Skip to main content

Advertisement

Log in

Phosphorylated CRMP2 Regulates Spinal Nociceptive Neurotransmission

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

The collapsin response mediator protein 2 (CRMP2) has emerged as a central node in assembling nociceptive signaling complexes involving voltage-gated ion channels. Concerted actions of post-translational modifications, phosphorylation and SUMOylation, of CRMP2 contribute to regulation of pathological pain states. In the present study, we demonstrate a novel role for CRMP2 in spinal nociceptive transmission. We found that, of six possible post-translational modifications, three phosphorylation sites on CRMP2 were critical for regulating calcium influx in dorsal root ganglion sensory neurons. Of these, only CRMP2 phosphorylated at serine 522 by cyclin-dependent kinase 5 (Cdk5) contributed to spinal neurotransmission in a bidirectional manner. Accordingly, expression of a non-phosphorylatable CRMP2 (S522A) decreased the frequency of spontaneous excitatory postsynaptic currents (sEPSCs), whereas expression of a constitutively phosphorylated CRMP2 (S522D) increased the frequency of sEPSCs. The presynaptic nature of CRMP2’s actions was further confirmed by pharmacological antagonism of Cdk5-mediated CRMP2 phosphorylation with S-N-benzy-2-acetamido-3-methoxypropionamide ((S)-lacosamide; (S)-LCM) which (i) decreased sEPSC frequency, (ii) increased paired-pulse ratio, and (iii) reduced the presynaptic distribution of CaV2.2 and NaV1.7, two voltage-gated ion channels implicated in nociceptive signaling. (S)-LCM also inhibited depolarization-evoked release of the pro-nociceptive neurotransmitter calcitonin gene-related peptide (CGRP) in the spinal cord. Increased CRMP2 phosphorylation in rats with spared nerve injury (SNI) was decreased by intrathecal administration of (S)-LCM resulting in a loss of presynaptic localization of CaV2.2 and NaV1.7. Together, these findings indicate that CRMP2 regulates presynaptic excitatory neurotransmission in spinal cord and may play an important role in regulating pathological pain. Novel targeting strategies to inhibit CRMP2 phosphorylation by Cdk5 may have great potential for the treatment of chronic pain.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. Costigan M, Scholz J, Woolf CJ (2009) Neuropathic pain: a maladaptive response of the nervous system to damage. Annu Rev Neurosci 32:1–32.https://doi.org/10.1146/annurev.neuro.051508.135531

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Moulin D, Boulanger A, Clark AJ, Clarke H, Dao T, Finley GA, Furlan A, Gilron I et al (2014) Pharmacological management of chronic neuropathic pain: revised consensus statement from the Canadian Pain Society. Pain Res Manag 19(6):328–335

    Article  PubMed  PubMed Central  Google Scholar 

  3. Price TJ, Basbaum AI, Bresnahan J, Chambers JF, De Koninck Y, Edwards RR, Ji RR, Katz J et al (2018) Transition to chronic pain: opportunities for novel therapeutics. Nat Rev Neurosci 19:383–384. https://doi.org/10.1038/s41583-018-0012-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Todd AJ (2010) Neuronal circuitry for pain processing in the dorsal horn. Nat Rev Neurosci 11(12):823–836. https://doi.org/10.1038/nrn2947

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. D'Mello R, Dickenson AH (2008) Spinal cord mechanisms of pain. Br J Anaesth 101(1):8–16.https://doi.org/10.1093/bja/aen088

    Article  CAS  PubMed  Google Scholar 

  6. Latremoliere A, Woolf CJ (2009) Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J Pain 10(9):895–926. https://doi.org/10.1016/j.jpain.2009.06.012

    Article  PubMed  PubMed Central  Google Scholar 

  7. Divito CB, Underhill SM (2014) Excitatory amino acid transporters: roles in glutamatergic neurotransmission. Neurochem Int 73:172–180. https://doi.org/10.1016/j.neuint.2013.12.008

    Article  CAS  PubMed  Google Scholar 

  8. Dolphin AC (2012) Calcium channel auxiliary alpha2delta and beta subunits: trafficking and one step beyond. Nat Rev Neurosci 13(8):542–555. https://doi.org/10.1038/nrn3311

    Article  CAS  PubMed  Google Scholar 

  9. Khanna R, Zougman A, Stanley EF (2007) A proteomic screen for presynaptic terminal N-type calcium channel (CaV2.2) binding partners. J Biochem Mol Biol 40(3):302–314

    CAS  PubMed  Google Scholar 

  10. Chi XX, Schmutzler BS, Brittain JM, Hingtgen CM, Nicol GD, Khanna R (2009) Regulation of N-type voltage-gated calcium (CaV2.2) channels and transmitter release by collapsin response mediator protein-2 (CRMP-2) in sensory neurons. J Cell Sci 122(23):4351–4362

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Brittain JM, Piekarz AD, Wang Y, Kondo T, Cummins TR, Khanna R (2009) An atypical role for collapsin response mediator protein 2 (CRMP-2) in neurotransmitter release via interaction with presynaptic voltage-gated calcium channels. J Biol Chem 284(45):31375–31390. https://doi.org/10.1074/jbc.M109.009951

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Brittain JM, Duarte DB, Wilson SM, Zhu W, Ballard C, Johnson PL, Liu N, Xiong W et al (2011) Suppression of inflammatory and neuropathic pain by uncoupling CRMP-2 from the presynaptic Ca(2)(+) channel complex. Nat Med 17(7):822–829. https://doi.org/10.1038/nm.2345

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Francois-Moutal L, Wang Y, Moutal A, Cottier KE, Melemedjian OK, Yang X, Wang Y, Ju W et al (2015) A membrane-delimited N-myristoylated CRMP2 peptide aptamer inhibits CaV2.2 trafficking and reverses inflammatory and postoperative pain behaviors. Pain 156(7):1247–1264. https://doi.org/10.1097/j.pain.0000000000000147

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Moutal A, Li W, Wang Y, Ju W, Luo S, Cai S, Francois-Moutal L, Perez-Miller S et al (2017) Homology-guided mutational analysis reveals the functional requirements for antinociceptive specificity of collapsin response mediator protein 2-derived peptides. Br J Pharmacol 175:2244–2260. https://doi.org/10.1111/bph.13737

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Piekarz AD, Due MR, Khanna M, Wang B, Ripsch MS, Wang R, Meroueh SO, Vasko MR et al (2012) CRMP-2 peptide mediated decrease of high and low voltage-activated calcium channels, attenuation of nociceptor excitability, and anti-nociception in a model of AIDS therapy-induced painful peripheral neuropathy. Mol Pain 8(1):54. https://doi.org/10.1186/1744-8069-8-54

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ripsch MS, Ballard CJ, Khanna M, Hurley JH, White FA, Khanna R (2012) A peptide uncoupling CRMP-2 from the presynaptic Ca2+ channel complex demonstrate efficacy in animal models of migraine and AIDS therapy-induced neuropathy. Transl Neurosci 3(1):1–8

    Article  PubMed  Google Scholar 

  17. Khanna R, Wilson SM, Brittain JM, Weimer J, Sultana R, Butterfield A, Hensley K (2012) Opening Pandora’s jar: a primer on the putative roles of CRMP2 in a panoply of neurodegenerative, sensory and motor neuron, and central disorders. Future Neurol 7(6):749–771. https://doi.org/10.2217/fnl.12.68

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Quach TT, Honnorat J, Kolattukudy PE, Khanna R, Duchemin AM (2015) CRMPs: critical molecules for neurite morphogenesis and neuropsychiatric diseases. Mol Psychiatry 20(9):1037–1045. https://doi.org/10.1038/mp.2015.77

    Article  CAS  PubMed  Google Scholar 

  19. Moutal A, Francois-Moutal L, Brittain JM, Khanna M, Khanna R (2014) Differential neuroprotective potential of CRMP2 peptide aptamers conjugated to cationic, hydrophobic, and amphipathic cell penetrating peptides. Front Cell Neurosci 8:471. https://doi.org/10.3389/fncel.2014.00471

    Article  PubMed  Google Scholar 

  20. Dustrude ET, Wilson SM, Ju W, Xiao Y, Khanna R (2013) CRMP2 protein SUMOylation modulates NaV1.7 channel trafficking. J Biol Chem 288(34):24316–24331. https://doi.org/10.1074/jbc.M113.474924

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Brustovetsky T, Pellman JJ, Yang XF, Khanna R, Brustovetsky N (2014) Collapsin response mediator protein 2 (CRMP2) interacts with N-methyl-D-aspartate (NMDA) receptor and Na+/Ca2+ exchanger and regulates their functional activity. J Biol Chem 289(11):7470–7482. https://doi.org/10.1074/jbc.M113.518472

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Dustrude ET, Moutal A, Yang X, Wang Y, Khanna M, Khanna R (2016) Hierarchical CRMP2 posttranslational modifications control NaV1.7 function. Proc Natl Acad Sci 113(52):E8443–E8452. https://doi.org/10.1073/pnas.1610531113

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Dustrude ET, Perez-Miller S, Francois-Moutal L, Moutal A, Khanna M, Khanna R (2017) A single structurally conserved SUMOylation site in CRMP2 controls NaV1.7 function. Channels (Austin) 11:316–328. https://doi.org/10.1080/19336950.2017.1299838

    Article  Google Scholar 

  24. Francois-Moutal L, Dustrude ET, Wang Y, Brustovetsky T, Dorame A, Ju W, Moutal A, Perez-Miller S et al (2018) Inhibition of the Ubc9 E2 SUMO-conjugating enzyme-CRMP2 interaction decreases NaV1.7 currents and reverses experimental neuropathic pain. Pain. https://doi.org/10.1097/j.pain.0000000000001294

    Article  PubMed  PubMed Central  Google Scholar 

  25. Ju W, Li Q, Wilson SM, Brittain JM, Meroueh L, Khanna R (2013) SUMOylation alters CRMP2 regulation of calcium influx in sensory neurons. Channels 7(3):153–159

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Moutal A, Dustrude ET, Largent-Milnes TM, Vanderah TW, Khanna M, Khanna R (2017) Blocking CRMP2 SUMOylation reverses neuropathic pain. Mol Psychiatry. https://doi.org/10.1038/mp.2017.117

    Article  Google Scholar 

  27. Cole AR, Causeret F, Yadirgi G, Hastie CJ, McLauchlan H, McManus EJ, Hernandez F, Eickholt BJ et al (2006) Distinct priming kinases contribute to differential regulation of collapsin response mediator proteins by glycogen synthase kinase-3 in vivo. J Biol Chem 281(24):16591–16598. https://doi.org/10.1074/jbc.M513344200

    Article  CAS  PubMed  Google Scholar 

  28. Yoshimura T, Kawano Y, Arimura N, Kawabata S, Kikuchi A, Kaibuchi K (2005) GSK-3beta regulates phosphorylation of CRMP-2 and neuronal polarity. Cell 120(1):137–149. https://doi.org/10.1016/j.cell.2004.11.012

    Article  CAS  PubMed  Google Scholar 

  29. Arimura N, Inagaki N, Chihara K, Menager C, Nakamura N, Amano M, Iwamatsu A, Goshima Y et al (2000) Phosphorylation of collapsin response mediator protein-2 by Rho-kinase. Evidence for two separate signaling pathways for growth cone collapse. J Biol Chem 275(31):23973–23980. https://doi.org/10.1074/jbc.M001032200

    Article  CAS  PubMed  Google Scholar 

  30. Varrin-Doyer M, Vincent P, Cavagna S, Auvergnon N, Noraz N, Rogemond V, Honnorat J, Moradi-Ameli M et al (2009) Phosphorylation of collapsin response mediator protein 2 on Tyr-479 regulates CXCL12-induced T lymphocyte migration. J Biol Chem 284(19):13265–13276. https://doi.org/10.1074/jbc.M807664200

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Uchida Y, Ohshima T, Yamashita N, Ogawara M, Sasaki Y, Nakamura F, Goshima Y (2009) Semaphorin3A signaling mediated by Fyn-dependent tyrosine phosphorylation of collapsin response mediator protein 2 at tyrosine 32. J Biol Chem 284(40):27393–27401. https://doi.org/10.1074/jbc.M109.000240

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ji RR, Kawasaki Y, Zhuang ZY, Wen YR, Zhang YQ (2007) Protein kinases as potential targets for the treatment of pathological pain. Handb Exp Pharmacol 177:359–389

    Article  CAS  Google Scholar 

  33. Uchida Y, Ohshima T, Sasaki Y, Suzuki H, Yanai S, Yamashita N, Nakamura F, Takei K et al (2005) Semaphorin3A signalling is mediated via sequential Cdk5 and GSK3beta phosphorylation of CRMP2: implication of common phosphorylating mechanism underlying axon guidance and Alzheimer’s disease. Genes Cells 10(2):165–179. https://doi.org/10.1111/j.1365-2443.2005.00827.x

    Article  CAS  PubMed  Google Scholar 

  34. Brittain JM, Wang Y, Eruvwetere O, Khanna R (2012) Cdk5-mediated phosphorylation of CRMP-2 enhances its interaction with CaV2.2. FEBS Lett 586(21):3813–3818. https://doi.org/10.1016/j.febslet.2012.09.022

    Article  CAS  PubMed  Google Scholar 

  35. Moutal A, Cai S, Luo S, Voisin R, Khanna R (2018) CRMP2 is necessary for neurofibromatosis type 1 related pain. Channels 12(1):47–50. https://doi.org/10.1080/19336950.2017.1370524

    Article  PubMed  Google Scholar 

  36. Moutal A, Luo S, Largent-Milnes TM, Vanderah TW, Khanna R (2018) Cdk5-mediated CRMP2 phosphorylation is necessary and sufficient for peripheral neuropathic pain. Neurobiol Pain. https://doi.org/10.1016/j.ynpai.2018.07.003

    Article  PubMed  Google Scholar 

  37. Hergenroeder GW, Redell JB, Choi HA, Schmitt L, Donovan W, Francisco GE, Schmitt K, Moore AN et al (2018) Increased levels of circulating glial fibrillary acidic protein and collapsin response mediator protein-2 autoantibodies in the acute stage of spinal cord injury predict the subsequent development of neuropathic pain. J Neurotrauma 35:2530–2539. https://doi.org/10.1089/neu.2018.5675

    Article  PubMed  PubMed Central  Google Scholar 

  38. Moutal A, Yang X, Li W, Gilbraith KB, Luo S, Cai S, Francois-Moutal L, Chew LA et al (2017) CRISPR/Cas9 editing of Nf1 gene identifies CRMP2 as a therapeutic target in neurofibromatosis type 1-related pain that is reversed by (S)-lacosamide. Pain 158(12):2301–2319. https://doi.org/10.1097/j.pain.0000000000001002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Moutal A, Chew LA, Yang X, Wang Y, Yeon SK, Telemi E, Meroueh S, Park KD et al (2016) (S)-Lacosamide inhibition of CRMP2 phosphorylation reduces postoperative and neuropathic pain behaviors through distinct classes of sensory neurons identified by constellation pharmacology. Pain 157(7):1448–1463. https://doi.org/10.1097/j.pain.0000000000000555

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhang SH, Yu J, Lou GD, Tang YY, Wang RR, Hou WW, Chen Z (2016) Widespread pain sensitization after partial infraorbital nerve transection in MRL/MPJ mice. Pain 157(3):740–749. https://doi.org/10.1097/j.pain.0000000000000432

    Article  PubMed  Google Scholar 

  41. Duan B, Cheng L, Bourane S, Britz O, Padilla C, Garcia-Campmany L, Krashes M, Knowlton W et al (2014) Identification of spinal circuits transmitting and gating mechanical pain. Cell 159(6):1417–1432. https://doi.org/10.1016/j.cell.2014.11.003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Moutal A, Sun L, Yang X, Li W, Cai S, Luo S, Khanna R (2018) CRMP2-neurofibromin interface drives NF1-related pain. Neuroscience 381:79–90. https://doi.org/10.1016/j.neuroscience.2018.04.002

    Article  CAS  PubMed  Google Scholar 

  43. Moutal A, Wang Y, Yang X, Ji Y, Luo S, Dorame A, Bellampalli SS, Chew LA et al (2017) Dissecting the role of the CRMP2-neurofibromin complex on pain behaviors. Pain 158(11):2203–2221. https://doi.org/10.1097/j.pain.0000000000001026

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Xie JY, Chew LA, Yang X, Wang Y, Qu C, Wang Y, Federici LM, Fitz SD et al (2016) Sustained relief of ongoing experimental neuropathic pain by a CRMP2 peptide aptamer with low abuse potential. Pain 157(9):2124–2140. https://doi.org/10.1097/j.pain.0000000000000628

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Moutal A, Eyde N, Telemi E, Park KD, Xie JY, Dodick DW, Porreca F, Khanna R (2016) Efficacy of (S)-lacosamide in preclinical models of cephalic pain. Pain Rep 1(1):e565. https://doi.org/10.1097/PR9.0000000000000565

    Article  PubMed  PubMed Central  Google Scholar 

  46. Moutal A, Francois-Moutal L, Perez-Miller S, Cottier K, Chew LA, Yeon SK, Dai J, Park KD et al (2016) (S)-Lacosamide binding to collapsin response mediator protein 2 (CRMP2) regulates CaV2.2 activity by subverting its phosphorylation by Cdk5. Mol Neurobiol 53(3):1959–1976. https://doi.org/10.1007/s12035-015-9141-2

    Article  CAS  PubMed  Google Scholar 

  47. Ibrahim MM, Patwardhan A, Gilbraith KB, Moutal A, Yang X, Chew LA, Largent-Milnes T, Malan TP et al (2017) Long-lasting antinociceptive effects of green light in acute and chronic pain in rats. Pain 158(2):347–360. https://doi.org/10.1097/j.pain.0000000000000767

    Article  PubMed  PubMed Central  Google Scholar 

  48. Chi XX, Schmutzler BS, Brittain JM, Wang Y, Hingtgen CM, Nicol GD, Khanna R (2009) Regulation of N-type voltage-gated calcium channels (Cav2.2) and transmitter release by collapsin response mediator protein-2 (CRMP-2) in sensory neurons. J Cell Sci 122(Pt 23):4351–4362. https://doi.org/10.1242/jcs.053280

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Pacchioni AM, Vallone J, Worley PF, Kalivas PW (2009) Neuronal pentraxins modulate cocaine-induced neuroadaptations. J Pharmacol Exp Ther 328(1):183–192. https://doi.org/10.1124/jpet.108.143115

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Duan JH, Hodgdon KE, Hingtgen CM, Nicol GD (2014) N-type calcium current, Cav2.2, is enhanced in small-diameter sensory neurons isolated from Nf1+/− mice. Neuroscience 270:192–202. https://doi.org/10.1016/j.neuroscience.2014.04.021

    Article  CAS  PubMed  Google Scholar 

  51. Wilson SM, Toth PT, Oh SB, Gillard SE, Volsen S, Ren D, Philipson LH, Lee EC et al (2000) The status of voltage-dependent calcium channels in alpha 1E knock-out mice. JNeurosci 20(23):8566–8571

    Article  CAS  Google Scholar 

  52. Wheeler DG, Groth RD, Ma H, Barrett CF, Owen SF, Safa P, Tsien RW (2012) Ca(V)1 and Ca(V)2 channels engage distinct modes of Ca(2+) signaling to control CREB-dependent gene expression. Cell 149(5):1112–1124

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. McIntosh JM, Corpuz GO, Layer RT, Garrett JE, Wagstaff JD, Bulaj G, Vyazovkina A, Yoshikami D et al (2000) Isolation and characterization of a novel conus peptide with apparent antinociceptive activity. J Biol Chem 275(42):32391–32397. https://doi.org/10.1074/jbc.M003619200

    Article  CAS  PubMed  Google Scholar 

  54. Wang Y, Brittain JM, Jarecki BW, Park KD, Wilson SM, Wang B, Hale R, Meroueh SO et al (2010) In silico docking and electrophysiological characterization of lacosamide binding sites on collapsin response mediator protein-2 identifies a pocket important in modulating sodium channel slow inactivation. J Biol Chem 285(33):25296–25307. https://doi.org/10.1074/jbc.M110.128801

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zucker RS, Regehr WG (2002) Short-term synaptic plasticity. Annu Rev Physiol 64:355–405. https://doi.org/10.1146/annurev.physiol.64.092501.114547

    Article  CAS  PubMed  Google Scholar 

  56. Zhang Y, Xiao X, Zhang XM, Zhao ZQ, Zhang YQ (2012) Estrogen facilitates spinal cord synaptic transmission via membrane-bound estrogen receptors: implications for pain hypersensitivity. J Biol Chem 287(40):33268–33281. https://doi.org/10.1074/jbc.M112.368142

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Schou WS, Ashina S, Amin FM, Goadsby PJ, Ashina M (2017) Calcitonin gene-related peptide and pain: a systematic review. J Headache Pain 18(1):34. https://doi.org/10.1186/s10194-017-0741-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Nitzan-Luques A, Devor M, Tal M (2011) Genotype-selective phenotypic switch in primary afferent neurons contributes to neuropathic pain. Pain 152(10):2413–2426. https://doi.org/10.1016/j.pain.2011.07.012

    Article  CAS  PubMed  Google Scholar 

  59. Lee SE, Kim JH (2007) Involvement of substance P and calcitonin gene-related peptide in development and maintenance of neuropathic pain from spinal nerve injury model of rat. Neurosci Res 58(3):245–249. https://doi.org/10.1016/j.neures.2007.03.004

    Article  CAS  PubMed  Google Scholar 

  60. Decosterd I, Woolf CJ (2000) Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain 87(2):149–158

    Article  CAS  PubMed  Google Scholar 

  61. Cizkova D, Marsala J, Lukacova N, Marsala M, Jergova S, Orendacova J, Yaksh TL (2002) Localization of N-type Ca2+ channels in the rat spinal cord following chronic constrictive nerve injury. Exp Brain Res 147(4):456–463. https://doi.org/10.1007/s00221-002-1217-3

    Article  CAS  PubMed  Google Scholar 

  62. Ip JP, Fu AK, Ip NY (2014) CRMP2: functional roles in neural development and therapeutic potential in neurological diseases. Neuroscientist 20:589–598. https://doi.org/10.1177/1073858413514278

    Article  CAS  PubMed  Google Scholar 

  63. Chew LA, Khanna R (2018) CRMP2 and voltage-gated ion channels: potential roles in neuropathic pain. Neuronal Signaling 2:16. https://doi.org/10.1042/NS20170220

    Article  Google Scholar 

  64. Moutal A, Eyde N, Telemi E, Park KD, Xie JY, Dodick DW, Porreca F, Khanna R (2016) Efficacy of (S)-lacosamide in preclinical models of cephalic pain. Pain Rep 1(1):1–10. https://doi.org/10.1097/PR9.0000000000000565

    Article  Google Scholar 

  65. Marques JM, Rodrigues RJ, Valbuena S, Rozas JL, Selak S, Marin P, Aller MI, Lerma J (2013) CRMP2 tethers kainate receptor activity to cytoskeleton dynamics during neuronal maturation. J Neurosci 33(46):18298–18310. https://doi.org/10.1523/JNEUROSCI.3136-13.2013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Petratos S, Ozturk E, Azari MF, Kenny R, Lee JY, Magee KA, Harvey AR, McDonald C et al (2012) Limiting multiple sclerosis related axonopathy by blocking Nogo receptor and CRMP-2 phosphorylation. Brain 135(Pt 6):1794–1818. https://doi.org/10.1093/brain/aws100

    Article  PubMed  PubMed Central  Google Scholar 

  67. Zhou Y, Danbolt NC (2014) Glutamate as a neurotransmitter in the healthy brain. J Neural Transm 121(8):799–817. https://doi.org/10.1007/s00702-014-1180-8

    Article  CAS  PubMed  Google Scholar 

  68. Gemes G, Bangaru ML, Wu HE, Tang Q, Weihrauch D, Koopmeiners AS, Cruikshank JM, Kwok WM et al (2011) Store-operated Ca2+ entry in sensory neurons: functional role and the effect of painful nerve injury. J Neurosci 31(10):3536–3549. https://doi.org/10.1523/JNEUROSCI.5053-10.2011

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Kadoyama K, Matsuura K, Nakamura-Hirota T, Takano M, Otani M, Matsuyama S (2015) Changes in the expression of collapsin response mediator protein-2 during synaptic plasticity in the mouse hippocampus. J Neurosci Res 93(11):1684–1692. https://doi.org/10.1002/jnr.23626

    Article  CAS  PubMed  Google Scholar 

  70. Zhang J, Zhao B, Zhu X, Li J, Wu F, Li S, Gong X, Cha C et al (2018) Phosphorylation and SUMOylation of CRMP2 regulate the formation and maturation of dendritic spines. Brain Res Bull 139:21–30. https://doi.org/10.1016/j.brainresbull.2018.02.004

    Article  CAS  PubMed  Google Scholar 

  71. Chai Z, Wang C, Huang R, Wang Y, Zhang X, Wu Q, Wang Y, Wu X et al (2017) CaV2.2 gates calcium-independent but voltage-dependent secretion in mammalian sensory neurons. Neuron 96(6):1317–1326 e1314. https://doi.org/10.1016/j.neuron.2017.10.028

    Article  CAS  PubMed  Google Scholar 

  72. Fioravante D, Regehr WG (2011) Short-term forms of presynaptic plasticity. Curr Opin Neurobiol 21(2):269–274. https://doi.org/10.1016/j.conb.2011.02.003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Tomoyose O, Kodama D, Ono H, Tanabe M (2014) Presynaptic inhibitory effects of fluvoxamine, a selective serotonin reuptake inhibitor, on nociceptive excitatory synaptic transmission in spinal superficial dorsal horn neurons of adult mice. J Pharmacol Sci 126(2):136–145

    Article  CAS  PubMed  Google Scholar 

  74. Meng J, Wang J, Lawrence G, Dolly JO (2007) Synaptobrevin I mediates exocytosis of CGRP from sensory neurons and inhibition by botulinum toxins reflects their anti-nociceptive potential. J Cell Sci 120(Pt 16):2864–2874. https://doi.org/10.1242/jcs.012211

    Article  CAS  PubMed  Google Scholar 

  75. McCoy ES, Taylor-Blake B, Street SE, Pribisko AL, Zheng J, Zylka MJ (2013) Peptidergic CGRPalpha primary sensory neurons encode heat and itch and tonically suppress sensitivity to cold. Neuron 78(1):138–151. https://doi.org/10.1016/j.neuron.2013.01.030

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was supported by a grant from the National Natural Science Foundation of China (81603088) to J.Y., a grant from the National Institutes of Health to JMW (R01NS082283), National Institutes of Health awards (R01NS098772 from the National Institute of Neurological Disorders and Stroke and R01DA042852 from the National Institute on Drug Abuse to R.K.), and a Neurofibromatosis New Investigator Award from the Department of Defense Congressionally Directed Military Medical Research and Development Program (NF1000099) to R.K. S.S.B. and N.Y.N.P. were supported by funds to the Undergraduate Biology Research Program from the University of Arizona’s Senior Vice President for Research’s office, and the University of Arizona’s Native American Cancer Prevention Program (N.Y.N.P.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Rajesh Khanna.

Ethics declarations

Conflict of Interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Jie Yu and Aubin Moutal are co-first authors.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yu, J., Moutal, A., Dorame, A. et al. Phosphorylated CRMP2 Regulates Spinal Nociceptive Neurotransmission. Mol Neurobiol 56, 5241–5255 (2019). https://doi.org/10.1007/s12035-018-1445-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-018-1445-6

Keywords

Navigation