DOI QR코드

DOI QR Code

Spinal Gap Junction Channels in Neuropathic Pain

  • Jeon, Young Hoon (Department of Anesthesiology and Pain Medicine, School of Dentistry, Kyungpook National University) ;
  • Youn, Dong Ho (Department of Oral Physiology, School of Dentistry, Kyungpook National University)
  • Received : 2015.03.16
  • Accepted : 2015.06.25
  • Published : 2015.10.01

Abstract

Damage to peripheral nerves or the spinal cord is often accompanied by neuropathic pain, which is a complex, chronic pain state. Increasing evidence indicates that alterations in the expression and activity of gap junction channels in the spinal cord are involved in the development of neuropathic pain. Thus, this review briefly summarizes evidence that regulation of the expression, coupling, and activity of spinal gap junction channels modulates pain signals in neuropathic pain states induced by peripheral nerve or spinal cord injury. We particularly focus on connexin 43 and pannexin 1 because their regulation vastly attenuates symptoms of neuropathic pain. We hope that the study of gap junction channels eventually leads to the development of a suitable treatment tool for patients with neuropathic pain.

Keywords

References

  1. Scholz J, Woolf CJ. Can we conquer pain? Nat Neurosci 2002; 5 Suppl: 1062-7. https://doi.org/10.1038/nn942
  2. Scholz J, Woolf CJ. The neuropathic pain triad: neurons, immune cells and glia. Nat Neurosci 2007; 10: 1361-8. https://doi.org/10.1038/nn1992
  3. Shubayev VI, Angert M, Dolkas J, Campana WM, Palenscar K, Myers RR. TNFalpha-induced MMP-9 promotes macrophage recruitment into injured peripheral nerve. Mol Cell Neurosci 2006; 31: 407-15. https://doi.org/10.1016/j.mcn.2005.10.011
  4. Bethea JR. Spinal cord injury-induced inflammation: a dual-edged sword. Prog Brain Res 2000; 128: 33-42. https://doi.org/10.1016/S0079-6123(00)28005-9
  5. Farahani R, Pina-Benabou MH, Kyrozis A, Siddiq A, Barradas PC, Chiu FC, et al. Alterations in metabolism and gap junction expression may determine the role of astrocytes as "good samaritans" or executioners. Glia 2005; 50:351-61. https://doi.org/10.1002/glia.20213
  6. Faulkner JR, Herrmann JE, Woo MJ, Tansey KE, Doan NB, Sofroniew MV. Reactive astrocytes protect tissue and preserve function after spinal cord injury. J Neurosci 2004; 24:2143-55. https://doi.org/10.1523/JNEUROSCI.3547-03.2004
  7. Tsuda M, Shigemoto-Mogami Y, Koizumi S, Mizokoshi A, Kohsaka S, Salter MW, et al. P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature 2003; 424: 778-83. https://doi.org/10.1038/nature01786
  8. Gwak YS, Kang J, Unabia GC, Hulsebosch CE. Spatial and temporal activation of spinal glial cells: role of gliopathy in central neuropathic pain following spinal cord injury in rats. Exp Neurol 2012; 234: 362-72. https://doi.org/10.1016/j.expneurol.2011.10.010
  9. Wang X, Arcuino G, Takano T, Lin J, Peng WG, Wan P, et al. P2X7 receptor inhibition improves recovery after spinal cord injury. Nat Med 2004; 10: 821-7. https://doi.org/10.1038/nm1082
  10. Peng W, Cotrina ML, Han X, Yu H, Bekar L, Blum L, et al. Systemic administration of an antagonist of the ATP-sensitive receptor P2X7 improves recovery after spinal cord injury. Proc Natl Acad Sci U S A 2009; 106: 12489-93. https://doi.org/10.1073/pnas.0902531106
  11. Springer JE, Azbill RD, Knapp PE. Activation of the caspase-3 apoptotic cascade in traumatic spinal cord injury. Nat Med 1999; 5: 943-6. https://doi.org/10.1038/11387
  12. Collo G, Neidhart S, Kawashima E, Kosco-Vilbois M, North RA, Buell G. Tissue distribution of the P2X7 receptor. Neuropharmacology 1997; 36: 1277-83. https://doi.org/10.1016/S0028-3908(97)00140-8
  13. Kawasaki Y, Zhang L, Cheng JK, Ji RR. Cytokine mechanisms of central sensitization: distinct and overlapping role of interleukin-1beta, interleukin-6, and tumor necrosis factoralpha in regulating synaptic and neuronal activity in the superficial spinal cord. J Neurosci 2008; 28: 5189-94. https://doi.org/10.1523/JNEUROSCI.3338-07.2008
  14. Cotrina ML, Nedergaard M. Physiological and pathological functions of P2X7 receptor in the spinal cord. Purinergic Signal 2009; 5: 223-32. https://doi.org/10.1007/s11302-009-9138-2
  15. Reyes EP, Cerpa V, Corvalan L, Retamal MA. Cxs and Panx-hemichannels in peripheral and central chemosensing in mammals. Front Cell Neurosci 2014; 8: 123. https://doi.org/10.3389/fncel.2014.00123
  16. Takeuchi H, Suzumura A. Gap junctions and hemichannels composed of connexins: potential therapeutic targets for neurodegenerative diseases. Front Cell Neurosci 2014; 8: 189.
  17. Goldberg GS, Lampe PD, Nicholson BJ. Selective transfer of endogenous metabolites through gap junctions composed of different connexins. Nat Cell Biol 1999; 1: 457-9. https://doi.org/10.1038/15693
  18. De Vuyst E, Decrock E, De Bock M, Yamasaki H, Naus CC, Evans WH, et al. Connexin hemichannels and gap junction channels are differentially influenced by lipopolysaccharide and basic fibroblast growth factor. Mol Biol Cell 2007; 18:34-46. https://doi.org/10.1091/mbc.e06-03-0182
  19. Retamal MA, Froger N, Palacios-Prado N, Ezan P, Saez PJ, Saez JC, et al. Cx43 hemichannels and gap junction channels in astrocytes are regulated oppositely by proinflammatory cytokines released from activated microglia. J Neurosci 2007; 27: 13781-92. https://doi.org/10.1523/JNEUROSCI.2042-07.2007
  20. Laird DW. The gap junction proteome and its relationship to disease. Trends Cell Biol 2010; 20: 92-101. https://doi.org/10.1016/j.tcb.2009.11.001
  21. Giaume C, Leybaert L, Naus CC, Saez JC. Connexin and pannexin hemichannels in brain glial cells: properties, pharmacology, and roles. Front Pharmacol 2013; 4: 88.
  22. Stout CE, Costantin JL, Naus CC, Charles AC. Intercellular calcium signaling in astrocytes via ATP release through connexin hemichannels. J Biol Chem 2002; 277: 10482-8. https://doi.org/10.1074/jbc.M109902200
  23. Ye ZC, Wyeth MS, Baltan-Tekkok S, Ransom BR. Functional hemichannels in astrocytes: a novel mechanism of glutamate release. J Neurosci 2003; 23: 3588-96. https://doi.org/10.1523/JNEUROSCI.23-09-03588.2003
  24. Chen G, Park CK, Xie RG, Berta T, Nedergaard M, Ji RR. Connexin-43 induces chemokine release from spinal cord astrocytes to maintain late-phase neuropathic pain in mice. Brain 2014; 137: 2193-209. https://doi.org/10.1093/brain/awu140
  25. Wu A, Green CR, Rupenthal ID, Moalem-Taylor G. Role of gap junctions in chronic pain. J Neurosci Res 2012; 90:337-45. https://doi.org/10.1002/jnr.22764
  26. Rozental R, Giaume C, Spray DC. Gap junctions in the nervous system. Brain Res Brain Res Rev 2000; 32: 11-5. https://doi.org/10.1016/S0165-0173(99)00095-8
  27. Spataro LE, Sloane EM, Milligan ED, Wieseler-Frank J, Schoeniger D, Jekich BM, et al. Spinal gap junctions:potential involvement in pain facilitation. J Pain 2004; 5:392-405. https://doi.org/10.1016/j.jpain.2004.06.006
  28. Roh DH, Yoon SY, Seo HS, Kang SY, Han HJ, Beitz AJ, et al. Intrathecal injection of carbenoxolone, a gap junction decoupler, attenuates the induction of below-level neuropathic pain after spinal cord injury in rats. Exp Neurol 2010; 224: 123-32. https://doi.org/10.1016/j.expneurol.2010.03.002
  29. Rohlmann A, Laskawi R, Hofer A, Dermietzel R, Wolff JR. Astrocytes as rapid sensors of peripheral axotomy in the facial nucleus of rats. Neuroreport 1994; 5: 409-12. https://doi.org/10.1097/00001756-199401120-00009
  30. Ochalski PA, Frankenstein UN, Hertzberg EL, Nagy JI. Connexin-43 in rat spinal cord: localization in astrocytes and identification of heterotypic astro-oligodendrocytic gap junctions. Neuroscience 1997; 76: 931-45.
  31. Theriault E, Frankenstein UN, Hertzberg EL, Nagy JI. Connexin43 and astrocytic gap junctions in the rat spinal cord after acute compression injury. J Comp Neurol 1997; 382: 199-214. https://doi.org/10.1002/(SICI)1096-9861(19970602)382:2<199::AID-CNE5>3.0.CO;2-Z
  32. Cronin M, Anderson PN, Cook JE, Green CR, Becker DL. Blocking connexin43 expression reduces inflammation and improves functional recovery after spinal cord injury. Mol Cell Neurosci 2008; 39: 152-60. https://doi.org/10.1016/j.mcn.2008.06.005
  33. Chen MJ, Kress B, Han X, Moll K, Peng W, Ji RR, et al. Astrocytic CX43 hemichannels and gap junctions play a crucial role in development of chronic neuropathic pain following spinal cord injury. Glia 2012; 60: 1660-70. https://doi.org/10.1002/glia.22384
  34. O'Carroll SJ, Alkadhi M, Nicholson LF, Green CR. Connexin 43 mimetic peptides reduce swelling, astrogliosis, and neuronal cell death after spinal cord injury. Cell Commun Adhes 2008; 15: 27-42. https://doi.org/10.1080/15419060802014164
  35. Huang C, Han X, Li X, Lam E, Peng W, Lou N, et al. Critical role of connexin 43 in secondary expansion of traumatic spinal cord injury. J Neurosci 2012; 32: 3333-8. https://doi.org/10.1523/JNEUROSCI.1216-11.2012
  36. Xu Q, Cheong YK, He SQ, Tiwari V, Liu J, Wang Y, et al. Suppression of spinal connexin 43 expression attenuates mechanical hypersensitivity in rats after an L5 spinal nerve injury. Neurosci Lett 2014; 566: 194-9. https://doi.org/10.1016/j.neulet.2014.03.004
  37. Sandilos JK, Bayliss DA. Physiological mechanisms for the modulation of pannexin 1 channel activity. J Physiol 2012; 590: 6257-66. https://doi.org/10.1113/jphysiol.2012.240911
  38. Bao L, Locovei S, Dahl G. Pannexin membrane channels are mechanosensitive conduits for ATP. FEBS Lett 2004; 572:65-8. https://doi.org/10.1016/j.febslet.2004.07.009
  39. Bravo D, Ibarra P, Retamal J, Pelissier T, Laurido C, Hernandez A, et al. Pannexin 1: a novel participant in neuropathic pain signaling in the rat spinal cord. Pain 2014; 155: 2108-15. https://doi.org/10.1016/j.pain.2014.07.024

Cited by

  1. Breakthrough Cancer Pain Is Associated with Spinal Gap Junction Activation via Regulation of Connexin 43 in a Mouse Model vol.11, pp.1662-5102, 2017, https://doi.org/10.3389/fncel.2017.00207
  2. In silico target network analysis of de novo-discovered, tick saliva-specific microRNAs reveals important combinatorial effects in their interference with vertebrate host physiology vol.23, pp.8, 2017, https://doi.org/10.1261/rna.061168.117
  3. 신경병증 통증 모델에서 Carbenoxolone과 P2x receptor 길항제의 효과 vol.17, pp.2, 2015, https://doi.org/10.5762/kais.2016.17.2.123
  4. The role of connexin43 in neuropathic pain induced by spinal cord injury vol.51, pp.6, 2015, https://doi.org/10.1093/abbs/gmz038
  5. Simultaneous Activation of Mu and Delta Opioid Receptors Reduces Allodynia and Astrocytic Connexin 43 in an Animal Model of Neuropathic Pain vol.56, pp.11, 2015, https://doi.org/10.1007/s12035-019-1607-1
  6. Blockage of Electrical Synapses Can Alleviate Neuropathic Pain?*Blockage of Electrical Synapses Can Alleviate Neuropathic Pain? vol.32, pp.4, 2015, https://doi.org/10.3166/dea-2020-0080
  7. Distinct roles for innexin gap junctions and hemichannels in mechanosensation vol.9, pp.None, 2015, https://doi.org/10.7554/elife.50597
  8. Pannexin 1 Transgenic Mice: Human Diseases and Sleep-Wake Function Revision vol.22, pp.10, 2015, https://doi.org/10.3390/ijms22105269
  9. Mechanisms of ATP release in pain: role of pannexin and connexin channels vol.17, pp.4, 2015, https://doi.org/10.1007/s11302-021-09822-6
  10. Effects of cyanocobalamin and its combination with morphine on neuropathic rats and the relationship between these effects and thrombospondin-4 expression vol.35, pp.1, 2022, https://doi.org/10.3344/kjp.2022.35.1.66