1932

Abstract

Itch is a unique sensation that helps organisms scratch away external threats; scratching itself induces an immune response that can contribute to more itchiness. Itch is induced chemically in the peripheral nervous system via a wide array of receptors. Given the superficial localization of itch neuron terminals, cells that dwell close to the skin contribute significantly to itch. Certain mechanical stimuli mediated by recently discovered circuits also contribute to the itch sensation. Ultimately, in the spinal cord, and likely in the brain, circuits that mediate touch, pain, and itch engage in cross modulation. Much of itch perception is still a mystery, but we present in this review the known ligands and receptors associated with itch. We also describe experiments and findings from investigations into the spinal and supraspinal circuitry responsible for the sensation of itch.

Keyword(s): brainDRGitchpruritusskinspinal cord
Loading

Article metrics loading...

/content/journals/10.1146/annurev-neuro-083019-024537
2020-07-08
2024-04-25
Loading full text...

Full text loading...

/deliver/fulltext/neuro/43/1/annurev-neuro-083019-024537.html?itemId=/content/journals/10.1146/annurev-neuro-083019-024537&mimeType=html&fmt=ahah

Literature Cited

  1. Akiyama T, Carstens MI, Carstens E 2011. Transmitters and pathways mediating inhibition of spinal itch-signaling neurons by scratching and other counterstimuli. PLOS ONE 6:7e22665
    [Google Scholar]
  2. Akiyama T, Nguyen T, Curtis E, Nishida K, Devireddy J et al. 2015. A central role for spinal dorsal horn neurons that express neurokinin-1 receptors in chronic itch. Pain 156:1240–46
    [Google Scholar]
  3. Alemi F, Kwon E, Poole DP, Lieu T, Lyo V et al. 2013. The TGR5 receptor mediates bile acid–induced itch and analgesia. J. Clin. Investig. 123:1513–30
    [Google Scholar]
  4. Andoh T, Nagasawa T, Satoh M, Kuraishi Y 1998. Substance P induction of itch-associated response mediated by cutaneous NK1 tachykinin receptors in mice. J. Pharmacol. Exp. Ther. 286:1140–45
    [Google Scholar]
  5. Arai I, Tsuji M, Takeda H, Akiyama N, Saito S 2013. A single dose of interleukin-31 (IL-31) causes continuous itch-associated scratching behaviour in mice. Exp. Dermatol. 22:656–81
    [Google Scholar]
  6. Barr TP, Garzia C, Guha S, Fletcher EK, Nguyen N et al. 2019. PAR2 pepducin-based suppression of inflammation and itch in atopic dermatitis models. J. Investig. Dermatol. 139:412–21
    [Google Scholar]
  7. Bell JK, McQueen DS, Rees JL 2004. Involvement of histamine H4 and H1 receptors in scratching induced by histamine receptor agonists in BalbC mice. Br. J. Pharmacol. 142:374–80
    [Google Scholar]
  8. Bourane S, Duan B, Koch SC, Dalet A, Britz O et al. 2015. Gate control of mechanical itch by a subpopulation of spinal cord interneurons. Science 350:550–54
    [Google Scholar]
  9. Cevikbas F, Wang X, Akiyama T, Kempkes C, Savinko T et al. 2014. A sensory neuron-expressed IL-31 receptor mediates T helper cell–dependent itch: involvement of TRPV1 and TRPA1. J. Allergy Clin. Immunol. 133:448–60
    [Google Scholar]
  10. Chan LS, Robinson N, Xu L 2001. Expression of interleukin-4 in the epidermis of transgenic mice results in a pruritic inflammatory skin disease: an experimental animal model to study atopic dermatitis. J. Investig. Dermatol. 117:977–83
    [Google Scholar]
  11. Chen Y, Fang Q, Wang Z, Zhang J, MacLeod A et al. 2016. Transient receptor potential vanilloid 4 ion channel functions as a pruriceptor in epidermal keratinocytes to evoke histaminergic itch. J. Biol. Chem. 291:10252–62
    [Google Scholar]
  12. Christensen AJ, Iyer SM, Francois A, Vyas S, Ramakrishnan C et al. 2016. In vivo interrogation of spinal mechanosensory circuits. Cell Rep 17:1699–710
    [Google Scholar]
  13. Chung K, Pitcher T, Grant AD, Hewitt E, Lindstrom E et al. 2019. Cathepsin S acts via protease-activated receptor 2 to activate sensory neurons and induce itch-like behaviour. Neurobiol. Pain 6:100032
    [Google Scholar]
  14. Coulie PJ, Ghys L, Rihoux JP 1991. Histamine-induced wheal, flare, and pruritus: inhibition by cetirizine, terfenadine, and placebo. Drug Dev. Res. 23:269–74
    [Google Scholar]
  15. Davidson S, Zhang X, Khasabov SG, Moser HR, Honda CN et al. 2012. Pruriceptive spinothalamic tract neurons: physiological properties and projection targets in the primate. J. Neurophysiol. 108:1711–23
    [Google Scholar]
  16. Davidson S, Zhang X, Khasabov SG, Simone DA, Giesler GJ 2009. Relief of itch by scratching: state-dependent inhibition of primate spinothalamic tract neurons. Nat. Neurosci. 12:544–46
    [Google Scholar]
  17. Dillon SR, Sprecher C, Hammond A, Bilsborough J, Rosenfeld-Franklin M et al. 2004. Interleukin 31, a cytokine produced by activated T cells, induces dermatitis in mice. Nat. Immunol. 5:752–60
    [Google Scholar]
  18. Dong XT, Dong XZ. 2018. Peripheral and central mechanisms of itch. Neuron 98:482–94
    [Google Scholar]
  19. Dong XZ, Han SK, Zylka MJ, Simon MI, Anderson DJ 2001. A diverse family of GPCRs expressed in specific subsets of nociceptive sensory neurons. Cell 106:619–32
    [Google Scholar]
  20. Emrick JJ, Mathur A, Wei J, Gracheva EO, Gronert K et al. 2018. Tissue-specific contributions of Tmem79 to atopic dermatitis and mast cell-mediated histaminergic itch. PNAS 115:E12091–100
    [Google Scholar]
  21. Feng J, Luo J, Yang P, Du J, Kim BS et al. 2018. Piezo2 channel–Merkel cell signaling modulates the conversion of touch to itch. Science 360:530–33
    [Google Scholar]
  22. Feng J, Yang P, Mack MR, Dryn D, Luo J et al. 2017. Sensory TRP channels contribute differentially to skin inflammation and persistent itch. Nat. Commun. 8:980
    [Google Scholar]
  23. Fleming MS, Ramos D, Han SB, Zhao J, Son YJ et al. 2012. The majority of dorsal spinal cord gastrin releasing peptide is synthesized locally whereas neuromedin B is highly expressed in pain- and itch-sensing somatosensory neurons. Mol. Pain 8:52
    [Google Scholar]
  24. Follansbee T, Akiyama T, Fujii M, Davoodi A, Nagamine M 2018. Effects of pruritogens and algogens on rostral ventromedial medullary ON and OFF cells. J. Neurophysiol. 120:2156–63
    [Google Scholar]
  25. Fukuoka M, Miyachi Y, Ikoma A 2013. Mechanically evoked itch in humans. Pain 154:897–904
    [Google Scholar]
  26. Gao ZR, Chen WZ, Liu MZ, Chen XJ, Wan LI et al. 2019. Tac1-expressing neurons in the periaqueductal gray facilitate the itch-scratching cycle via descending regulation. Neuron 101:45–59
    [Google Scholar]
  27. Gotoh Y, Omori Y, Andoh T, Kuraishi Y 2011. Tonic inhibition of allergic itch signaling by the descending noradrenergic system in mice. J. Pharmacol. Sci. 115:417–20
    [Google Scholar]
  28. Green D, Limjunyawong N, Gour N, Pundir P, Dong X 2019. A mast-cell-specific receptor mediates neurogenic inflammation and pain. Neuron 101:412–20
    [Google Scholar]
  29. Grimstad Ø, Sawanobori Y, Vestergaard C, Bilsborough J, Olsen UB et al. 2008. Anti-interleukin-31-antibodies ameliorate scratching behaviour in NC/Nga mice: a model of atopic dermatitis. Exp. Dermatol. 18:35–43
    [Google Scholar]
  30. Han L, Ma C, Liu Q, Weng HJ, Cui Y 2013. A subpopulation of nociceptors specifically linked to itch. Nat. Neurosci. 16:174–82
    [Google Scholar]
  31. Han Q, Liu D, Convertino M, Wang Z, Jiang C et al. 2018. miRNA-711 binds and activates TRPA1 extracellularly to evoke acute and chronic pruritus. Neuron 99:449–63
    [Google Scholar]
  32. Han SK, Dong X, Hwang JI, Zylka MJ, Anderson DJ et al. 2002. Orphan G protein-coupled receptors MrgA1 and MrgC11 are distinctively activated by RF-amide-related peptides through the Gαq/11 pathway. PNAS 99:14740–45
    [Google Scholar]
  33. Hashimoto T, Ohata H, Momose K 2004. Itch-scratch responses induced by lysophosphatidic acid in mice. Pharmacology 72:51–56
    [Google Scholar]
  34. Herde L, Forster C, Strupf M, Handwerker HO 2007. Itch induced by a novel method leads to limbic deactivations—a functional MRI study. J. Neurophysiol. 98:2347–56
    [Google Scholar]
  35. Hill RZ, Morita T, Brem RB, Bautista DM 2018. S1PR3 mediates itch and pain via distinct TRP channel-dependent pathways. J. Neurosci. 38:7833–43
    [Google Scholar]
  36. Hosogi M, Schmelz M, Miyachi Y, Ikoma A 2006. Bradykinin is a potent pruritogen in atopic dermatitis: a switch from pain to itch. Pain 126:16–23
    [Google Scholar]
  37. Huang CC, Yang W, Guo C, Jiang H, Li F et al. 2018. Anatomical and functional dichotomy of ocular itch and pain. Nat. Med. 24:1268–76
    [Google Scholar]
  38. Huang J, Polgár E, Solinski HJ, Mishra SK, Tseng PY et al. 2018. Circuit dissection of the role of somatostatin in itch and pain. Nat. Neurosci. 21:707–16
    [Google Scholar]
  39. Huang T, Lin S-H, Malewicz NM, Zhang Y, Zhang Y et al. 2018. Identifying the pathways required for coping behaviours associated with sustained pain. Nature 565:86–90
    [Google Scholar]
  40. Imamachi N, Park GH, Lee H, Anderson DJ, Simon MI et al. 2009. TRPV1-expressing primary afferents generate behavioral responses to pruritogens via multiple mechanisms. PNAS 106:11330–35
    [Google Scholar]
  41. Jurcakova D, Ru F, Undem B 2019. Allergen-induced histaminergic and non-histaminergic activation of itch C-fiber nerve terminals in mouse skin. Neuroscience 410:55–58
    [Google Scholar]
  42. Kardon AP, Polgar E, Hachisuka J, Snyder LM, Cameron D et al. 2014. Dynorphin acts as a neuromodulator to inhibit itch in the dorsal horn of the spinal cord. Neuron 82:573–86
    [Google Scholar]
  43. Kasutani K, Fujii E, Ohyama S, Adachi H, Hasegawa M et al. 2014. Anti-IL-31 receptor antibody is shown to be a potential therapeutic option for treating itch and dermatitis in mice. Br. J. Pharmacol. 171:5049–58
    [Google Scholar]
  44. Kido-Nakahara M, Buddenkotte J, Kempkes C, Ikoma A, Cevikbas F et al. 2014. Neural peptidase endothelin-converting enzyme 1 regulates endothelin 1-induced pruritus. J. Clin. Investig. 124:2683–95
    [Google Scholar]
  45. Kittaka H, Uchida K, Fukuta N, Tominaga M 2017. Lysophosphatidic acid–induced itch is mediated by signalling of LPA5 receptor, phospholipase D and TRPA1/TRPV1. J. Physiol. 595:2681–98
    [Google Scholar]
  46. Ko M, Naughton NN. 2000. An experimental itch model in monkeys: characterization of intrathecal morphine-induced scratching and antinociception. Anesthesiology 92:795–805
    [Google Scholar]
  47. Lagerstrom M, Rogoz K, Abrahamsen B, Persson E, Reinius B et al. 2010. VGLUT2-dependent sensory neurons in the TRPV1 population regulate pain and itch. Neuron 68:529–42
    [Google Scholar]
  48. LaMotte RH, Dong XZ, Ringkamp M 2014. Sensory neurons and circuits mediating itch. Nat. Rev. Neurosci. 15:19–31
    [Google Scholar]
  49. Lembo PM, Grazzini E, Groblewski T, O'Donnell D, Roy MO et al. 2002. Proenkephalin A gene products activate a new family of sensory neuron-specific GPCRs. Nat. Neurosci. 5:201–9
    [Google Scholar]
  50. Lewis T. 1942. Pain New York: Macmillan
  51. Lieu T, Jayaweera G, Zhao P, Poole DP, Jensen D et al. 2014. The bile acid receptor TGR5 activates the TRPA1 channel to induce itch in mice. Gastroenterology 147:1417–28
    [Google Scholar]
  52. Liu B, Tai Y, Achanta S, Kaelberer MM, Caceres AI et al. 2016. IL-33/ST2 signaling excites sensory neurons and mediates itch response in a mouse model of poison ivy contact allergy. PNAS 113:4E7572–79
    [Google Scholar]
  53. Liu Q, Sikand P, Ma C, Tang Z, Han L et al. 2012. Mechanisms of itch evoked by β-alanine. J. Neurosci. 32:14532–37
    [Google Scholar]
  54. Liu Q, Tang Z, Surdenikova L, Kim S, Patel KN et al. 2009. Sensory neuron-specific GPCR Mrgprs are itch receptors mediating chloroquine-induced pruritus. Cell 139:1353–65
    [Google Scholar]
  55. Liu Q, Weng HJ, Patel KN, Tang Z, Bai H et al. 2011. The distinct roles of two GPCRs, MrgprC11 and PAR2, in itch and hyperalgesia. Sci. Signal. 4:181ra45
    [Google Scholar]
  56. Liu T, Berta T, Xu ZZ, Park CK, Zhang L et al. 2012. TLR3 deficiency impairs spinal cord synaptic transmission, central sensitization, and pruritus in mice. J. Clin. Investig. 122:2195–207
    [Google Scholar]
  57. Liu T, Xu ZZ, Park CK, Berta T, Ji RR 2010. Toll-like receptor 7 mediates pruritus. Nat. Neurosci. 13:1460–62
    [Google Scholar]
  58. Liu X, Liu ZC, Sun Y, Ross M, Kim S et al. 2011. Unidirectional cross-activation of GRPR by MOR1D uncouples itch and analgesia induced by opioids. Cell 147:447–58
    [Google Scholar]
  59. Liu Y, Samad O, Zhang L, Duan B, Tong Q et al. 2010. VGLUT2-dependent glutamate release from nociceptors is required to sense pain and suppress itch. Neuron 68:543–56
    [Google Scholar]
  60. Mack M, Kim BS. 2018. The itch-scratch cycle: a neuroimmune perspective. Trends Immunol 39:980–91
    [Google Scholar]
  61. McNeil BD, Pundir P, Meeker S, Han L, Undem BJ et al. 2015. Identification of a mast cell specific receptor crucial for pseudoallergic drug reactions. Nature 519:237–41
    [Google Scholar]
  62. Meixiong J, Anderson M, Limjunyawong N, Sabbagh M, Hu E et al. 2019a. Activation of mast-cell-expressed mas-related G-protein-coupled receptors drives non-histaminergic itch. Immunity 50:1163–71
    [Google Scholar]
  63. Meixiong J, Vasavda C, Green D, Zheng Q, Qi L et al. 2019b. Identification of a bilirubin receptor may mediate a component of cholestatic itch. eLife 8:e44116
    [Google Scholar]
  64. Meixiong J, Vasavda C, Snyder SH, Dong X 2019c. MRGPRX4 is a G protein-coupled receptor activated by bile acids that may contribute to cholestatic pruritus. PNAS 116:10525–30
    [Google Scholar]
  65. Metcalfe DD, Baram D, Mekori YA 1997. Mast cells. Physiol. Rev. 77:1033–79
    [Google Scholar]
  66. Mishra SK, Hoon MA. 2013. The cells and circuitry for itch responses in mice. Science 340:968–71
    [Google Scholar]
  67. Morita T, McClain SP, Batia LM, Pellegrino M, Wilson SR et al. 2015. HTR7 mediates serotonergic acute and chronic itch. Neuron 87:124–38
    [Google Scholar]
  68. Mu D, Deng J, Liu KF, Wu ZY, Shi YF et al. 2017. A central neural circuit for itch sensation. Science 357:695–99
    [Google Scholar]
  69. Munanairi A, Liu X-Y, Barry DM, Yang Q, Yin J-B et al. 2018. Non-canonical opioid signaling inhibits itch transmission in the spinal cord of mice. Cell Rep 23:866–77
    [Google Scholar]
  70. Oetjen LK, Mack MR, Feng J, Whelan TM, Niu H et al. 2017. Sensory neurons co-opt classical immune signaling pathways to mediate chronic itch. Cell 171:217–28
    [Google Scholar]
  71. Pagani M, Albisetti G, Sivakumar N, Wildner H, Santello M et al. 2019. How gastrin-releasing peptide opens the spinal gate for itch. Neuron 103:102–17
    [Google Scholar]
  72. Palkar R, Ongun S, Catich E, Li N, Broad N et al. 2018. Cooling relief of acute and chronic itch requires TRPM8 channels and neurons. J. Investig. Dermatol. 138:1391–99
    [Google Scholar]
  73. Pan H, Fatima M, Li A, Lee H, Cai W et al. 2019. Identification of a spinal circuit for mechanical and persistent spontaneous itch. Neuron 103:1135–49.e6
    [Google Scholar]
  74. Reddy VB, Azimi E, Chu L, Lerner EA 2018. Mas-related G-protein coupled receptors and cowhage-induced itch. J. Investig. Dermatol. 138:461–64
    [Google Scholar]
  75. Reddy VB, Iuga AO, Shimada SG, LaMotte RH, Lerner EA 2008. Cowhage-evoked itch is mediated by a novel cysteine protease: a ligand of protease-activated receptors. J. Neurosci. 28:4331–35
    [Google Scholar]
  76. Reddy VB, Shimada SG, Sikand P, LaMotte RH, Lerner EA 2010. Cathepsin S elicits itch and signals via protease-activated receptors. J. Investig. Dermatol. 130:1468–70
    [Google Scholar]
  77. Reddy VB, Sun S, Azimi E, Elmariah SB, Dong X et al. 2015. Redefining the concept of protease-activated receptors: Cathepsin S evokes itch via activation of Mrgprs. Nat. Commun. 6:7864
    [Google Scholar]
  78. Ross SE, Mardinly AR, McCord AE, Zurawski J, Cohen S et al. 2010. Loss of inhibitory interneurons in the dorsal spinal cord and elevated itch inBhlhb5 mutant mice. Neuron 65:886–98
    [Google Scholar]
  79. Sakata D, Uruno T, Matsubara K, Andoh T, Yamamura K et al. 2019. Selective role of neurokinin B in IL-31-induced itch response in mice. J. Allergy Clin. Immunol. 144:1130–33.e8
    [Google Scholar]
  80. Salvatierra J, Bustamante MD, Meixiong J, Tierney E, Dong X et al. 2018. A disease mutation reveals a role for NaV1.9 in acute itch. J. Clin. Investig. 128:5434–47
    [Google Scholar]
  81. Samineni VK, Grajales-Reyes JG, Sundaram SS, Gereau RW 2018. Cell type-specific modulation of sensory and affective components of itch in the periaqueductal gray. bioRxiv 486332. https://doi.org/10.1101/486332
    [Crossref]
  82. Sanjel B, Maeng HJ, Shim WS 2019. BAM8-22 and its receptor MRGPRX1 may attribute to cholestatic pruritus. Sci. Rep. 9:10888
    [Google Scholar]
  83. Schmelz M, Schmidt R, Bickel A, Handwerker HO, Torebjork HE 1997. Specific C-receptors for itch in human skin. J. Neurosci. 17:8003–8
    [Google Scholar]
  84. Schwendinger-Schreck J, Wilson SR, Bautista DM 2015. Interactions between keratinocytes and somatosensory neurons in itch. Pharmacology of Itch A Cowan, G Yosipovitch 177–90 Heidelberg: Springer
    [Google Scholar]
  85. Shim WS, Tak MH, Lee MH, Kim M, Koo JY et al. 2007. TRPV1 mediates histamine-induced itching via the activation of phospholipase A2 and 12-lipoxygenase. J. Neurosci. 27:2331–37
    [Google Scholar]
  86. Shinohara T, Harada M, Ogi K, Maruyama M, Fujii R et al. 2004. Identification of a G protein-coupled receptor specifically responsive to β-alanine. J. Biol. Chem. 279:23559–64
    [Google Scholar]
  87. Sikand P, Dong X, Lamotte RH 2011. BAM8-22 peptide produces itch and nociceptive sensations in humans independent of histamine release. J. Neurosci. 31:7563–67
    [Google Scholar]
  88. Solinski HJ, Kriegbaum MC, Tseng PY, Earnest TW, Gu XL et al. 2019. Nppb neurons are sensors of mast cell-induced itch. Cell Rep 26:3561–73
    [Google Scholar]
  89. Solorzano C, Villafuerte D, Meda K, Cervikbas F, Braz J et al. 2015. Primary afferent and spinal cord expression of gastrin-releasing peptide: message, protein, and antibody concerns. J. Neurosci. 35:648–57
    [Google Scholar]
  90. Stefansson K, Brattsand M, Roosterman D, Kempkes C, Bocheva G et al. 2008. Activation of proteinase-activated receptor-2 by human kallikrein-related peptidases. J. Investig. . Dermatol 128:18–25
    [Google Scholar]
  91. Stokes JR, Romero FA, Allan RJ, Phillips PG, Hackman F et al. 2012. The effects of an H3 receptor antagonist (PF-03654746) with fexofenadin on reducing allergic rhinitis symptoms. J. Allergy Clin. Immunol. 129:409–12
    [Google Scholar]
  92. Su X, Chen M, Yuan Y, Li Y, Guo S et al. 2019. Central processing of itch in the midbrain reward center. Neuron 102:858–72
    [Google Scholar]
  93. Sun S, Xu Q, Guo C, Guan Y, Liu Q, Dong X 2017. Leaky gate model: intensity-dependent coding of pain and itch in the spinal cord. Neuron 93:840–53
    [Google Scholar]
  94. Sun YG, Chen ZF. 2007. A gastrin-releasing peptide receptor mediates the itch sensation in the spinal cord. Nature 448:700–3
    [Google Scholar]
  95. Sun YG, Zhao ZQ, Meng XL, Yin J, Liu XY et al. 2009. Cellular basis of itch sensation. Science 325:1531–34
    [Google Scholar]
  96. Trentin PG, Fernandes MB, D'Orleans-Juste P, Rae GA 2006. Endothelin-1 causes pruritus in mice. Exp. Biol. Med. 231:1146–51
    [Google Scholar]
  97. Tseng PY, Zheng Q, Zhe Li, Dong X 2019. MrgprX1 mediates neuronal excitability and itch through tetrodotoxin-resistant sodium channels. Itch 4:3e28
    [Google Scholar]
  98. Usoskin D, Furlan A, Islam S, Abdo H, Lönnerberg P et al. 2015. Unbiased classification of sensory neuron types by large-scale single-cell RNA sequencing. Nat. Neurosci. 18:145–53
    [Google Scholar]
  99. Wang X, Zhang J, Eberhart D, Urban R, Meda K et al. 2013. Excitatory superficial dorsal horn interneurons are functionally heterogeneous and required for the full behavioral expression of pain and itch. Neuron 78:312–24
    [Google Scholar]
  100. Wilson SR, The L, Batia LM, Beattie K, Katibah GE et al. 2013. The epithelial cell-derived atopic dermatitis cytokine TSLP activates neurons to induce itch. Cell 155:285–95
    [Google Scholar]
  101. Xu Y, Lopes C, Wende H, Guo Z, Cheng L et al. 2013. Ontogeny of excitatory spinal neurons processing distinct somatic sensory modalities. J. Neurosci. 33:14738–48
    [Google Scholar]
  102. Yamaguchi T, Nagasawa T, Satoh M, Kuraishi Y 1999. Itch-associated response induced by intradermal serotonin through 5-HT2 receptors in mice. Neurosci. Res. 35:77–83
    [Google Scholar]
  103. Yosipovitch G, Fast K, Bernhard J 2005. Noxious heat and scratching decrease histamine-induced itch and skin blood flow. J. Investig. Dermatol. 125:1268–72
    [Google Scholar]
  104. Yu H, Zhao T, Liu S, Wu Q, Johnson O et al. 2019. MRGPRX4 is a novel bile acid receptor in cholestatic itch. bioRxiv 633446. https://doi.org/10.1101/633446
    [Crossref]
  105. Zhao ZQ, Liu XY, Jeffry J, Karunarathne WK, Li JL et al. 2014. Descending control of itch transmission by the serotonergic system via 5-HT1A-facilitated GRP-GRPR signaling. Neuron 84:821–34
    [Google Scholar]
  106. Zheng T, Oh MH, Oh SY, Schroeder JT, Glick AB et al. 2009. Transgenic expression of interleukin-13 in the skin induces a pruritic dermatitis and skin remodeling. J. Investig. Dermatol. 129:742–51
    [Google Scholar]
/content/journals/10.1146/annurev-neuro-083019-024537
Loading
/content/journals/10.1146/annurev-neuro-083019-024537
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error