2021.04.30

Abstract

Background: We aimed to investigate candidates for serological biomarkers of neuropathic pain in individuals with neuromyelitis optica spectrum disorder (NMOSD). Methods: We analyzed 38 sera samples from 38 participants with NMOSD in National Cancer Center. Neuropathic pain was evaluated using the painDETECT questionnaire. Pain with neuropathic components (painDETECT score ≥ 13) was observed in 22 participants, among whom 17 had definite neuropathic pain (painDETECT score ≥ 19). The remaining 16 participants had non-neuropathic pain (painDETECT score < 13). Serum glial fibrillary acidic protein (GFAP) levels were assessed using a single-molecule array assay. Several cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, IL-10, and IL-17A, were measured by a multiplex bead-based immunoassay. Results: In comparison of NMOSD participants with neuropathic pain components (or definite neuropathic pain) and those with non-neuropathic pain, the absolute values of serum GFAP, TNF-α, IL-6, and IL-10 levels were higher in participants with neuropathic pain components (or definite neuropathic pain), but these findings did not reach statistical significance. Conclusions: Further larger-scale investigations to find reliable serological biomarkers for neuropathic pain in NMOSD are warranted.

Keywords

Acknowledgement

This research was supported by a grant funded by the Korean Society of Pain and Autonomic Disorders.

References

  1. International Association for the Study of Pain (IASP). IASP terminology [Internet]. Washington, D.C. (USA): IASP; c2017 [accessed 2019 Apr 1]. Available from: www.iasp-pain.org/terminology.
  2. Bradl M, Kanamori Y, Nakashima I, Misu T, Fujihara K, Lassmann H, et al. Pain in neuromyelitis optica--prevalence, pathogenesis and therapy. Nat Rev Neurol 2014;10:529-536. https://doi.org/10.1038/nrneurol.2014.129
  3. Hyun JW, Jang H, Yu J, Park NY, Kim SH, Huh SY, et al. Comparison of neuropathic pain in neuromyelitis optica spectrum disorder and multiple sclerosis. J Clin Neurol 2020;16:124-130. https://doi.org/10.3988/jcn.2020.16.1.124
  4. Watanabe M, Nakamura Y, Michalak Z, Isobe N, Barro C, Leppert D, et al. Serum GFAP and neurofilament light as biomarkers of disease activity and disability in NMOSD. Neurology 2019;93:e1299-e1311. https://doi.org/10.1212/WNL.0000000000008160
  5. Uzawa A, Mori M, Arai K, Sato Y, Hayakawa S, Masuda S, et al. Cytokine and chemokine profiles in neuromyelitis optica: significance of interleukin-6. Mult Scler 2010;16:1443-1452. https://doi.org/10.1177/1352458510379247
  6. Sung JK, Choi JH, Jeong J, Kim WJ, Lee DJ, Lee SC, et al. Korean version of the painDETECT questionnaire: a study for cultural adaptation and validation. Pain Pract 2017;17:494-504. https://doi.org/10.1111/papr.12472
  7. Wingerchuk DM, Banwell B, Bennett JL, Cabre P, Carroll W, Chitnis T, et al. International consensus diagnostic criteria for neuromyelitis optica spectrum disorders. Neurology 2015;85:177-189. https://doi.org/10.1212/WNL.0000000000001729
  8. Kim Y, Kim G, Kong BS, Lee JE, Oh YM, Hyun JW, et al. Large-scale in-house cell-based assay for evaluating the serostatus in patients with neuromyelitis optica spectrum disorder based on new diagnostic criteria. J Clin Neurol 2017;13:175-180. https://doi.org/10.3988/jcn.2017.13.2.175
  9. Freynhagen R, Tolle TR, Gockel U, Baron R. The painDETECT project-far more than a screening tool on neuropathic pain. Curr Med Res Opin 2016;32:1033-1057. https://doi.org/10.1185/03007995.2016.1157460
  10. Lennon VA, Wingerchuk DM, Kryzer TJ, Pittock SJ, Lucchinetti CF, Fujihara K, et al. A serum autoantibody marker of neuromyelitis optica: distinction from multiple sclerosis. Lancet 2004;364:2106-2112. https://doi.org/10.1016/S0140-6736(04)17551-X
  11. Hung AL, Lim M, Doshi TL. Targeting cytokines for treatment of neuropathic pain. Scand J Pain 2017;17:287-293. https://doi.org/10.1016/j.sjpain.2017.08.002
  12. Wei XH, Zang Y, Wu CY, Xu JT, Xin WJ, Liu XG. Peri-sciatic administration of recombinant rat TNF-alpha induces mechanical allodynia via upregulation of TNF-alpha in dorsal root ganglia and in spinal dorsal horn: the role of NF-kappa B pathway. Exp Neurol 2007;205:471-484. https://doi.org/10.1016/j.expneurol.2007.03.012
  13. Zanella JM, Burright EN, Hildebrand K, Hobot C, Cox M, Christoferson L, et al. Effect of etanercept, a tumor necrosis factor-alpha inhibitor, on neuropathic pain in the rat chronic constriction injury model. Spine (Phila Pa 1976) 2008;33:227-234. https://doi.org/10.1097/BRS.0b013e318162340a
  14. Wang K, Bao JP, Yang S, Hong X, Liu L, Xie XH, et al. A cohort study comparing the serum levels of pro- or anti-inflammatory cytokines in patients with lumbar radicular pain and healthy subjects. Eur Spine J 2016;25:1428-1434. https://doi.org/10.1007/s00586-015-4349-4
  15. Zu B, Pan H, Zhang XJ, Yin ZS. Serum levels of the inflammatory cytokines in patients with lumbar radicular pain due to disc herniation. Asian Spine J 2016;10:843-849. https://doi.org/10.4184/asj.2016.10.5.843
  16. US Food and Drug. Information on Tumor Necrosis Factor (TNF) Blockers (marketed as Remicade, Enbrel, Humira, Cimzia, and Simponi) [Internet]. Washington, D.C., USA: US Food and Drug; c2021 [accessed 2021 Feb 25]. Available from: https://www.fda.gov/drugs/postmarket-drug-safety-information-patients-and-providers/information-tumor-necrosis-factor-tnf-blockers-marketed-remicade-enbrel-humira-cimzia-and-simponi.
  17. Kemanetzoglou E, Andreadou E. CNS demyelination with TNF-α blockers. Curr Neurol Neurosci Rep 2017;17:36. https://doi.org/10.1007/s11910-017-0742-1
  18. Ramer MS, Murphy PG, Richardson PM, Bisby MA. Spinal nerve lesion-induced mechanoallodynia and adrenergic sprouting in sensory ganglia are attenuated in interleukin-6 knockout mice. Pain 1998;78:115-121. https://doi.org/10.1016/S0304-3959(98)00121-3
  19. Guptarak J, Wanchoo S, Durham-Lee J, Wu Y, Zivadinovic D, Paulucci-Holthauzen A, et al. Inhibition of IL-6 signaling: a novel therapeutic approach to treating spinal cord injury pain. Pain 2013;154:1115-1128. https://doi.org/10.1016/j.pain.2013.03.026
  20. Araki M, Matsuoka T, Miyamoto K, Kusunoki S, Okamoto T, Murata M, et al. Efficacy of the anti-IL-6 receptor antibody tocilizumab in neuromyelitis optica: a pilot study. Neurology 2014;82:1302-1306. https://doi.org/10.1212/WNL.0000000000000317
  21. Kim CF, Moalem-Taylor G. Interleukin-17 contributes to neuroinflammation and neuropathic pain following peripheral nerve injury in mice. J Pain 2011;12:370-383. https://doi.org/10.1016/j.jpain.2010.08.003
  22. Day YJ, Liou JT, Lee CM, Lin YC, Mao CC, Chou AH, et al. Lack of interleukin-17 leads to a modulated micro-environment and amelioration of mechanical hypersensitivity after peripheral nerve injury in mice. Pain 2014;155:1293-1302. https://doi.org/10.1016/j.pain.2014.04.004
  23. Shao Q, Li Y, Wang Q, Zhao J. IL-10 and IL-1β mediate neuropathic-pain like behavior in the ventrolateral orbital cortex. Neurochem Res 2015;40:733-739. https://doi.org/10.1007/s11064-015-1521-5