1932

Abstract

The study of chronic pain continues to generate ever-increasing numbers of publications, but safe and efficacious treatments for chronic pain remain elusive. Recognition of sex-specific mechanisms underlying chronic pain has resulted in a surge of studies that include both sexes. A predominant focus has been on identifying sex differences, yet many newly identified cellular mechanisms and alterations in gene expression are conserved between the sexes. Here we review sex differences and similarities in cellular and molecular signals that drive the generation and resolution of neuropathic pain. The mix of differences and similarities reflects degeneracy in peripheral and central signaling processes by which neurons, immune cells, and glia codependently drive pain hypersensitivity. Recent findings identifying critical signaling nodes foreshadow the development of rationally designed, broadly applicable analgesic strategies. However, the paucity of effective, safe pain treatments compels targeted therapies as well to increase therapeutic options that help reduce the global burden of suffering.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pharmtox-051421-112259
2023-01-20
2024-05-20
Loading full text...

Full text loading...

/deliver/fulltext/pharmtox/63/1/annurev-pharmtox-051421-112259.html?itemId=/content/journals/10.1146/annurev-pharmtox-051421-112259&mimeType=html&fmt=ahah

Literature Cited

  1. 1.
    Raja SN, Carr DB, Cohen M, Finnerup NB, Flor H et al. 2020. The revised International Association for the Study of Pain definition of pain: concepts, challenges, and compromises. Pain 161:1976–82
    [Google Scholar]
  2. 2.
    Shupler MS, Kramer JK, Cragg JJ, Jutzeler CR, Whitehurst DGT. 2019. Pan-Canadian estimates of chronic pain prevalence from 2000 to 2014: a repeated cross-sectional survey analysis. J. Pain 20:557–65
    [Google Scholar]
  3. 3.
    Zelaya CE, Dahlhamer JM, Lucas JW, Connor EM. 2020. Chronic pain and high-impact chronic pain among US adults, 2019. NCHS Data Brief 390:1–8
    [Google Scholar]
  4. 4.
    Gaskin DJ, Richard P. 2012. The economic costs of pain in the United States. J. Pain 13:715–24
    [Google Scholar]
  5. 5.
    Treede R-D, Rief W, Barke A, Aziz Q, Bennett MI et al. 2019. Chronic pain as a symptom or a disease: the IASP Classification of Chronic Pain for the International Classification of Diseases (ICD-11). Pain 160:19–27
    [Google Scholar]
  6. 6.
    van Hecke O, Austin SK, Khan RA, Smith BH, Torrance N 2014. Neuropathic pain in the general population: a systematic review of epidemiological studies. Pain 155:654–62
    [Google Scholar]
  7. 7.
    Mogil JS. 2012. Sex differences in pain and pain inhibition: multiple explanations of a controversial phenomenon. Nat. Rev. Neurosci. 13:859–66
    [Google Scholar]
  8. 8.
    NIH (Natl. Inst. Health) 2015. Consideration of sex as a biological variable in NIH-funded research NIH Notice NOT-OD-15-102 NIH Bethesda, MA:
  9. 9.
    Bale TL, Epperson CN. 2017. Sex as a biological variable: who, what, when, why, and how. Neuropsychopharmacology 42:386–96
    [Google Scholar]
  10. 10.
    Mapplebeck JC, Beggs S, Salter MW. 2017. Molecules in pain and sex: a developing story. Mol. Brain 10:9
    [Google Scholar]
  11. 11.
    Linher-Melville K, Shah A, Singh G. 2020. Sex differences in neuro(auto)immunity and chronic sciatic nerve pain. Biol. Sex Differ. 11:62
    [Google Scholar]
  12. 12.
    Rosen S, Ham B, Mogil JS. 2017. Sex differences in neuroimmunity and pain. J. Neurosci. Res. 95:500–8
    [Google Scholar]
  13. 13.
    Keller AF, Beggs S, Salter MW, De Koninck Y. 2007. Transformation of the output of spinal lamina I neurons after nerve injury and microglia stimulation underlying neuropathic pain. Mol. Pain 3:27
    [Google Scholar]
  14. 14.
    Salter MW, Stevens B. 2017. Microglia emerge as central players in brain disease. Nat. Med. 23:1018–27
    [Google Scholar]
  15. 15.
    Tsuda M, Shigemoto-Mogami Y, Koizumi S, Mizokoshi A, Kohsaka S et al. 2003. P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature 424:778–83
    [Google Scholar]
  16. 16.
    Inoue K, Tsuda M. 2018. Microglia in neuropathic pain: cellular and molecular mechanisms and therapeutic potential. Nat. Rev. Neurosci. 19:138–52
    [Google Scholar]
  17. 17.
    Tam TH, Salter MW. 2021. Purinergic signalling in spinal pain processing. Purinergic Signal 17:49–54
    [Google Scholar]
  18. 18.
    Guan Z, Kuhn JA, Wang X, Colquitt B, Solorzano C et al. 2016. Injured sensory neuron-derived CSF1 induces microglial proliferation and DAP12-dependent pain. Nat. Neurosci. 19:94–101
    [Google Scholar]
  19. 19.
    Beggs S, Trang T, Salter MW 2012. P2X4R+ microglia drive neuropathic pain. Nat. Neurosci. 15:1068–73
    [Google Scholar]
  20. 20.
    Masuda T, Iwamoto S, Yoshinaga R, Tozaki-Saitoh H, Nishiyama A et al. 2014. Transcription factor IRF5 drives P2X4R+-reactive microglia gating neuropathic pain. Nat. Commun. 5:3771
    [Google Scholar]
  21. 21.
    Masuda T, Tsuda M, Yoshinaga R, Tozaki-Saitoh H, Ozato K et al. 2012. IRF8 is a critical transcription factor for transforming microglia into a reactive phenotype. Cell Rep. 1:334–40
    [Google Scholar]
  22. 22.
    Mapplebeck JCS, Dalgarno R, Tu Y, Moriarty O, Beggs S et al. 2018. Microglial P2X4R-evoked pain hypersensitivity is sexually dimorphic in rats. Pain 159:1752–63
    [Google Scholar]
  23. 23.
    Masuda T, Ozono Y, Mikuriya S, Kohro Y, Tozaki-Saitoh H et al. 2016. Dorsal horn neurons release extracellular ATP in a VNUT-dependent manner that underlies neuropathic pain. Nat. Commun. 7:12529
    [Google Scholar]
  24. 24.
    Sorge RE, Mapplebeck JC, Rosen S, Beggs S, Taves S et al. 2015. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat. Neurosci. 18:1081–83
    [Google Scholar]
  25. 25.
    Agalave NM, Larsson M, Abdelmoaty S, Su J, Baharpoor A et al. 2014. Spinal HMGB1 induces TLR4-mediated long-lasting hypersensitivity and glial activation and regulates pain-like behavior in experimental arthritis. Pain 155:1802–13
    [Google Scholar]
  26. 26.
    Fernandez-Zafra T, Gao T, Jurczak A, Sandor K, Hore Z et al. 2019. Exploring the transcriptome of resident spinal microglia after collagen antibody-induced arthritis. Pain 160:224–36
    [Google Scholar]
  27. 27.
    Chen G, Luo X, Qadri MY, Berta T, Ji RR 2018. Sex-dependent glial signaling in pathological pain: distinct roles of spinal microglia and astrocytes. Neurosci. Bull. 34:98–108
    [Google Scholar]
  28. 28.
    Saika F, Matsuzaki S, Kobayashi D, Ideguchi Y, Nakamura TY et al. 2020. Chemogenetic regulation of CX3CR1-expressing microglia using Gi-DREADD exerts sex-dependent anti-allodynic effects in mouse models of neuropathic pain. Front. Pharmacol. 11:925
    [Google Scholar]
  29. 29.
    Taves S, Berta T, Liu DL, Gan S, Chen G et al. 2016. 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. Immunity 55:70–81
    [Google Scholar]
  30. 30.
    Westlund KN, Montera MA, Goins AE, Alles SRA, Suri N et al. 2021. Single-dose P2 X4R single-chain fragment variable antibody permanently reverses chronic pain in male mice. Int. J. Mol. Sci. 22:13612
    [Google Scholar]
  31. 31.
    Luo X, Fitzsimmons B, Mohan A, Zhang L, Terrando N et al. 2018. 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. Immunity 72:34–44
    [Google Scholar]
  32. 32.
    Mapplebeck JCS, Lorenzo LE, Lee KY, Gauthier C, Muley MM et al. 2019. Chloride dysregulation through downregulation of KCC2 mediates neuropathic pain in both sexes. Cell Rep 28:590–96.e4
    [Google Scholar]
  33. 33.
    Tsujikawa S, DeMeulenaere KE, Centeno MV, Ghazisaeidi S, Martin ME et al. 2022. Regulation of neuropathic pain by microglial Orai1 channels. bioRxiv 2022.09.02.506250. https://doi.org/10.1101/2022.09.02.506250
    [Crossref]
  34. 34.
    Trang T, Beggs S, Wan X, Salter MW 2009. P2X4-receptor-mediated synthesis and release of brain-derived neurotrophic factor in microglia is dependent on calcium and p38-mitogen-activated protein kinase activation. J. Neurosci. 29:3518–28
    [Google Scholar]
  35. 35.
    Saika F, Matsuzaki S, Kishioka S, Kiguchi N. 2021. Chemogenetic activation of CX3CR1-expressing spinal microglia using Gq-DREADD elicits mechanical allodynia in male mice. Cells 10:874
    [Google Scholar]
  36. 36.
    Kiguchi N, Fukazawa Y, Saika A, Uta D, Saika F et al. 2021. Chemogenetic activation of central gastrin-releasing peptide-expressing neurons elicits itch-related scratching behavior in male and female mice. Pharmacol. Res. Perspect. 9:e00790
    [Google Scholar]
  37. 37.
    Grace PM, Wang X, Strand KA, Baratta MV, Zhang Y et al. 2018. DREADDed microglia in pain: implications for spinal inflammatory signaling in male rats. Exp. Neurol. 304:125–31
    [Google Scholar]
  38. 38.
    Watkins LR, Maier SF. 2002. Beyond neurons: evidence that immune and glial cells contribute to pathological pain states. Physiol. Rev. 82:981–1011
    [Google Scholar]
  39. 39.
    Calvo M, Dawes JM, Bennett DL. 2012. The role of the immune system in the generation of neuropathic pain. Lancet Neurol. 11:629–42
    [Google Scholar]
  40. 40.
    Hu P, McLachlan EM. 2002. Macrophage and lymphocyte invasion of dorsal root ganglia after peripheral nerve lesions in the rat. Neuroscience 112:23–38
    [Google Scholar]
  41. 41.
    Liu T, van Rooijen N, Tracey DJ. 2000. Depletion of macrophages reduces axonal degeneration and hyperalgesia following nerve injury. Pain 86:25–32
    [Google Scholar]
  42. 42.
    Barclay J, Clark AK, Ganju P, Gentry C, Patel S et al. 2007. Role of the cysteine protease cathepsin S in neuropathic hyperalgesia. Pain 130:225–34
    [Google Scholar]
  43. 43.
    Yu X, Liu H, Hamel KA, Morvan MG, Yu S et al. 2020. Dorsal root ganglion macrophages contribute to both the initiation and persistence of neuropathic pain. Nat. Commun. 11:264
    [Google Scholar]
  44. 44.
    Simeoli R, Montague K, Jones HR, Castaldi L, Chambers D et al. 2017. Exosomal cargo including microRNA regulates sensory neuron to macrophage communication after nerve trauma. Nat. Commun. 8:1778
    [Google Scholar]
  45. 45.
    Cobos EJ, Nickerson CA, Gao F, Chandran V, Bravo-Caparrós I et al. 2018. Mechanistic differences in neuropathic pain modalities revealed by correlating behavior with global expression profiling. Cell Rep. 22:1301–12
    [Google Scholar]
  46. 46.
    Vega-Avelaira D, Géranton SM, Fitzgerald M. 2009. Differential regulation of immune responses and macrophage/neuron interactions in the dorsal root ganglion in young and adult rats following nerve injury. Mol. Pain 5:70
    [Google Scholar]
  47. 47.
    Luo X, Huh Y, Bang S, He Q, Zhang L et al. 2019. Macrophage Toll-like receptor 9 contributes to chemotherapy-induced neuropathic pain in male mice. J. Neurosci. 39:6848–64
    [Google Scholar]
  48. 48.
    Luo X, Chen O, Wang Z, Bang S, Ji J et al. 2021. IL-23/IL-17A/TRPV1 axis produces mechanical pain via macrophage-sensory neuron crosstalk in female mice. Neuron 109:2691–706.e5
    [Google Scholar]
  49. 49.
    Silva CEA, Guimarães RM, Cunha TM. 2021. Sensory neuron-associated macrophages as novel modulators of neuropathic pain. Pain Rep. 6:e873
    [Google Scholar]
  50. 50.
    Tu Y, Muley MM, Beggs S, Salter MW. 2022. Microglia-independent peripheral neuropathic pain in male and female mice. Pain 163:e112944
    [Google Scholar]
  51. 51.
    Old EA, Nadkarni S, Grist J, Gentry C, Bevan S et al. 2014. Monocytes expressing CX3CR1 orchestrate the development of vincristine-induced pain. J. Clin. Investig. 124:2023–36
    [Google Scholar]
  52. 52.
    Leung L, Cahill CM. 2010. TNF-α and neuropathic pain—a review. J. Neuroinflamm. 7:27
    [Google Scholar]
  53. 53.
    Del Rivero T, Fischer R, Yang F, Swanson KA, Bethea JR. 2019. Tumor necrosis factor receptor 1 inhibition is therapeutic for neuropathic pain in males but not in females. Pain 160:922–31
    [Google Scholar]
  54. 54.
    Haydon PG. 2001. GLIA: listening and talking to the synapse. Nat. Rev. Neurosci. 2:185–93
    [Google Scholar]
  55. 55.
    Escartin C, Galea E, Lakatos A, O'Callaghan JP, Petzold GC et al. 2021. Reactive astrocyte nomenclature, definitions, and future directions. Nat. Neurosci. 24:312–25
    [Google Scholar]
  56. 56.
    Halassa MM, Fellin T, Takano H, Dong JH, Haydon PG. 2007. Synaptic islands defined by the territory of a single astrocyte. J. Neurosci. 27:6473–77
    [Google Scholar]
  57. 57.
    Gao YJ, Ji RR. 2010. Targeting astrocyte signaling for chronic pain. Neurotherapeutics 7:482–93
    [Google Scholar]
  58. 58.
    Li T, Chen X, Zhang C, Zhang Y, Yao W. 2019. An update on reactive astrocytes in chronic pain. J. Neuroinflamm. 16:140
    [Google Scholar]
  59. 59.
    Donnelly CR, Andriessen AS, Chen G, Wang K, Jiang C et al. 2020. Central nervous system targets: glial cell mechanisms in chronic pain. Neurotherapeutics 17:846–60
    [Google Scholar]
  60. 60.
    Zhang J, De Koninck Y. 2006. Spatial and temporal relationship between monocyte chemoattractant protein-1 expression and spinal glial activation following peripheral nerve injury. J. Neurochem. 97:772–83
    [Google Scholar]
  61. 61.
    Zhuang ZY, Gerner P, Woolf CJ, Ji RR 2005. ERK is sequentially activated in neurons, microglia, and astrocytes by spinal nerve ligation and contributes to mechanical allodynia in this neuropathic pain model. Pain 114:149–59
    [Google Scholar]
  62. 62.
    Echeverry S, Shi XQ, Zhang J. 2008. Characterization of cell proliferation in rat spinal cord following peripheral nerve injury and the relationship with neuropathic pain. Pain 135:37–47
    [Google Scholar]
  63. 63.
    Chen Z, Doyle TM, Luongo L, Largent-Milnes TM, Giancotti LA et al. 2019. Sphingosine-1-phosphate receptor 1 activation in astrocytes contributes to neuropathic pain. PNAS 116:10557–62
    [Google Scholar]
  64. 64.
    Doyle TM, Chen Z, Durante M, Salvemini D 2019. Activation of sphingosine-1-phosphate receptor 1 in the spinal cord produces mechanohypersensitivity through the activation of inflammasome and IL-1β pathway. J. Pain 20:956–64
    [Google Scholar]
  65. 65.
    Stockstill K, Wahlman C, Braden K, Chen Z, Yosten GL et al. 2020. Sexually dimorphic therapeutic response in bortezomib-induced neuropathic pain reveals altered pain physiology in female rodents. Pain 161:177–84
    [Google Scholar]
  66. 66.
    Moalem G, Xu K, Yu L 2004. T lymphocytes play a role in neuropathic pain following peripheral nerve injury in rats. Neuroscience 129:767–77
    [Google Scholar]
  67. 67.
    Lopes DM, Malek N, Edye M, Jager SB, McMurray S et al. 2017. Sex differences in peripheral not central immune responses to pain-inducing injury. Sci. Rep. 7:16460
    [Google Scholar]
  68. 68.
    Du B, Ding YQ, Xiao X, Ren HY, Su BY, Qi JG. 2018. CD4+ αβ T cell infiltration into the leptomeninges of lumbar dorsal roots contributes to the transition from acute to chronic mechanical allodynia after adult rat tibial nerve injuries. J. Neuroinflamm. 15:81
    [Google Scholar]
  69. 69.
    Cao L, DeLeo J. 2008. CNS-infiltrating CD4+ T lymphocytes contribute to murine spinal nerve transection-induced neuropathic pain. Eur. J. Immunol. 38:448–58
    [Google Scholar]
  70. 70.
    Costigan M, Moss A, Latremoliere A, Johnston C, Verma-Gandhu M et al. 2009. T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. J. Neurosci. 29:14415–22
    [Google Scholar]
  71. 71.
    Laumet G, Ma J, Robison AJ, Kumari S, Heijnen CJ, Kavelaars A. 2019. T cells as an emerging target for chronic pain therapy. Front. Mol. Neurosci. 12:216
    [Google Scholar]
  72. 72.
    Kobayashi Y, Kiguchi N, Fukazawa Y, Saika F, Maeda T, Kishioka S. 2015. Macrophage-T cell interactions mediate neuropathic pain through the glucocorticoid-induced tumor necrosis factor ligand system. J. Biol. Chem. 290:12603–13
    [Google Scholar]
  73. 73.
    Pasciuto E, Burton OT, Roca CP, Lagou V, Rajan WD et al. 2020. Microglia require CD4 T cells to complete the fetal-to-adult transition. Cell 182:625–40.e24
    [Google Scholar]
  74. 74.
    Costigan M, Moss A, Latremoliere A, Johnston C, Verma-Gandhu M et al. 2009. T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. J. Neurosci. 29:14415–22
    [Google Scholar]
  75. 75.
    Gensel JC, Donahue RR, Bailey WM, Taylor BK. 2019. Sexual dimorphism of pain control: analgesic effects of pioglitazone and azithromycin in chronic spinal cord injury. J. Neurotrauma 36:2372–76
    [Google Scholar]
  76. 76.
    Santos DFS, Donahue RR, Laird DE, Oliveira MCG, Taylor BK. 2022. The PPARγ agonist pioglitazone produces a female-predominant inhibition of hyperalgesia associated with surgical incision, peripheral nerve injury, and painful diabetic neuropathy. Neuropharmacology 205:108907
    [Google Scholar]
  77. 77.
    Kuhn JA, Vainchtein ID, Braz J, Hamel K, Bernstein M et al. 2021. Regulatory T-cells inhibit microglia-induced pain hypersensitivity in female mice. eLife 10:e69056
    [Google Scholar]
  78. 78.
    Krukowski K, Eijkelkamp N, Laumet G, Hack CE, Li Y et al. 2016. CD8+ T cells and endogenous IL-10 are required for resolution of chemotherapy-induced neuropathic pain. J. Neurosci. 36:11074–83
    [Google Scholar]
  79. 79.
    Davoli-Ferreira M, de Lima KA, Fonseca MM, Guimarães RM, Gomes FI et al. 2020. Regulatory T cells counteract neuropathic pain through inhibition of the Th1 response at the site of peripheral nerve injury. Pain 161:1730–43
    [Google Scholar]
  80. 80.
    Fischer R, Sendetski M, Del Rivero T, Martinez GF, Bracchi-Ricard V et al. 2019. TNFR2 promotes Treg-mediated recovery from neuropathic pain across sexes. PNAS 116:17045–50
    [Google Scholar]
  81. 81.
    Woolf CJ, Salter MW. 2000. Neuronal plasticity: increasing the gain in pain. Science 288:1765–69
    [Google Scholar]
  82. 82.
    Alsaloum M, Waxman SG. 2022. iPSCs and DRGs: stepping stones to new pain therapies. Trends Mol. Med. 28:110–22
    [Google Scholar]
  83. 83.
    Basbaum AI, Bautista DM, Scherrer G, Julius D 2009. Cellular and molecular mechanisms of pain. Cell 139:267–84
    [Google Scholar]
  84. 84.
    Zamponi GW, Lewis RJ, Todorovic SM, Arneric SP, Snutch TP. 2009. Role of voltage-gated calcium channels in ascending pain pathways. Brain Res. Rev. 60:84–89
    [Google Scholar]
  85. 85.
    Chandra S, Wang Z, Tao X, Chen O, Luo X et al. 2020. Computer-aided discovery of a new Nav1.7 inhibitor for treatment of pain and itch. Anesthesiology 133:611–27
    [Google Scholar]
  86. 86.
    O'Brien MS, Philpott HTA, McDougall JJ. 2019. Targeting the Nav1.8 ion channel engenders sex-specific responses in lysophosphatidic acid-induced joint neuropathy. Pain 160:269–78
    [Google Scholar]
  87. 87.
    Smith MT, Cabot PJ, Ross FB, Robertson AD, Lewis RJ. 2002. The novel N-type calcium channel blocker, AM336, produces potent dose-dependent antinociception after intrathecal dosing in rats and inhibits substance P release in rat spinal cord slices. Pain 96:119–27
    [Google Scholar]
  88. 88.
    Luo ZD, Chaplan SR, Higuera ES, Sorkin LS, Stauderman KA et al. 2001. Upregulation of dorsal root ganglion α2δ calcium channel subunit and its correlation with allodynia in spinal nerve-injured rats. J. Neurosci. 21:1868–75
    [Google Scholar]
  89. 89.
    Altier C, Dale CS, Kisilevsky AE, Chapman K, Castiglioni AJ et al. 2007. Differential role of N-type calcium channel splice isoforms in pain. J. Neurosci. 27:6363–73
    [Google Scholar]
  90. 90.
    Chaplan SR, Pogrel JW, Yaksh TL. 1994. Role of voltage-dependent calcium channel subtypes in experimental tactile allodynia. J. Pharmacol. Exp. Ther. 269:1117–23
    [Google Scholar]
  91. 91.
    García-Caballero A, Gadotti VM, Stemkowski P, Weiss N, Souza IA et al. 2014. The deubiquitinating enzyme USP5 modulates neuropathic and inflammatory pain by enhancing Cav3.2 channel activity. Neuron 83:1144–58
    [Google Scholar]
  92. 92.
    Tomita S, Sekiguchi F, Deguchi T, Miyazaki T, Ikeda Y et al. 2019. Critical role of Cav3.2 T-type calcium channels in the peripheral neuropathy induced by bortezomib, a proteasome-inhibiting chemotherapeutic agent, in mice. Toxicology 413:33–39
    [Google Scholar]
  93. 93.
    Messinger RB, Naik AK, Jagodic MM, Nelson MT, Lee WY et al. 2009. In vivo silencing of the CaV3.2 T-type calcium channels in sensory neurons alleviates hyperalgesia in rats with streptozocin-induced diabetic neuropathy. Pain 145:184–95
    [Google Scholar]
  94. 94.
    Staats PS, Yearwood T, Charapata SG, Presley RW, Wallace MS et al. 2004. Intrathecal ziconotide in the treatment of refractory pain in patients with cancer or AIDS: a randomized controlled trial. JAMA 291:63–70
    [Google Scholar]
  95. 95.
    Donnelly CR, Jiang C, Andriessen AS, Wang K, Wang Z et al. 2021. STING controls nociception via type I interferon signalling in sensory neurons. Nature 591:275–80
    [Google Scholar]
  96. 96.
    Wang YY, Shen D, Zhao LJ, Zeng N, Hu TH. 2019. Sting is a critical regulator of spinal cord injury by regulating microglial inflammation via interacting with TBK1 in mice. Biochem. Biophys. Res. Commun. 517:741–48
    [Google Scholar]
  97. 97.
    Morozzi G, Rothen J, Toussaint G, De Lange K, Westritschnig K et al. 2021. STING regulates peripheral nerve regeneration and colony stimulating factor 1 receptor (CSF1R) processing in microglia. iScience 24:103434
    [Google Scholar]
  98. 98.
    Prescott SA, Ma Q, De Koninck Y. 2014. Normal and abnormal coding of somatosensory stimuli causing pain. Nat. Neurosci. 17:183–91
    [Google Scholar]
  99. 99.
    Craig ADB. 2003. Pain mechanisms: labeled lines versus convergence in central processing. Annu. Rev. Neurosci. 26:1–30
    [Google Scholar]
  100. 100.
    Coull JA, Beggs S, Boudreau D, Boivin D, Tsuda M et al. 2005. BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain. Nature 438:1017–21
    [Google Scholar]
  101. 101.
    Doyon N, Vinay L, Prescott SA, De Koninck Y. 2016. Chloride regulation: a dynamic equilibrium crucial for synaptic inhibition. Neuron 89:1157–72
    [Google Scholar]
  102. 102.
    Hildebrand ME, Xu J, Dedek A, Li Y, Sengar AS et al. 2016. Potentiation of synaptic GluN2B NMDAR currents by Fyn kinase is gated through BDNF-mediated disinhibition in spinal pain processing. Cell Rep. 17:2753–65
    [Google Scholar]
  103. 103.
    Pelkey KA, Askalan R, Paul S, Kalia LV, Nguyen TH et al. 2002. Tyrosine phosphatase STEP is a tonic brake on induction of long-term potentiation. Neuron 34:127–38
    [Google Scholar]
  104. 104.
    Costigan M, Befort K, Karchewski L, Griffin RS, D'Urso D et al. 2002. Replicate high-density rat genome oligonucleotide microarrays reveal hundreds of regulated genes in the dorsal root ganglion after peripheral nerve injury. BMC Neurosci. 3:16
    [Google Scholar]
  105. 105.
    Tegeder I, Costigan M, Griffin RS, Abele A, Belfer I et al. 2006. GTP cyclohydrolase and tetrahydrobiopterin regulate pain sensitivity and persistence. Nat. Med. 12:1269–77
    [Google Scholar]
  106. 106.
    Latremoliere A, Latini A, Andrews N, Cronin SJ, Fujita M et al. 2015. Reduction of neuropathic and inflammatory pain through inhibition of the tetrahydrobiopterin pathway. Neuron 86:1393–406
    [Google Scholar]
  107. 107.
    Tavares-Ferreira D, Ray PR, Sankaranarayanan I, Mejia GL, Wangzhou A et al. 2022. Sex differences in nociceptor translatomes contribute to divergent prostaglandin signaling in male and female mice. Biol. Psychiatry 91:129–40
    [Google Scholar]
  108. 108.
    Niehaus JK, Taylor-Blake B, Loo L, Simon JM, Zylka MJ. 2021. Spinal macrophages resolve nociceptive hypersensitivity after peripheral injury. Neuron 109:1274–82.e6
    [Google Scholar]
  109. 109.
    Renthal W, Tochitsky I, Yang L, Cheng YC, Li E et al. 2020. Transcriptional reprogramming of distinct peripheral sensory neuron subtypes after axonal injury. Neuron 108:128–44.e9
    [Google Scholar]
  110. 110.
    Tansley S, Uttam S, Ureña Guzmán A, Yaqubi M, Pacis A et al. 2022. Single-cell RNA sequencing reveals time- and sex-specific responses of mouse spinal cord microglia to peripheral nerve injury and links ApoE to chronic pain. Nat. Commun. 13:843
    [Google Scholar]
  111. 111.
    Kohno K, Shirasaka R, Yoshihara K, Mikuriya S, Tanaka K et al. 2022. A spinal microglia population involved in remitting and relapsing neuropathic pain. Science 376:86–90
    [Google Scholar]
  112. 112.
    Ghazisaeidi S, Muley MM, Tu Y, Kolahdouzan M, Sengar AS et al. 2022. Conserved transcriptional programming across sex and species after peripheral nerve injury predicts treatments for neuropathic pain. bioRxiv 2022.05.30.494054. https://doi.org/10.1101/2022.05.30.494054
    [Crossref]
  113. 113.
    Fillingim RB 2017. Sex, gender, and pain. Principles of Gender-Specific Medicine M Legato 481–96 London: Academic
    [Google Scholar]
  114. 114.
    Poleshuck EL, Green CR. 2008. Socioeconomic disadvantage and pain. Pain 136:235–38
    [Google Scholar]
  115. 115.
    Prego-Domínguez J, Skillgate E, Orsini N, Takkouche B. 2021. Social factors and chronic pain: the modifying effect of sex in the Stockholm Public Health Cohort Study. Rheumatology 61:1802–9
    [Google Scholar]
  116. 116.
    Leo RJ. 2005. Chronic pain and comorbid depression. Curr. Treat. Opt. Neurol. 7:403–12
    [Google Scholar]
  117. 117.
    Sramek JJ, Murphy MF, Cutler NR. 2016. Sex differences in the psychopharmacological treatment of depression. Dialogues Clin. Neurosci. 18:447–57
    [Google Scholar]
  118. 118.
    Gureje O. 2008. Comorbidity of pain and anxiety disorders. Curr. Psychiatry Rep. 10:318–22
    [Google Scholar]
  119. 119.
    Eid RS, Gobinath AR, Galea LAM. 2019. Sex differences in depression: insights from clinical and preclinical studies. Progr. Neurobiol. 176:86–102
    [Google Scholar]
  120. 120.
    Okifuji A, Hare BD. 2015. The association between chronic pain and obesity. J. Pain Res. 8:399–408
    [Google Scholar]
  121. 121.
    Salinero AE, Anderson BM, Zuloaga KL. 2018. Sex differences in the metabolic effects of diet-induced obesity vary by age of onset. Int. J. Obes. 42:1088–91
    [Google Scholar]
  122. 122.
    Schopflocher D, Taenzer P, Jovey R. 2011. The prevalence of chronic pain in Canada. Pain Res. Manag. 16:445–50
    [Google Scholar]
  123. 123.
    Tsang A, Von Korff M, Lee S, Alonso J, Karam E et al. 2008. Common chronic pain conditions in developed and developing countries: gender and age differences and comorbidity with depression-anxiety disorders. J. Pain 9:883–91
    [Google Scholar]
  124. 124.
    Lopes DM, Malek N, Edye M, Jager SB, McMurray S et al. 2017. Sex differences in peripheral not central immune responses to pain-inducing injury. Sci. Rep. 7:16460
    [Google Scholar]
  125. 125.
    Chernov AV, Hullugundi SK, Eddinger KA, Dolkas J, Remacle AG et al. 2020. A myelin basic protein fragment induces sexually dimorphic transcriptome signatures of neuropathic pain in mice. J. Biol. Chem. 295:10807–21
    [Google Scholar]
  126. 126.
    Fiore NT, Yin Z, Guneykaya D, Gauthier CD, Hayes JP et al. 2022. Sex-specific transcriptome of spinal microglia in neuropathic pain due to peripheral nerve injury. Glia 70:675–96
    [Google Scholar]
  127. 127.
    Parisien M, Samoshkin A, Tansley SN, Piltonen MH, Martin LJ et al. 2019. Genetic pathway analysis reveals a major role for extracellular matrix organization in inflammatory and neuropathic pain. Pain 160:932–44
    [Google Scholar]
  128. 128.
    Stephens KE, Chen Z, Sivanesan E, Raja SN, Linderoth B et al. 2018. RNA-seq of spinal cord from nerve-injured rats after spinal cord stimulation. Mol. Pain 14:1744806918817429
    [Google Scholar]
  129. 129.
    Ahlström FHG, Mätlik K, Viisanen H, Blomqvist KJ, Liu X et al. 2021. Spared nerve injury causes sexually dimorphic mechanical allodynia and differential gene expression in spinal cords and dorsal root ganglia in rats. Mol. Neurobiol. 58:5396–419
    [Google Scholar]
/content/journals/10.1146/annurev-pharmtox-051421-112259
Loading
/content/journals/10.1146/annurev-pharmtox-051421-112259
Loading

Data & Media loading...

Supplemental Material

Supplementary Data

  • 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