Skip to main content

Advertisement

Log in

Kinesins: Motor Proteins as Novel Target for the Treatment of Chronic Pain

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

A Correction to this article was published on 17 September 2020

This article has been updated

Abstract

Kinesins are one of the neoteric and efficacious targets recently reported to play an important role in the initiation and progression of chronic pain. Kinesins are anterograde microtubule-based motor proteins that are involved in trafficking of receptors including nociceptors and progression of pain. The specific kinesin and regulatory proteins interplay is crucial for the delivery of nociceptors to the synapse. If this complex and less understood interplay is inhibited, it may result in a decrease in central sensitization, and thus attenuation of pain. This review is focused on the transportation process of receptors/cargos, the role of regulatory proteins influencing the respective kinesin, and their relationship with chronic pain. The review also features specific strategies adopted by researchers for targeting kinesin and chronic pain. Considering the recent preclinical success of kinesin inhibition in pain, it is expected that inhibitors for kinesin or enzymes responsible for kinesin activation could be developed or repurposed as alternative, safe, and potential therapies 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

Similar content being viewed by others

Change history

  • 17 September 2020

    The original version of this article unfortunately contained mistakes.

References

  1. Kamerman PR, Wadley AL, Davis K, Hietaharju A, Jain P, Kopf A, Meyer A-C, Raja SN et al (2015) World Health Organization (WHO) essential medicines lists: where are the drugs to treat neuropathic pain? Pain 156:793–797

    Article  Google Scholar 

  2. Murray CJ, Lopez AD (2013) Measuring the global burden of disease. N Engl J Med 369:448–457

    Article  CAS  Google Scholar 

  3. Tiwari V, Yang F, He SQ, Shechter R, Zhang C, Shu B, Zhang T, Tiwari V et al (2016) Activation of peripheral μ-opioid receptors by Dermorphin [D-Arg2, Lys4] (1-4) amide leads to modality-preferred inhibition of neuropathic pain. Anesthesiology 124(3):706–720

    Article  CAS  Google Scholar 

  4. Vlaeyen JW, Linton SJ (2012) Fear-avoidance model of chronic musculoskeletal pain: 12 years on. Pain 153:1144–1147

    Article  Google Scholar 

  5. Belvisi MG (2003) Sensory nerves and airway inflammation: role of Aδ and C-fibres. Pulm Pharmacol Ther 16:1–7

    Article  CAS  Google Scholar 

  6. Craig A (2003) Pain mechanisms: labeled lines versus convergence in central processing. Annu Rev Neurosci 26:1–30

    Article  CAS  Google Scholar 

  7. Cook SP, Vulchanova L, Hargreaves KM, Elde R, Mccleskey EW (1997) Distinct ATP receptors on pain-sensing and stretch-sensing neurons. Nature 387:505–508

    Article  CAS  Google Scholar 

  8. Mckemy DD (2005) How cold is it? TRPM8 and TRPA1 in the molecular logic of cold sensation. Mol Pain 1:16

    Article  Google Scholar 

  9. Ploghaus A, Tracey I, Gati JS, Clare S, Menon RS, Matthews PM, Rawlins JNP (1999) Dissociating pain from its anticipation in the human brain. Science 284:1979–1981

    Article  CAS  Google Scholar 

  10. Coutaux A, Adam F, Willer J-C, Le Bars D (2005) Hyperalgesia and allodynia: peripheral mechanisms. Joint Bone Spine 72:359–371

    Article  Google Scholar 

  11. Woolf CJ (2000) Neuronal plasticity: increasing the gain in pain. Science 288:1765–1768

    Article  CAS  Google Scholar 

  12. Woolf CJ, Chong M-S (1993) Preemptive analgesia-treating postoperative pain by preventing the establishment of central sensitization. Anesth Analg 77:362–379

    Article  CAS  Google Scholar 

  13. Hirokawa N, Takemura R (2003) Biochemical and molecular characterization of diseases linked to motor proteins. Trends Biochem Sci 28:558–565

    Article  CAS  Google Scholar 

  14. Hirokawa N, Noda Y, Tanaka Y, Niwa S (2009) Kinesin superfamily motor proteins and intracellular transport. Nat Rev Mol Cell Biol 10:682–696

    Article  CAS  Google Scholar 

  15. Sablin EP, Case RB, Dai SC, Hart CL, Ruby A, Vale RD, Fletterick RJ (1998) Direction determination in the minus-end-directed kinesin motor ncd. Nature 395:813–816

    Article  CAS  Google Scholar 

  16. Pollard TD, Goldman RD (2017) The Cytoskeleton. Cold Spring Harbor Laboratory Press, New York

  17. Wiche G (1998) Role of plectin in cytoskeleton organization and dynamics. J Cell Sci 111:2477–2486

    CAS  PubMed  Google Scholar 

  18. Squire J, Parry DA (2005) Fibrous proteins: muscle and molecular motors. Gulf Professional Publishing, Houston

    Google Scholar 

  19. Tanenbaum ME, Macurek L, Janssen A, Geers EF, Alvarez-Fernandez M assembly. Curr Biol 19:1703–1711

  20. Imanishi M, Endres NF, Gennerich A Vale, RD (2006) Autoinhibition regulates the motility of the C. elegans intraflagellar transport motor OSM-3. J. Cell Biol, 174:931–937

    Article  CAS  Google Scholar 

  21. Hammond JW, Cai D, Blasius TL, Li Z, Jiang Y, Jih GT, Meyhofer E, Verhey KJ (2009a) Mammalian Kinesin-3 motors are dimeric in vivo and move by processive motility upon release of autoinhibition. PLoS Biol 7:e1000072

    Article  Google Scholar 

  22. Lwin KM, Li D, Bretscher A (2016) Kinesin-related Smy1 enhances the Rab-dependent association of myosin-V with secretory cargo. Mol Biol Cell 27:2450–2462

    Article  CAS  Google Scholar 

  23. Twelvetrees AE, Pernigo S, Sanger A, Guedes-Dias P, Schiavo G, Steiner RA, Dodding MP, Holzbaur EL (2016) The dynamic localization of cytoplasmic dynein in neurons is driven by kinesin-1. Neuron 90:1000–1015

    Article  CAS  Google Scholar 

  24. Karcher RL, Deacon SW, Gelfand VI (2002) Motor–cargo interactions: the key to transport specificity. Trends Cell Biol 12:21–27

    Article  CAS  Google Scholar 

  25. Akhmanova A, Hammer JA (2010) Linking molecular motors to membrane cargo. Curr Opin Cell Biol 22:479–487

    Article  CAS  Google Scholar 

  26. Berk A, Zipursky S, Lodish H (2000) Molecular cell biology 4th edn. National Center for Biotechnology InformationÕs Bookshelf

  27. Ren B-X, Gu X-P, Zheng Y-G, Liu C-L, Wang D, Sun Y-E, Ma Z-L (2012) Intrathecal injection of metabotropic glutamate receptor subtype 3 and 5 agonist/antagonist attenuates bone cancer pain by inhibition of spinal astrocyte activation in a mouse model. Anesthesiology 116:122–132

    Article  CAS  Google Scholar 

  28. Verhey KJ, Hammond JW (2009) Traffic control: regulation of kinesin motors. Nat Rev Mol Cell Biol 10:765–777

    Article  CAS  Google Scholar 

  29. Verhey KJ, Meyer D, Deehan R, Blenis J, Schnapp BJ, Rapoport TA, Margolis B (2001) Cargo of kinesin identified as JIP scaffolding proteins and associated signaling molecules. J Cell Biol 152:959–970

    Article  CAS  Google Scholar 

  30. Byrd DT, Kawasaki M, Walcoff M, Hisamoto N, Matsumoto K, Jin Y (2001) UNC-16, a JNK-signaling scaffold protein, regulates vesicle transport in C. elegans. Neuron, 32:(5)787–800

    Article  CAS  Google Scholar 

  31. Cavalli V, Kujala P, Klumperman J, Goldstein LS (2005) Sunday Driver links axonal transport to damage signaling. J Cell Biol 168:775–787

    Article  CAS  Google Scholar 

  32. Cai D, Mcewen DP, Martens JR, Meyhofer E, Verhey KJ (2009) Single molecule imaging reveals differences in microtubule track selection between kinesin motors. PLoS Biol 7:e1000216

    Article  Google Scholar 

  33. Dunn S, Morrison EE, Liverpool TB, Molina-París C, Cross RA, Alonso MC, Peckham M (2008) Differential trafficking of Kif5c on tyrosinated and detyrosinated microtubules in live cells. J Cell Sci 121:1085–1095

    Article  CAS  Google Scholar 

  34. Konishi Y, Setou M (2009) Tubulin tyrosination navigates the kinesin-1 motor domain to axons. Nat Neurosci 12:559–567

    Article  CAS  Google Scholar 

  35. Reed NA, Cai D, Blasius TL, Jih GT, Meyhofer E, Gaertig J, Verhey KJ (2006) Microtubule acetylation promotes kinesin-1 binding and transport. Curr Biol 16:2166–2172

    Article  CAS  Google Scholar 

  36. Ikegami K, Heier RL, Taruishi M, Takagi H, Mukai M, Shimma S, Taira S, Hatanaka K et al (2007) Loss of α-tubulin polyglutamylation in ROSA22 mice is associated with abnormal targeting of KIF1A and modulated synaptic function. Proc Natl Acad Sci 104:3213–3218

    Article  CAS  Google Scholar 

  37. Rinaldi F, Bassi MT, Todeschini A, Rota S, Arnoldi A, Padovani A, Filosto M (2015) A novel mutation in motor domain of KIF5A associated with an HSP/axonal neuropathy phenotype. J Clin Neuromuscul Dis 16:153–158

    Article  Google Scholar 

  38. Su Y-Y, Ye M, Li L, Liu C, Pan J, Liu W-W, Jiang Y, Jiang X-Y et al (2013a) KIF5B promotes the forward transport and axonal function of the voltage-gated sodium channel Nav1. 8. J Neurosci 33:17884–17896

    Article  CAS  Google Scholar 

  39. Wang N, Xu J (2015a) Functions of kinesin superfamily proteins in neuroreceptor trafficking. Biomed Res Int 2015:639301

    PubMed  PubMed Central  Google Scholar 

  40. Liu M, Liu Y, Hou B, Bu D, Shi L, Gu X, Ma Z (2015b) Kinesin superfamily protein 17 contributes to the development of bone cancer pain by participating in NR2B transport in the spinal cord of mice. Oncol Rep 33:1365–1371

    Article  CAS  Google Scholar 

  41. Bo J, Zhang W, Sun X, Yang Y, Liu X, Jiang M, Ma Z, Gu X (2014) The cyclic AMP response element-binding protein antisense oligonucleotide induced anti-nociception and decreased the expression of KIF17 in spinal cord after peripheral nerve injury in mice. Int J Clin Exp Med 7:5181–5191

    PubMed  PubMed Central  Google Scholar 

  42. Dilley A, Richards N, Pulman KG, Bove GM (2013) Disruption of fast axonal transport in the rat induces behavioral changes consistent with neuropathic pain. J Pain 14:1437–1449

    Article  CAS  Google Scholar 

  43. Ni K, Zhou Y, Sun YE, Liu Y, Gu XP, Ma ZL (2014) Intrathecal injection of selected peptide Myr-RC-13 attenuates bone cancer pain by inhibiting KIF17 and NR2B expression. Pharmacol Biochem Behav 122:228–233

    Article  CAS  Google Scholar 

  44. Liu Y, Liang Y, Hou B, Liu M, Yang X, Liu C, Zhang J, Zhang W et al (2014) The inhibitor of calcium/calmodulin-dependent protein kinase II KN93 attenuates bone cancer pain via inhibition of KIF17/NR2B trafficking in mice. Pharmacol Biochem Behav 124:19–26

    Article  CAS  Google Scholar 

  45. Xing B-M, Yang Y-R, Du J-X, Chen H-J, Qi C, Huang Z-H, Zhang Y, Wang Y (2012a) Cyclin-dependent kinase 5 controls TRPV1 membrane trafficking and the heat sensitivity of nociceptors through KIF13B. J Neurosci 32:14709–14721

    Article  CAS  Google Scholar 

  46. Liu J, Du J, Yang Y, Wang Y (2015a) Phosphorylation of TRPV1 by cyclin-dependent kinase 5 promotes TRPV1 surface localization, leading to inflammatory thermal hyperalgesia. Exp Neurol 273:253–262

    Article  CAS  Google Scholar 

  47. Yin X, Takei Y, Kido MA, Hirokawa N (2011) Molecular motor KIF17 is fundamental for memory and learning via differential support of synaptic NR2A/2B levels. Neuron 70:310–325

    Article  CAS  Google Scholar 

  48. Brüggemann I, Schulz S, Wiborny D, Höllt V (2000) Colocalization of the mu-opioid receptor and calcium/calmodulin-dependent kinase II in distinct pain-processing brain regions. Brain Res Mol Brain Res 85(1–2):239–250

    Article  Google Scholar 

  49. Carlton SM (2002) Localization of CaMKIIalpha in rat primary sensory neurons: increase in inflammation. Brain Res 947(2):252–259

    Article  CAS  Google Scholar 

  50. Caterina MJ, Leffler A, Malmberg A, Martin W, Trafton J, Petersen-Zeitz K, Koltzenburg M, Basbaum A et al (2000) Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science 288:306–313

    Article  CAS  Google Scholar 

  51. Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D (1997) The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 389:816–824

    Article  CAS  Google Scholar 

  52. Camprubi-Robles M, Planells-Cases R, Ferrer-Montiel A (2009) Differential contribution of SNARE-dependent exocytosis to inflammatory potentiation of TRPV1 in nociceptors. FASEB J 23:3722–3733

    Article  CAS  Google Scholar 

  53. Morenilla-Palao C, Planells-Cases R, Garcia-Sanz N, Ferrer-Montiel A (2004) Regulated exocytosis contributes to protein kinase C potentiation of vanilloid receptor activity. J Biol Chem 279:25665–25672

    Article  CAS  Google Scholar 

  54. Xu Y, Zhang F, Su Z, Mcnew JA, Shin YK (2005) Hemifusion in SNARE-mediated membrane fusion. Nat Struct Mol Biol 12:417–422

    Article  CAS  Google Scholar 

  55. Züchner S, Vance JM (2006) Mechanisms of disease: a molecular genetic update on hereditary axonal neuropathies. Nat Rev Neurol 2:45

    Article  Google Scholar 

  56. Vallat J-M, Tazir M, Magdelaine C, Sturtz F, Grid D (2005) Autosomal-recessive Charcot-Marie-Tooth diseases. J Neuropathol Exp Neurol 64:363–370

    Article  CAS  Google Scholar 

  57. Manganelli F, Tozza S, Pisciotta C, Bellone E, Iodice R, Nolano M, Geroldi A, Capponi S et al (2014) Charcot-Marie-Tooth disease: frequency of genetic subtypes in a Southern Italy population. J Peripher Nerv Syst 19:292–298

    Article  CAS  Google Scholar 

  58. Crimella C, Baschirotto C, Arnoldi A, Tonelli A, Tenderini E, Airoldi G, Martinuzzi A, Trabacca A et al (2012) Mutations in the motor and stalk domains of KIF5A in spastic paraplegia type 10 and in axonal Charcot-Marie-Tooth type 2. Clin Genet 82(2):157–164

    Article  CAS  Google Scholar 

  59. López E, Casasnovas C, Giménez J, Santamaría R, Jm TERRAZAS, Volpini V (2015) Identification of two novel KIF5A mutations in hereditary spastic paraplegia associated with mild peripheral neuropathy. J Neurol Sci 358(1–2):422–427

    Article  Google Scholar 

  60. Levinson SR, Luo S, Henry MA (2012) The role of sodium channels in chronic pain. Muscle Nerve 46(2):155–165

    Article  CAS  Google Scholar 

  61. Coward K, Plumpton C, Facer P, Birch R, Carlstedt T, Tate S, Bountra C, Anand P (2000) Immunolocalization of SNS/PN3 and NaN/SNS2 sodium channels in human pain states. Pain, 85:(1-2)41–50

    Article  CAS  Google Scholar 

  62. Hirokawa N, Niwa S, Tanaka Y (2010) Molecular motors in neurons: transport mechanisms and roles in brain function, development, and disease. Neuron, 68:(4)610–638

    Article  CAS  Google Scholar 

  63. Purcell JW, Davis J, Reddy M, Martin S, Samayoa K, Vo H, Thomsen K, Bean P et al (2010) Activity of the kinesin spindle protein inhibitor ispinesib (SB-715992) in models of breast cancer. Clin Cancer Res 16:566–576

    Article  CAS  Google Scholar 

  64. Guillaud L, Setou M, Hirokawa N (2003) KIF17 dynamics and regulation of NR2B trafficking in hippocampal neurons. J Neurosci 23:131–140

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors wish to express their thanks to Prof. D.K. Singh for his editorial contribution and to the Director, NIPER Ahmedabad for providing necessary facilities and infrastructure.

Funding

This work is supported by Department of Pharmaceuticals, Ministry of Chemical and Fertilizers, Govt of India, National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Gandhinagar, Gujarat, India and Department of Science and Technology (DST), Government of India, Early Career Research Grant (ECR/2016/001846) awarded to Dr. Vinod Tiwari and by NIH grants (R01NS094664, R01NS094224, and R01DA033390) for Yuan-Xiang Tao.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Vinod Tiwari.

Ethics declarations

Conflict of Interest

The authors declare that they have no conflict of interest.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shantanu, P.A., Sharma, D., Sharma, M. et al. Kinesins: Motor Proteins as Novel Target for the Treatment of Chronic Pain. Mol Neurobiol 56, 3854–3864 (2019). https://doi.org/10.1007/s12035-018-1327-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-018-1327-y

Keywords

Navigation