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.

  • Review Article
  • Published:

Astrocytes in chronic pain and itch

Abstract

Astrocytes are critical for maintaining the homeostasis of the CNS. Increasing evidence suggests that a number of neurological and neuropsychiatric disorders, including chronic pain, may result from astrocyte ‘gliopathy’. Indeed, in recent years there has been substantial progress in our understanding of how astrocytes can regulate nociceptive synaptic transmission via neuronal–glial and glial–glial cell interactions, as well as the involvement of spinal and supraspinal astrocytes in the modulation of pain signalling and the maintenance of neuropathic pain. A role of astrocytes in the pathogenesis of chronic itch is also emerging. These developments suggest that targeting the specific pathways that are responsible for astrogliopathy may represent a novel approach to develop therapies for chronic pain and chronic itch.

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

Fig. 1: Homeostatic functions of astrocytes.
Fig. 2: Astrocyte–neuron and astrocyte–microglial cell interactions in neuropathic pain.
Fig. 3: Propagation of chronic itch through disinhibition and astrocyte activation.
Fig. 4: Future directions in the investigation of astrocytes in chronic pain.

Similar content being viewed by others

References

  1. Herculano-Houzel, S. The glia/neuron ratio: how it varies uniformly across brain structures and species and what that means for brain physiology and evolution. Glia 62, 1377–1391 (2014).

    Article  PubMed  Google Scholar 

  2. Ben Haim, L. & Rowitch, D. H. Functional diversity of astrocytes in neural circuit regulation. Nat. Rev. Neurosci. 18, 31–41 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Giaume, C. & McCarthy, K. D. Control of gap-junctional communication in astrocytic networks. Trends Neurosci. 19, 319–325 (1996).

    Article  CAS  PubMed  Google Scholar 

  4. Ji, R. R., Berta, T. & Nedergaard, M. Glia and pain: is chronic pain a gliopathy? Pain 154, S10–S28 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Mathewson, A. J. & Berry, M. Observations on the astrocyte response to a cerebral stab wound in adult rats. Brain Res. 327, 61–69 (1985).

    Article  CAS  PubMed  Google Scholar 

  6. Kimelberg, H. K. & Nedergaard, M. Functions of astrocytes and their potential as therapeutic targets. Neurotherapeutics 7, 338–353 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Iadecola, C. & Nedergaard, M. Glial regulation of the cerebral microvasculature. Nat. Neurosci. 10, 1369–1376 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Oberheim, N. A. et al. Loss of astrocytic domain organization in the epileptic brain. J. Neurosci. 28, 3264–3276 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Gao, Y. J. & Ji, R.-R. Targeting astrocyte signaling for chronic pain. Neurotherapeutics. 7, 482–493 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bushong, E. A., Martone, M. E., Jones, Y. Z. & Ellisman, M. H. Protoplasmic astrocytes in CA1 stratum radiatum occupy separate anatomical domains. J. Neurosci. 22, 183–192 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Oberheim, N. A. et al. Uniquely hominid features of adult human astrocytes. J. Neurosci. 29, 3276–3287 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Halassa, M. M., Fellin, T., Takano, H., Dong, J. H. & Haydon, P. G. Synaptic islands defined by the territory of a single astrocyte. J. Neurosci. 27, 6473–6477 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Oberheim, N. A., Wang, X., Goldman, S. & Nedergaard, M. Astrocytic complexity distinguishes the human brain. Trends Neurosci. 29, 547–553 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Han, X. et al. Forebrain engraftment by human glial progenitor cells enhances synaptic plasticity and learning in adult mice. Cell Stem Cell 12, 342–353 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Laug, D., Glasgow, S. M. & Deneen, B. A glial blueprint for gliomagenesis. Nat. Rev. Neurosci. 19, 393–403 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Clarke, L. E. & Barres, B. A. Emerging roles of astrocytes in neural circuit development. Nat. Rev. Neurosci. 14, 311–321 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Molofsky, A. V. et al. Astrocytes and disease: a neurodevelopmental perspective. Genes Dev. 26, 891–907 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ji, R.-R., Xu, Z. Z. & Gao, Y. J. Emerging targets in neuroinflammation-driven chronic pain. Nat. Rev. Drug Discov. 13, 533–548 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ransohoff, R. M. How neuroinflammation contributes to neurodegeneration. Science 353, 777–783 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Heppner, F. L., Ransohoff, R. M. & Becher, B. Immune attack: the role of inflammation in Alzheimer disease. Nat. Rev. Neurosci. 16, 358–372 (2015).

    Article  CAS  PubMed  Google Scholar 

  21. Ji, R.-R., Chamessian, A. & Zhang, Y. Q. Pain regulation by non-neuronal cells and inflammation. Science 354, 572–577 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Grace, P. M., Hutchinson, M. R., Maier, S. F. & Watkins, L. R. Pathological pain and the neuroimmune interface. Nat. Rev. Immunol. 14, 217–231 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Costigan, M., Scholz, J. & Woolf, C. J. Neuropathic pain: a maladaptive response of the nervous system to damage. Annu. Rev. Neurosci. 32, 1–32 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ji, R.-R., Nackley, A., Huh, Y., Terrando, N. & Maixner, W. Neuroinflammation and central sensitization in chronic and widespread pain. Anesthesiology 129, 343–366 (2018).

    Article  PubMed  Google Scholar 

  25. Tsuda, M. Modulation of pain and itch by spinal glia. Neurosci. Bull. 34, 178–185 (2018).

    Article  PubMed  Google Scholar 

  26. Verkhratsky, A. et al. Neurological diseases as primary gliopathies: a reassessment of neurocentrism. ASN Neuro. 4, e0008 (2012).

    Article  Google Scholar 

  27. Simard, M. & Nedergaard, M. The neurobiology of glia in the context of water and ion homeostasis. Neurosci. 129, 877–896 (2004).

    Article  CAS  Google Scholar 

  28. Cui, Y. et al. Astroglial Kir4.1 in the lateral habenula drives neuronal bursts in depression. Nature 554, 323–327 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Cornell-Bell, A. H., Finkbeiner, S. M., Cooper, M. S. & Smith, S. J. Glutamate induces calcium waves in cultured astrocytes: long-range glial signaling. Science 247, 470–473 (1990).

    Article  CAS  PubMed  Google Scholar 

  30. Nedergaard, M. & Verkhratsky, A. Artifact versus reality — how astrocytes contribute to synaptic events. Glia 60, 1013–1023 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Djukic, B., Casper, K. B., Philpot, B. D., Chin, L. S. & McCarthy, K. D. Conditional knock-out of Kir4.1 leads to glial membrane depolarization, inhibition of potassium and glutamate uptake, and enhanced short-term synaptic potentiation. J. Neurosci. 27, 11354–11365 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Verkhratsky, A. & Nedergaard, M. Astroglial cradle in the life of the synapse. Philos. Trans. R. Soc. Lond. B Biol. Sci. 369, 20130595 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Eroglu, C. & Barres, B. A. Regulation of synaptic connectivity by glia. Nature 468, 223–231 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Araque, A., Parpura, V., Sanzgiri, R. P. & Haydon, P. G. Tripartite synapses: glia, the unacknowledged partner. Trends Neurosci. 22, 208–215 (1999).

    Article  CAS  PubMed  Google Scholar 

  35. Sun, W. et al. Glutamate-dependent neuroglial calcium signaling differs between young and adult brain. Science 339, 197–200 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ding, F. et al. α1-Adrenergic receptors mediate coordinated Ca2+ signaling of cortical astrocytes in awake, behaving mice. Cell Calcium 54, 387–394 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Srinivasan, R. et al. Ca2+ signaling in astrocytes from Ip3r2–/– mice in brain slices and during startle responses in vivo. Nat. Neurosci. 18, 708–717 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Paukert, M. et al. Norepinephrine controls astroglial responsiveness to local circuit activity. Neuron 82, 1263–1270 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Wang, F. et al. Astrocytes modulate neural network activity by Ca2+-dependent uptake of extracellular K(+). Sci. Signal. 5, ra26 (2012).

    PubMed  PubMed Central  Google Scholar 

  40. Iliff, J. J. et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci. Transl. Med. 4, 147ra111 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Nedergaard, M. Garbage truck of the brain. Science 340, 1529–1530 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Goldman, N. et al. Adenosine A1 receptors mediate local anti-nociceptive effects of acupuncture. Nat. Neurosci. 13, 883–888 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Zhang, J. M. et al. ATP released by astrocytes mediates glutamatergic activity-dependent heterosynaptic suppression. Neuron 40, 971–982 (2003).

    Article  CAS  PubMed  Google Scholar 

  44. Foley, J. C., McIver, S. R. & Haydon, P. G. Gliotransmission modulates baseline mechanical nociception. Mol. Pain 7, 93 (2011). This study, together with Fujita et al. (2014) and Liu et al. (2016), provides evidence for the existence of an astrocyte-mediated pain suppression system under homeostatic conditions.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Fujita, T. et al. Neuronal transgene expression in dominant-negative SNARE mice. J. Neurosci. 34, 16594–16604 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Liu, C. C. et al. Interferon α inhibits spinal cord synaptic and nociceptive transmission via neuronal–glial interactions. Sci. Rep. 6, 34356 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ohara, P. T., Vit, J. P., Bhargava, A. & Jasmin, L. Evidence for a role of connexin 43 in trigeminal pain using RNA interference in vivo. J. Neurophysiol. 100, 3064–3073 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Morioka, N. et al. Downregulation of spinal astrocytic connexin43 leads to upregulation of interleukin-6 and cyclooxygenase-2 and mechanical hypersensitivity in mice. Glia 66, 428–444 (2018).

    Article  PubMed  Google Scholar 

  49. Garrison, C. J., Dougherty, P. M., Kajander, K. C. & Carlton, S. M. Staining of glial fibrillary acidic protein (GFAP) in lumbar spinal cord increases following a sciatic nerve constriction injury. Brain Res. 565, 1–7 (1991). This study provides early evidence for a possible involvement of astrocytes in pain.

    Article  CAS  PubMed  Google Scholar 

  50. Garrison, C. J., Dougherty, P. M. & Carlton, S. M. GFAP expression in lumbar spinal cord of naive and neuropathic rats treated with MK-801. Exp. Neurol. 129, 237–243 (1994).

    Article  CAS  PubMed  Google Scholar 

  51. Nesic, O. et al. Transcriptional profiling of spinal cord injury-induced central neuropathic pain. J. Neurochem. 95, 998–1014 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Zhuang, Z. Y. et al. A peptide c-Jun N-terminal kinase (JNK) inhibitor blocks mechanical allodynia after spinal nerve ligation: respective roles of JNK activation in primary sensory neurons and spinal astrocytes for neuropathic pain development and maintenance. J. Neurosci. 26, 3551–3560 (2006). This study demonstrates that astrocytic JNK signalling contributes to the maintenance of nerve injury-induced neuropathic pain.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Song, P. & Zhao, Z. Q. The involvement of glial cells in the development of morphine tolerance. Neurosci. Res. 39, 281–286 (2001). This study provides early evidence for an involvement of astrocytes in chronic morphine-induced antinociceptive tolerance.

    Article  CAS  PubMed  Google Scholar 

  54. Raghavendra, V., Tanga, F. Y. & DeLeo, J. A. Complete Freunds adjuvant-induced peripheral inflammation evokes glial activation and proinflammatory cytokine expression in the CNS. Eur. J. Neurosci. 20, 467–473 (2004).

    Article  PubMed  Google Scholar 

  55. Gao, Y. J. et al. The c-Jun N-terminal kinase 1 (JNK1) in spinal astrocytes is required for the maintenance of bilateral mechanical allodynia under a persistent inflammatory pain condition. Pain 148, 309–319 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Guo, W. et al. Glial-cytokine-neuronal interactions underlying the mechanisms of persistent pain. J. Neurosci. 27, 6006–6018 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sun, S. et al. New evidence for the involvement of spinal fractalkine receptor in pain facilitation and spinal glial activation in rat model of monoarthritis. Pain 129, 64–75 (2007).

    Article  CAS  PubMed  Google Scholar 

  58. Honore, P. et al. Murine models of inflammatory, neuropathic and cancer pain each generates a unique set of neurochemical changes in the spinal cord and sensory neurons. Neurosci. 98, 585–598 (2000).

    Article  CAS  Google Scholar 

  59. Gao, Y. J. et al. Selective inhibition of JNK with a peptide inhibitor attenuates pain hypersensitivity and tumor growth in a mouse skin cancer pain model. Exp. Neurol. 219, 146–155 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zhang, H., Yoon, S. Y., Zhang, H. & Dougherty, P. M. Evidence that spinal astrocytes but not microglia contribute to the pathogenesis of Paclitaxel-induced painful neuropathy. J. Pain 13, 293–303 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Shi, Y., Gelman, B. B., Lisinicchia, J. G. & Tang, S. J. Chronic-pain-associated astrocytic reaction in the spinal cord dorsal horn of human immunodeficiency virus-infected patients. J. Neurosci. 32, 10833–10840 (2012). This study provides evidence for the persistent activation of spinal cord astrocytes in a human chronic pain condition.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Del Valle, L., Schwartzman, R. J. & Alexander, G. Spinal cord histopathological alterations in a patient with longstanding complex regional pain syndrome. Brain, Behav., Immun. 23, 85–91 (2009).

    Article  Google Scholar 

  63. Gwak, Y. S., Kang, J., Unabia, G. C. & Hulsebosch, C. E. Spatial and temporal activation of spinal glial cells: role of gliopathy in central neuropathic pain following spinal cord injury in rats. Exp. Neurol. 234, 362–372 (2012).

    Article  CAS  PubMed  Google Scholar 

  64. Tsuda, M. et al. JAK-STAT3 pathway regulates spinal astrocyte proliferation and neuropathic pain maintenance in rats. Brain 134, 1127–1139 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Kim, D. S. et al. Profiling of dynamically changed gene expression in dorsal root ganglia post peripheral nerve injury and a critical role of injury-induced glial fibrillary acidic protein in maintenance of pain behaviors [corrected]. Pain 143, 114–122 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. DeLeo, J. A., Rutkowski, M. D., Stalder, A. K. & Campbell, I. L. Transgenic expression of TNF by astrocytes increases mechanical allodynia in a mouse neuropathy model. Neuroreport 11, 599–602 (2000).

    Article  CAS  PubMed  Google Scholar 

  67. Menetski, J. et al. Mice overexpressing chemokine ligand 2 (CCL2) in astrocytes display enhanced nociceptive responses. Neuroscience 149, 706–714 (2007).

    Article  CAS  PubMed  Google Scholar 

  68. Gao, Y. J., Zhang, L. & Ji, R. R. Spinal injection of TNF-α-activated astrocytes produces persistent pain symptom mechanical allodynia by releasing monocyte chemoattractant protein-1. Glia 58, 1871–1880 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Chiang, C. Y., Sessle, B. J. & Dostrovsky, J. O. Role of astrocytes in pain. Neurochem. Res. 37, 2419-2431 (2012).

  70. Meller, S. T., Dykstra, C., Grzybycki, D., Murphy, S. & Gebhart, G. F. The possible role of glia in nociceptive processing and hyperalgesia in the spinal cord of the rat. Neuropharmacology 33, 1471–1478 (1994).

    Article  CAS  PubMed  Google Scholar 

  71. Watkins, L. R., Martin, D., Ulrich, P., Tracey, K. J. & Maier, S. F. Evidence for the involvement of spinal cord glia in subcutaneous formalin induced hyperalgesia in the rat. Pain 71, 225–235 (1997).

    Article  CAS  PubMed  Google Scholar 

  72. Ji, R. R., Kawasaki, Y., Zhuang, Z. Y., Wen, Y. R. & Decosterd, I. Possible role of spinal astrocytes in maintaining chronic pain sensitization: review of current evidence with focus on bFGF/JNK pathway. Neuron Glia Biol. 2, 259–269 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Chiang, C. Y. et al. Astroglial glutamate–glutamine shuttle is involved in central sensitization of nociceptive neurons in rat medullary dorsal horn. J. Neurosci. 27, 9068–9076 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Okada-Ogawa, A. et al. Astroglia in medullary dorsal horn (trigeminal spinal subnucleus caudalis) are involved in trigeminal neuropathic pain mechanisms. J. Neurosci. 29, 11161–11171 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Ren, K. & Dubner, R. Interactions between the immune and nervous systems in pain. Nat. Med. 16, 1267–1276 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Nam, Y. et al. Reversible induction of pain hypersensitivity following optogenetic stimulation of spinal astrocytes. Cell Rep. 17, 3049–3061 (2016). This study demonstrates that sterile astrocyte activation using optogenetic stimulation alone is sufficient to produce pain behaviours in naïve rats.

    Article  CAS  PubMed  Google Scholar 

  77. Salter, M. W. & Stevens, B. Microglia emerge as central players in brain disease. Nat. Med. 23, 1018–1027 (2017).

    Article  CAS  PubMed  Google Scholar 

  78. Inoue, K. & Tsuda, M. Microglia in neuropathic pain: cellular and molecular mechanisms and therapeutic potential. Nat. Rev. Neurosci. 19, 138–152 (2018).

    Article  CAS  PubMed  Google Scholar 

  79. Scholz, J. & Woolf, C. J. The neuropathic pain triad: neurons, immune cells and glia. Nat. Neurosci. 10, 1361–1368 (2007).

    Article  CAS  PubMed  Google Scholar 

  80. McMahon, S. B. & Malcangio, M. Current challenges in glia–pain biology. Neuron 64, 46–54 (2009).

    Article  CAS  PubMed  Google Scholar 

  81. Chen, G., Zhang, Y. Q., Qadri, Y. J., Serhan, C. N. & Ji, R. R. Microglia in pain: detrimental and protective roles in pathogenesis and resolution of pain. Neuron 100, 1292–1311 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Robinson, C. R., Zhang, H. & Dougherty, P. M. Astrocytes, but not microglia, are activated in oxaliplatin and bortezomib-induced peripheral neuropathy in the rat. Neuroscience 274, 308–317 (2014).

    Article  CAS  PubMed  Google Scholar 

  83. Luo, X. et al. Intrathecal administration of antisense oligonucleotide against p38α but not p38β MAP kinase isoform reduces neuropathic and postoperative pain and TLR4-induced pain in male mice. Brain Behav. Immun. 72, 34–44 (2018).

    Article  CAS  PubMed  Google Scholar 

  84. Taves, S. et al. Spinal inhibition of p38 MAP kinase reduces inflammatory and neuropathic pain in male but not female mice: Sex-dependent microglial signaling in the spinal cord. Brain Behav. Immun. 55, 70–81 (2016).

    Article  CAS  PubMed  Google Scholar 

  85. Sorge, R. E. et al. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat. Neurosci. 18, 1081–1083 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Chen, G., Luo, X., Qadri, M. Y., Berta, T. & Ji, R. R. Sex-dependent glial signaling in pathological pain: distinct roles of spinal microglia and astrocytes. Neurosci. Bull. 34, 98–108 (2018).

    Article  CAS  PubMed  Google Scholar 

  87. Yang, Y. et al. Delayed activation of spinal microglia contributes to the maintenance of bone cancer pain in female Wistar rats via P2X7 receptor and IL-18. J. Neurosci. 35, 7950–7963 (2015).

    Article  CAS  PubMed  Google Scholar 

  88. Pappalardo, L. W. et al. Nav1.5 in astrocytes plays a sex-specific role in clinical outcomes in a mouse model of multiple sclerosis. Glia 66, 2174–2187 (2018).

    Article  PubMed  Google Scholar 

  89. Zhang, J. & De Koninck, Y. Spatial and temporal relationship between monocyte chemoattractant protein-1 expression and spinal glial activation following peripheral nerve injury. J. Neurochem. 97, 772–783 (2006).

    Article  CAS  PubMed  Google Scholar 

  90. Raghavendra, V., Tanga, F. & DeLeo, J. A. Inhibition of microglial activation attenuates the development but not existing hypersensitivity in a rat model of neuropathy. J. Pharmacol. Exp. Ther. 306, 624–630 (2003).

    Article  CAS  PubMed  Google Scholar 

  91. Chen, G. et al. Connexin-43 induces chemokine release from spinal cord astrocytes to maintain late-phase neuropathic pain in mice. Brain 137, 2193–2209 (2014). This study demonstrates that astrocytes upregulate connexin 43 following nerve injury, driving the release of CXCL1, which induces central sensitization in nociceptive neurons to maintain neuropathic pain.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Obata, K. & Noguchi, K. MAPK activation in nociceptive neurons and pain hypersensitivity. Life Sci. 74, 2643–2653 (2004).

    Article  CAS  PubMed  Google Scholar 

  93. Ji, R. R., Gereau, R. W., Malcangio, M. & Strichartz, G. R. MAP kinase and pain. Brain Res. Rev. 60, 135–148 (2009).

    Article  CAS  PubMed  Google Scholar 

  94. Jin, S. X., Zhuang, Z. Y., Woolf, C. J. & Ji, R. R. p38 mitogen-activated protein kinase is activated after a spinal nerve ligation in spinal cord microglia and dorsal root ganglion neurons and contributes to the generation of neuropathic pain. J. Neurosci. 23, 4017–4022 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Tsuda, M., Mizokoshi, A., Shigemoto-Mogami, Y., Koizumi, S. & Inoue, K. Activation of p38 mitogen-activated protein kinase in spinal hyperactive microglia contributes to pain hypersensitivity following peripheral nerve injury. Glia 45, 89–95 (2004).

    Article  PubMed  Google Scholar 

  96. Katsura, H. et al. Transforming growth factor-activated kinase 1 induced in spinal astrocytes contributes to mechanical hypersensitivity after nerve injury. Glia 56, 723–733 (2008).

    Article  PubMed  Google Scholar 

  97. Mei, X. P. et al. Inhibition of spinal astrocytic c-Jun N-terminal kinase (JNK) activation correlates with the analgesic effects of ketamine in neuropathic pain. J. Neuroinflammation 8, 6 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Zhuang, Z. Y., Gerner, P., Woolf, C. J. & Ji, R. R. ERK is sequentially activated in neurons, microglia, and astrocytes by spinal nerve ligation and contributes to mechanical allodynia in this neuropathic pain model. Pain 114, 149–159 (2005).

    Article  PubMed  Google Scholar 

  99. Ji, R. R., Xu, Z. Z., Wang, X. & Lo, E. H. Matrix metalloprotease regulation of neuropathic pain. Trends Pharmacol. Sci. 30, 336–340 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Kawasaki, Y. et al. Distinct roles of matrix metalloproteases in the early- and late-phase development of neuropathic pain. Nat. Med. 14, 331–336 (2008). This paper demonstrates that MMP2 upregulation in spinal astrocytes after nerve injury contributes to neuropathic pain maintenance.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Kozai, T. et al. Tissue type plasminogen activator induced in rat dorsal horn astrocytes contributes to mechanical hypersensitivity following dorsal root injury. Glia 55, 595–603 (2007).

    Article  PubMed  Google Scholar 

  102. Jiang, L. et al. Selective suppression of the JNK–MMP2/9 signal pathway by tetramethylpyrazine attenuates neuropathic pain in rats. J. Neuroinflammation 14, 174 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Sung, B., Lim, G. & Mao, J. Altered expression and uptake activity of spinal glutamate transporters after nerve injury contribute to the pathogenesis of neuropathic pain in rats. J. Neurosci. 23, 2899–2910 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Xin, W. J., Weng, H. R. & Dougherty, P. M. Plasticity in expression of the glutamate transporters GLT-1 and GLAST in spinal dorsal horn glial cells following partial sciatic nerve ligation. Mol. Pain 5, 15 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Liaw, W. J. et al. Spinal glutamate uptake is critical for maintaining normal sensory transmission in rat spinal cord. Pain 115, 60–70 (2005).

    Article  CAS  PubMed  Google Scholar 

  106. Weng, H. R., Chen, J. H. & Cata, J. P. Inhibition of glutamate uptake in the spinal cord induces hyperalgesia and increased responses of spinal dorsal horn neurons to peripheral afferent stimulation. Neuroscience 138, 1351–1360 (2006).

    Article  CAS  PubMed  Google Scholar 

  107. Falnikar, A., Hala, T. J., Poulsen, D. J. & Lepore, A. C. GLT1 overexpression reverses established neuropathic pain-related behavior and attenuates chronic dorsal horn neuron activation following cervical spinal cord injury. Glia 64, 396–406 (2016).

    Article  PubMed  Google Scholar 

  108. Tackley, G. et al. Chronic neuropathic pain severity is determined by lesion level in aquaporin 4-antibody-positive myelitis. J. Neurol. Neurosurg. Psychiatry 88, 165–169 (2017).

    Article  PubMed  Google Scholar 

  109. Bao, F., Chen, M., Zhang, Y. & Zhao, Z. Hypoalgesia in mice lacking aquaporin-4 water channels. Brain Res. Bull. 83, 298–303 (2010).

    Article  CAS  PubMed  Google Scholar 

  110. Bradl, M. et al. Pain in neuromyelitis optica — prevalence, pathogenesis and therapy. Nat. Rev. Neurol. 10, 529–536 (2014).

    Article  PubMed  Google Scholar 

  111. Kang, J. et al. Connexin 43 hemichannels are permeable to ATP. J. Neurosci. 28, 4702–4711 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Chen, M. J. et al. Astrocytic CX43 hemichannels and gap junctions play a crucial role in development of chronic neuropathic pain following spinal cord injury. Glia 60, 1660–1670 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  113. Spataro, L. E. et al. Spinal gap junctions: potential involvement in pain facilitation. J. Pain 5, 392–405 (2004).

    Article  CAS  PubMed  Google Scholar 

  114. Bennett, M. V., Contreras, J. E., Bukauskas, F. F. & Saez, J. C. New roles for astrocytes: gap junction hemichannels have something to communicate. Trends Neurosci. 26, 610–617 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Cronin, M., Anderson, P. N., Cook, J. E., Green, C. R. & Becker, D. L. Blocking connexin43 expression reduces inflammation and improves functional recovery after spinal cord injury. Mol. Cell Neurosci. 39, 152–160 (2008).

    Article  CAS  PubMed  Google Scholar 

  116. Huang, C. et al. Critical role of connexin 43 in secondary expansion of traumatic spinal cord injury. J. Neurosci. 32, 3333–3338 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Garre, J. M. et al. FGF-1 induces ATP release from spinal astrocytes in culture and opens pannexin and connexin hemichannels. Proc. Natl Acad. Sci. USA 107, 22659–22664 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  118. Koyanagi, S. et al. Glucocorticoid regulation of ATP release from spinal astrocytes underlies diurnal exacerbation of neuropathic mechanical allodynia. Nat. Commun. 7, 13102 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Mousseau, M. et al. Microglial pannexin-1 channel activation is a spinal determinant of joint pain. Sci. Adv. 4, eaas9846 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Gao, Y. J. et al. JNK-induced MCP-1 production in spinal cord astrocytes contributes to central sensitization and neuropathic pain. J. Neurosci. 29, 4096–4108 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Zhang, Z. J. et al. Chemokine CCL2 and its receptor CCR2 in the medullary dorsal horn are involved in trigeminal neuropathic pain. J. Neuroinflammation 9, 136 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Jiang, B. C. et al. CXCL13 drives spinal astrocyte activation and neuropathic pain via CXCR5. J. Clin. Invest. 126, 745–761 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Yuan, S., Shi, Y. & Tang, S. J. Wnt signaling in the pathogenesis of multiple sclerosis-associated chronic pain. J. Neuroimmune Pharmacol. 7, 904–913 (2012).

    Article  PubMed  Google Scholar 

  124. Zhang, Y. K. et al. WNT signaling underlies the pathogenesis of neuropathic pain in rodents. J. Clin. Invest. 123, 2268–2286 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Liu, S. et al. Wnt/Ryk signaling contributes to neuropathic pain by regulating sensory neuron excitability and spinal synaptic plasticity in rats. Pain 156, 2572–2584 (2015).

    Article  CAS  PubMed  Google Scholar 

  126. Patti, G. J. et al. Metabolomics implicates altered sphingolipids in chronic pain of neuropathic origin. Nat. Chem. Biol. 8, 232–234 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Stockstill, K. et al. Dysregulation of sphingolipid metabolism contributes to bortezomib-induced neuropathic pain. J. Exp. Med. 215, 1301–1313 (2018). This study demonstrates that chemotherapy-induced sphingolipid metabolites activate astrocytes to release pro-inflammatory mediators and drive neuropathic pain.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Zarpelon, A. C. et al. Spinal cord oligodendrocyte-derived alarmin IL-33 mediates neuropathic pain. FASEB J. 30, 54–65 (2016).

    Article  CAS  PubMed  Google Scholar 

  129. Liu, S. et al. Spinal IL-33/ST2 signaling contributes to neuropathic pain via neuronal camkii-creb and astroglial jak2-stat3 cascades in mice. Anesthesiology 123, 1154–1169 (2015).

    Article  CAS  PubMed  Google Scholar 

  130. Suter, M. R., Wen, Y. R., Decosterd, I. & Ji, R. R. Do glial cells control pain? Neuron Glia Biol. 3, 255–268 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  131. Miyoshi, K., Obata, K., Kondo, T., Okamura, H. & Noguchi, K. Interleukin-18-mediated microglia/astrocyte interaction in the spinal cord enhances neuropathic pain processing after nerve injury. J. Neurosci. 28, 12775–12787 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Lu, Y. et al. TRAF6 upregulation in spinal astrocytes maintains neuropathic pain by integrating TNF-α and IL-1β signaling. Pain 155, 2618–2629 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. 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 

  134. Tanga, F. Y., Nutile-McMenemy, N. & DeLeo, J. A. The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proc. Natl Acad. Sci. USA 102, 5856–5861 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Li, Y. et al. Toll-like receptor 4 signaling contributes to Paclitaxel-induced peripheral neuropathy. J. Pain 15, 712–725 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Sorge, R. E. et al. Spinal cord Toll-like receptor 4 mediates inflammatory and neuropathic hypersensitivity in male but not female mice. J. Neurosci. 31, 15450–15454 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Bartley, E. J. & Fillingim, R. B. Sex differences in pain: a brief review of clinical and experimental findings. Br. J. Anaesth. 111, 52–58 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Woolf, C. J. Evidence for a central component of post-injury pain hypersensitivity. Nature 306, 686–688 (1983).

    Article  CAS  PubMed  Google Scholar 

  139. Ji, R. R., Kohno, T., Moore, K. A. & Woolf, C. J. Central sensitization and LTP: do pain and memory share similar mechanisms? Trends Neurosci. 26, 696–705 (2003).

    Article  CAS  PubMed  Google Scholar 

  140. Woolf, C. J. Central sensitization: implications for the diagnosis and treatment of pain. Pain 152, S2–S15 (2011).

    Article  PubMed  Google Scholar 

  141. Woolf, C. J. & Salter, M. W. Neuronal plasticity: increasing the gain in pain. Science 288, 1765–1769 (2000).

    Article  CAS  PubMed  Google Scholar 

  142. Latremoliere, A. & Woolf, C. J. Central sensitization: a generator of pain hypersensitivity by central neural plasticity. J. Pain 10, 895–926 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  143. Gao, Y. J. & Ji, R. R. Chemokines, neuronal–glial interactions, and central processing of neuropathic pain. Pharmacol. Ther. 126, 56–68 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. White, F. A. et al. Excitatory monocyte chemoattractant protein-1 signaling is up-regulated in sensory neurons after chronic compression of the dorsal root ganglion. Proc. Natl Acad. Sci. USA 102, 14092–14097 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Xie, R. G. et al. Spinal CCL2 promotes central sensitization, long-term potentiation, and inflammatory pain via CCR2: further insights into molecular, synaptic, and cellular mechanisms. Neurosci. Bull. 34, 13–21 (2018).

    Article  CAS  PubMed  Google Scholar 

  146. Gosselin, R. D. et al. Constitutive expression of CCR2 chemokine receptor and inhibition by MCP-1/CCL2 of GABA-induced currents in spinal cord neurones. J. Neurochem. 95, 1023–1034 (2005).

    Article  CAS  PubMed  Google Scholar 

  147. Zhang, Z. J., Cao, D. L., Zhang, X., Ji, R. R. & Gao, Y. J. Chemokine contribution to neuropathic pain: respective induction of CXCL1 and CXCR2 in spinal cord astrocytes and neurons. Pain 154, 2185–2197 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Sommer, C., Schafers, M., Marziniak, M. & Toyka, K. V. Etanercept reduces hyperalgesia in experimental painful neuropathy. J. Peripher. Nerv. Syst. 6, 67–72 (2001).

    Article  CAS  PubMed  Google Scholar 

  149. Milligan, E. D. & Watkins, L. R. Pathological and protective roles of glia in chronic pain. Nat. Rev. Neurosci. 10, 23–36 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Ren, K. & Torres, R. Role of interleukin-1β during pain and inflammation. Brain Res. Rev. 60, 57–64 (2009).

    Article  CAS  PubMed  Google Scholar 

  151. Zhang, R. X. et al. IL-1ra alleviates inflammatory hyperalgesia through preventing phosphorylation of NMDA receptor NR-1 subunit in rats. Pain 135, 232–239 (2008).

    Article  CAS  PubMed  Google Scholar 

  152. Li, W. W. et al. The NALP1 inflammasome controls cytokine production and nociception in a rat fracture model of complex regional pain syndrome. Pain 147, 277–286 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Kawasaki, Y., Zhang, L., Cheng, J. K. & Ji, R. R. Cytokine mechanisms of central sensitization: distinct and overlapping role of interleukin-1β, interleukin-6, and tumor necrosis factor-α in regulating synaptic and neuronal activity in the superficial spinal cord. J. Neurosci. 28, 5189–5194 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. 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 

  155. Zhang, H., Nei, H. & Dougherty, P. M. A p38 mitogen-activated protein kinase-dependent mechanism of disinhibition in spinal synaptic transmission induced by tumor necrosis factor-α. J. Neurosci. 30, 12844–12855 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Sandkuhler, J. Learning and memory in pain pathways. Pain 88, 113–118 (2000).

    Article  CAS  PubMed  Google Scholar 

  157. Sandkuhler, J. Models and mechanisms of hyperalgesia and allodynia. Physiol. Rev. 89, 707–758 (2009).

    Article  CAS  PubMed  Google Scholar 

  158. Liu, Y. L. et al. Tumor necrosis factor-α induces long-term potentiation of C-fiber evoked field potentials in spinal dorsal horn in rats with nerve injury: the role of NF-κB, JNK and p38 MAPK. Neuropharmacology 52, 708–715 (2007).

    Article  CAS  PubMed  Google Scholar 

  159. 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 

  160. Gruber-Schoffnegger, D. et al. Induction of thermal hyperalgesia and synaptic long-term potentiation in the spinal cord lamina I by TNF-α and IL-1β is mediated by glial cells. J. Neurosci. 33, 6540–6551 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Chirila, A. M. et al. Long-term potentiation of glycinergic synapses triggered by interleukin 1β. Proc. Natl Acad. Sci. USA 111, 8263–8268 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Kronschlager, M. T. et al. Gliogenic LTP spreads widely in nociceptive pathways. Science 354, 1144–1148 (2016). This study identifies a diffusible form of LTP, induced by small, glia-derived mediators, which can be carried through the CSF to distant synapses.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Xie, Y. F. et al. Involvement of glia in central sensitization in trigeminal subnucleus caudalis (medullary dorsal horn). Brain Behav. Immun. 21, 634–641 (2007).

    Article  CAS  PubMed  Google Scholar 

  164. Kobayashi, A. et al. Mechanisms involved in extraterritorial facial pain following cervical spinal nerve injury in rats. Mol. Pain 7, 12 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Mah, W. et al. A role for the purinergic receptor P2X3 in astrocytes in the mechanism of craniofacial neuropathic pain. Sci. Rep. 7, 13627 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Alshelh, Z. et al. Chronic neuropathic pain: it’s about the rhythm. J. Neurosci. 36, 1008–1018 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Wei, F., Guo, W., Zou, S., Ren, K. & Dubner, R. Supraspinal glial–neuronal interactions contribute to descending pain facilitation. J. Neurosci. 28, 10482–10495 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Ni, H. D. et al. Astrocyte activation in the periaqueductal gray promotes descending facilitation to cancer-induced bone pain through the JNK MAPK signaling pathway. Mol. Pain 15, 1744806919831909 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  169. Chen, F. L. et al. Activation of astrocytes in the anterior cingulate cortex contributes to the affective component of pain in an inflammatory pain model. Brain Res. Bull. 87, 60–66 (2012).

    Article  CAS  PubMed  Google Scholar 

  170. Kim, S. K. et al. Cortical astrocytes rewire somatosensory cortical circuits for peripheral neuropathic pain. J. Clin. Invest. 126, 1983–1997 (2016). This paper provides evidence that cortical astrocytes are activated following nerve injury, driving local synaptic plasticity and neuropathic pain via astrocytic TSP1.

    Article  PubMed  PubMed Central  Google Scholar 

  171. Loggia, M. L. et al. Evidence for brain glial activation in chronic pain patients. Brain 138, 604–615 (2015). This neuroimaging study demonstrates that glial activation is observed in the pain-processing brain regions of patients with chronic lower back pain.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Takata, N. et al. Optogenetic astrocyte activation evokes BOLD fMRI response with oxygen consumption without neuronal activity modulation. Glia 66, 2013–2023 (2018).

    Article  PubMed  Google Scholar 

  173. LaMotte, R. H., Dong, X. & Ringkamp, M. Sensory neurons and circuits mediating itch. Nat. Rev. Neurosci. 15, 19–31 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Moser, H. R. & Giesler, G. J. Jr. Itch and analgesia resulting from intrathecal application of morphine: contrasting effects on different populations of trigeminothalamic tract neurons. J. Neurosci. 33, 6093–6101 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Lee, H. & Ko, M. C. Distinct functions of opioid-related peptides and gastrin-releasing peptide in regulating itch and pain in the spinal cord of primates. Sci. Rep. 5, 11676 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Steinhoff, M. et al. Neurophysiological, neuroimmunological, and neuroendocrine basis of pruritus. J. Invest. Dermatol. 126, 1705–1718 (2006).

    Article  CAS  PubMed  Google Scholar 

  177. Dong, X. & Dong, X. Peripheral and central mechanisms of itch. Neuron 98, 482–494 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Sun, Y. G. & Chen, Z. F. A gastrin-releasing peptide receptor mediates the itch sensation in the spinal cord. Nature 448, 700–703 (2007).

    Article  CAS  PubMed  Google Scholar 

  179. Duan, B., Cheng, L. & Ma, Q. Spinal circuits transmitting mechanical pain and itch. Neurosci. Bull. 34, 186–193 (2018).

    Article  PubMed  Google Scholar 

  180. Ross, S. E. Pain and itch: insights into the neural circuits of aversive somatosensation in health and disease. Curr. Opin. Neurobiol. 21, 880–887 (2011).

    Article  CAS  PubMed  Google Scholar 

  181. Liu, Y. et al. VGLUT2-dependent glutamate release from nociceptors is required to sense pain and suppress itch. Neuron 68, 543–556 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Liu, T. & Ji, R.-R. in Itch: Mechanisms and Treatment (eds Carstens, E. & Akiyama, T.) 257-270 (CRC Press, 2014).

  183. Yosipovitch, G. & Bernhard, J. D. Chronic pruritus. N. Engl. J. Med. 368, 1625–1634 (2013).

    Article  CAS  PubMed  Google Scholar 

  184. Han, Q. et al. miRNA-711 binds and activates TRPA1 extracellularly to evoke acute and chronic pruritus. Neuron 99, 449–463 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Liu, T. & Ji, R. R. New insights into the mechanisms of itch: are pain and itch controlled by distinct mechanisms? Pflug. Arch. 465, 1671–1685 (2013).

    Article  CAS  Google Scholar 

  186. Ross, S. E. et al. Loss of inhibitory interneurons in the dorsal spinal cord and elevated itch in Bhlhb5 mutant mice. Neuron 65, 886–898 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Green, D. & Dong, X. Supporting itch: a new role for astrocytes in chronic itch. Nat. Med. 21, 841–842 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Tsuda, M. Astrocytes in the spinal dorsal horn and chronic itch. Neurosci. Res. 126, 9–14 (2018).

    Article  CAS  PubMed  Google Scholar 

  189. Du, L. et al. Spinal IL-33/ST2 signaling mediates chronic itch in mice through the astrocytic JAK2-STAT3 cascade. Glia 67, 1680–1693 (2019).

    PubMed  Google Scholar 

  190. Shiratori-Hayashi, M. et al. STAT3-dependent reactive astrogliosis in the spinal dorsal horn underlies chronic itch. Nat. Med. 21, 927–931 (2015).

    Article  CAS  PubMed  Google Scholar 

  191. Liu, T. et al. Toll-like receptor 4 contributes to chronic itch, alloknesis, and spinal astrocyte activation in male mice. Pain 157, 806–817 (2016). This study demonstrates that spinal astrocytes play an active role in regulating chronic but not acute itch.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Akiyama, T. et al. A central role for spinal dorsal horn neurons that express neurokinin-1 receptors in chronic itch. Pain 156, 1240–1246 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Bourane, S. et al. Gate control of mechanical itch by a subpopulation of spinal cord interneurons. Science 350, 550–554 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Miao, X. et al. TNF-α/TNFR1 signaling is required for the full expression of acute and chronic itch in mice via peripheral and central mechanisms. Neurosci. Bull. 34, 42–53 (2018).

    Article  CAS  PubMed  Google Scholar 

  195. Jing, P. B. et al. Chemokine receptor CXCR3 in the spinal cord contributes to chronic itch in mice. Neurosci. Bull. 34, 54–63 (2018).

    Article  CAS  PubMed  Google Scholar 

  196. Wilson, S. R. et al. The epithelial cell-derived atopic dermatitis cytokine TSLP activates neurons to induce itch. Cell 155, 285–295 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Liu, B. et al. IL-33/ST2 signaling excites sensory neurons and mediates itch response in a mouse model of poison ivy contact allergy. Proc. Natl Acad. Sci. USA 113, E7572–E7579 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Paus, R., Schmelz, M., Biro, T. & Steinhoff, M. Frontiers in pruritus research: scratching the brain for more effective itch therapy. J. Clin. Invest. 116, 1174–1186 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Global Industry Analysts, Inc. Pain Management — A Global Strategic Business Report (GIA, 2011).

  200. Gereau, R. W. T. et al. A pain research agenda for the 21st century. J. Pain 15, 1203–1214 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  201. Ringelstein, M. et al. Long-term therapy with interleukin 6 receptor blockade in highly active neuromyelitis optica spectrum disorder. JAMA Neurol. 72, 756–763 (2015).

    Article  PubMed  Google Scholar 

  202. Sommer, C. [Animal studies on neuropathic pain: the role of cytokines and cytokine receptors in pathogenesis and therapy]. Schmerz 13, 315–323 (1999).

    Article  CAS  PubMed  Google Scholar 

  203. Sato, K. L., Johanek, L. M., Sanada, L. S. & Sluka, K. A. Spinal cord stimulation reduces mechanical hyperalgesia and glial cell activation in animals with neuropathic pain. Anesth. Analg. 118, 464–472 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Chen, G., Park, C. K., Xie, R. G. & Ji, R. R. Intrathecal bone marrow stromal cells inhibit neuropathic pain via TGF-β secretion. J. Clin. Invest. 125, 3226–3240 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  205. Saunders, A. et al. Molecular diversity and specializations among the cells of the adult mouse brain. Cell 174, 1015–1030.e1016 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Zeisel, A. et al. Molecular architecture of the mouse nervous system. Cell 174, 999–1014.e1022 (2018). This system-wide single-cell RNA-sequencing study demonstrates the existence of at least seven unique populations of astrocytes in the brain.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Liddelow, S. A. et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487 (2017). This study demonstrates that microglia, via the release of IL-1α, TNF and C1q, promote the acquisition of a pro-inflammatory ‘A1’ astrocyte phenotype.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Nahrendorf, M. & Swirski, F. K. Abandoning M1/M2 for a network model of macrophage function. Circ. Res. 119, 414–417 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Smith, M. T. & Haythornthwaite, J. A. How do sleep disturbance and chronic pain inter-relate? Insights from the longitudinal and cognitive–behavioral clinical trials literature. Sleep Med. Rev. 8, 119–132 (2004).

    Article  PubMed  Google Scholar 

  210. Xie, L. et al. Sleep drives metabolite clearance from the adult brain. Science 342, 373–377 (2013). This study demonstrates that enhanced glymphatic flow through channels formed by the perivascular endfeet of astrocytes drives the removal of proteinaceous waste from the extracellular space during sleep.

    Article  CAS  PubMed  Google Scholar 

  211. Plog, B. A. & Nedergaard, M. The glymphatic system in central nervous system health and disease: past, present, and future. Annu. Rev. Pathol. 13, 379–394 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Louveau, A. et al. Understanding the functions and relationships of the glymphatic system and meningeal lymphatics. J. Clin. Invest. 127, 3210–3219 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  213. Ringstad, G., Vatnehol, S. A. S. & Eide, P. K. Glymphatic MRI in idiopathic normal pressure hydrocephalus. Brain 140, 2691–2705 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  214. Myllyla, T. et al. Assessment of the dynamics of human glymphatic system by near-infrared spectroscopy. J. Biophotonics 11, e201700123 (2018).

    Article  PubMed  Google Scholar 

  215. von Holstein-Rathlou, S., Petersen, N. C. & Nedergaard, M. Voluntary running enhances glymphatic influx in awake behaving, young mice. Neurosci. Lett. 662, 253–258 (2018).

    Article  CAS  Google Scholar 

  216. Kimura, M. et al. Impaired pain-evoked analgesia after nerve injury in rats reflects altered glutamate regulation in the locus coeruleus. Anesthesiology 123, 899–908 (2015).

    Article  CAS  PubMed  Google Scholar 

  217. Jensen, T. S. et al. A new definition of neuropathic pain. Pain 152, 2204–2205 (2011).

    Article  PubMed  Google Scholar 

  218. van, H. O., Austin, S. K., Khan, R. A., Smith, B. H. & Torrance, N. Neuropathic pain in the general population: a systematic review of epidemiological studies. Pain 155, 654–662 (2013).

    Google Scholar 

  219. Calvo, M., Dawes, J. M. & Bennett, D. L. The role of the immune system in the generation of neuropathic pain. Lancet Neurol. 11, 629–642 (2012).

    Article  CAS  PubMed  Google Scholar 

  220. 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 

  221. Kim, S. H. & Chung, J. M. An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain 50, 355–363 (1992).

    Article  CAS  Google Scholar 

  222. Seltzer, Z., Dubner, R. & Shir, Y. A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 43, 205–218 (1990).

    Article  CAS  PubMed  Google Scholar 

  223. Decosterd, I. & Woolf, C. J. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain 87, 149–158 (2000).

    Article  CAS  PubMed  Google Scholar 

  224. Flatters, S. J. & Bennett, G. J. Ethosuximide reverses paclitaxel- and vincristine-induced painful peripheral neuropathy. Pain 109, 150–161 (2004).

    Article  CAS  PubMed  Google Scholar 

  225. Tsuda, M. et al. P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature 424, 778–783 (2003).

    Article  CAS  PubMed  Google Scholar 

  226. Coull, J. A. et al. BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain. Nature 438, 1017–1021 (2005).

    Article  CAS  PubMed  Google Scholar 

  227. Clark, A. K. et al. P2X7-dependent release of interleukin-1β and nociception in the spinal cord following lipopolysaccharide. J. Neurosci. 30, 573–582 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Hochstim, C., Deneen, B., Lukaszewicz, A., Zhou, Q. & Anderson, D. J. Identification of positionally distinct astrocyte subtypes whose identities are specified by a homeodomain code. Cell 133, 510–522 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Tsai, H. H. et al. Regional astrocyte allocation regulates CNS synaptogenesis and repair. Science 337, 358–362 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Hammond, T. R. et al. Single-cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes. Immunity 50, 253–271.e256 (2019).

    Article  CAS  PubMed  Google Scholar 

  231. Bannenberg, G. & Serhan, C. N. Specialized pro-resolving lipid mediators in the inflammatory response: an update. Biochim. Biophys. Acta 1801, 1260–1273 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Abdelmoaty, S. et al. Spinal actions of lipoxin A4 and 17(R)-resolvin D1 attenuate inflammation-induced mechanical hypersensitivity and spinal TNF release. PLOS ONE 8, e75543 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Singh, S. K. et al. Astrocytes assemble thalamocortical synapses by bridging NRX1α and NL1 via hevin. Cell 164, 183–196 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Mishra, S. K. & Hoon, M. A. The cells and circuitry for itch responses in mice. Science 340, 968–971 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Alexandre, C. et al. Decreased alertness due to sleep loss increases pain sensitivity in mice. Nat. Med. 23, 768–774 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Kim, D. S. et al. Thrombospondin-4 contributes to spinal sensitization and neuropathic pain states. J. Neurosci. 32, 8977–8987 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH grants DE17794, to R.-R.J., and DE22743, to R.-R.J. and M.N. M.N is also supported by the Lundbeck Foundation. C.R.D. is supported by a John J. Bonica Trainee Fellowship from the International Association for the Study of Pain and by NIH grant T32 GM08600.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding authors

Correspondence to Ru-Rong Ji or Maiken Nedergaard.

Ethics declarations

Competing Interests

The authors declare that there are no competing interests.

Additional information

Publisher’s note

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

Glossary

Neuroinflammation

A localized form of inflammation occurring in the peripheral nervous system and/or CNS.

Central sensitization

Forms of neuronal and/or synaptic plasticity characterized by increased responsiveness of nociceptive neurons in the CNS to their normal input.

Hemichannels

Assemblies composed of six connexin proteins, which form a pore that allows for the bidirectional flow of ions and signalling molecules.

Glymphatic system

A waste clearance system in the CNS that utilizes a unique network of perivascular tunnels formed by astrocytes.

Cytokines

A large and diverse class of small (<30 kDa) proteins, glycoproteins and peptides that are secreted by cells and exert specific biological functions, including pain modulations.

Satellite glial cells

A cell type in the peripheral nervous system that is roughly equivalent in function to astrocytes in the CNS.

Allodynia

Pain in response to a stimulus that does not normally provoke pain. Mechanical allodynia is a cardinal feature of chronic pain.

Hyperalgesia

Increased pain in response to a stimulus that ordinarily provokes pain.

Optogenetic

The use of light to activate or inhibit genetically encoded ion channels expressed within a defined cell type of interest.

Chemokines

A specific family of immunomodulatory cytokines named for their ability to induce directed chemotaxis of immune effector cells.

Analgesia

The absence of pain in response to stimulation that would normally be painful.

Inflammasome

A multimeric intracellular signalling complex responsible for the detection of pathogenic microorganisms and host-derived stressors, leading to the activation of caspase-1 and the induction of inflammation.

Nociceptors

Specialized neurons of the somatosensory nervous system that are capable of transducing nociceptive stimuli.

Immunotherapeutic

A therapeutic agent used to treat a disease by activating or suppressing the immune system.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ji, RR., Donnelly, C.R. & Nedergaard, M. Astrocytes in chronic pain and itch. Nat Rev Neurosci 20, 667–685 (2019). https://doi.org/10.1038/s41583-019-0218-1

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41583-019-0218-1

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