Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2022

01-12-2022 | Neuropathic Pain | Research

Activation of locus coeruleus-spinal cord noradrenergic neurons alleviates neuropathic pain in mice via reducing neuroinflammation from astrocytes and microglia in spinal dorsal horn

Authors: Juan Li, Yiyong Wei, Junli Zhou, Helin Zou, Lulin Ma, Chengxi Liu, Zhi Xiao, Xingfeng Liu, Xinran Tan, Tian Yu, Song Cao

Published in: Journal of Neuroinflammation | Issue 1/2022

Login to get access

Abstract

Background

The noradrenergic neurons of locus coeruleus (LC) project to the spinal dorsal horn (SDH), and release norepinephrine (NE) to inhibit pain transmission. However, its effect on pathological pain and the cellular mechanism in the SDH remains unclear. This study aimed to explore the analgesic effects and the anti-neuroinflammation mechanism of LC-spinal cord noradrenergic pathway (LC:SC) in neuropathic pain (NP) mice with sciatic chronic constriction injury.

Methods

The Designer Receptors Exclusively Activated by Designer Drugs (DREADD) was used to selectively activate LC:SC. Noradrenergic neuron-specific retro–adeno-associated virus was injected to the spinal cord. Pain threshold, LC and wide dynamic range (WDR) neuron firing, neuroinflammation (microglia and astrocyte activation, cytokine expression), and α2AR expression in SDH were evaluated.

Results

Activation of LC:SC with DREADD increased the mechanical and thermal nociceptive thresholds and reduced the WDR neuron firing. LC:SC activation (daily, 7 days) downregulated TNF-α and IL-1β expression, upregulated IL-4 and IL-10 expression in SDH, and inhibited microglia and astrocytes activation in NP mice. Immunofluorescence double staining confirmed that LC:SC activation decreased the expression of cytokines in microglia of the SDH. In addition, the effects of LC:SC activation could be reversed by intrathecal injection of yohimbine. Immunofluorescence of SDH showed that NE receptor α2B-AR was highly expressed in microglia in CCI mice.

Conclusion

These findings indicate that selective activation of LC:SC alleviates NP in mice by increasing the release of NE and reducing neuroinflammation of astrocytes and microglia in SDH.
Appendix
Available only for authorised users
Literature
1.
go back to reference Bouhassira D. Neuropathic pain: definition, assessment and epidemiology. Rev Neurol (Paris). 2019;175:16–25.CrossRef Bouhassira D. Neuropathic pain: definition, assessment and epidemiology. Rev Neurol (Paris). 2019;175:16–25.CrossRef
2.
go back to reference Finnerup NB, Haroutounian S, Kamerman P, Baron R, Bennett DLH, Bouhassira D, Cruccu G, Freeman R, Hansson P, Nurmikko T, et al. Neuropathic pain: an updated grading system for research and clinical practice. Pain. 2016;157:1599–606.PubMedPubMedCentralCrossRef Finnerup NB, Haroutounian S, Kamerman P, Baron R, Bennett DLH, Bouhassira D, Cruccu G, Freeman R, Hansson P, Nurmikko T, et al. Neuropathic pain: an updated grading system for research and clinical practice. Pain. 2016;157:1599–606.PubMedPubMedCentralCrossRef
3.
go back to reference Cao S, Li J, Yuan J, Zhang D, Yu T. Fast localization and sectioning of mouse locus coeruleus. Biomed Res Int. 2020;2020:4860735.PubMedPubMedCentral Cao S, Li J, Yuan J, Zhang D, Yu T. Fast localization and sectioning of mouse locus coeruleus. Biomed Res Int. 2020;2020:4860735.PubMedPubMedCentral
5.
go back to reference Llorca-Torralba M, Borges G, Neto F, Mico JA, Berrocoso E. Noradrenergic locus coeruleus pathways in pain modulation. Neuroscience. 2016;338:93–113.PubMedCrossRef Llorca-Torralba M, Borges G, Neto F, Mico JA, Berrocoso E. Noradrenergic locus coeruleus pathways in pain modulation. Neuroscience. 2016;338:93–113.PubMedCrossRef
7.
go back to reference Hayashida KI, Eisenach JC. Descending noradrenergic inhibition: an important mechanism of gabapentin analgesia in neuropathic pain. Adv Exp Med Biol. 2018;1099:93–100.PubMedCrossRef Hayashida KI, Eisenach JC. Descending noradrenergic inhibition: an important mechanism of gabapentin analgesia in neuropathic pain. Adv Exp Med Biol. 2018;1099:93–100.PubMedCrossRef
8.
go back to reference Hayashida KI, Kimuram M, Eisenach JC. Blockade of α2-adrenergic or metabotropic glutamate receptors induces glutamate release in the locus coeruleus to activate descending inhibition in rats with chronic neuropathic hypersensitivity. Neurosci Lett. 2018;676:41–5.PubMedPubMedCentralCrossRef Hayashida KI, Kimuram M, Eisenach JC. Blockade of α2-adrenergic or metabotropic glutamate receptors induces glutamate release in the locus coeruleus to activate descending inhibition in rats with chronic neuropathic hypersensitivity. Neurosci Lett. 2018;676:41–5.PubMedPubMedCentralCrossRef
9.
go back to reference Di Cesare ML, Micheli L, Crocetti L, Giovannoni MP, Vergelli C, Ghelardini C. α2 adrenoceptor: a target for neuropathic pain treatment. Mini Rev Med Chem. 2017;17:95–107. Di Cesare ML, Micheli L, Crocetti L, Giovannoni MP, Vergelli C, Ghelardini C. α2 adrenoceptor: a target for neuropathic pain treatment. Mini Rev Med Chem. 2017;17:95–107.
10.
go back to reference Seibt F, Schlichter R. Noradrenaline-mediated facilitation of inhibitory synaptic transmission in the dorsal horn of the rat spinal cord involves interlaminar communications. Eur J Neurosci. 2015;42:2654–65.PubMedCrossRef Seibt F, Schlichter R. Noradrenaline-mediated facilitation of inhibitory synaptic transmission in the dorsal horn of the rat spinal cord involves interlaminar communications. Eur J Neurosci. 2015;42:2654–65.PubMedCrossRef
11.
go back to reference Pluvinage JV, Haney MS, Smith BAH, Sun J, Iram T, Bonanno L, Li L, Lee DP, Morgens DW, Yang AC, et al. CD22 blockade restores homeostatic microglial phagocytosis in ageing brains. Nature. 2019;568:187–92.PubMedPubMedCentralCrossRef Pluvinage JV, Haney MS, Smith BAH, Sun J, Iram T, Bonanno L, Li L, Lee DP, Morgens DW, Yang AC, et al. CD22 blockade restores homeostatic microglial phagocytosis in ageing brains. Nature. 2019;568:187–92.PubMedPubMedCentralCrossRef
12.
go back to reference Nieman AN, Li G, Zahn NM, Mian MY, Mikulsky BN, Hoffman DA, Wilcox TM, Kehoe AS, Luecke IW, Poe MM, et al. Targeting nitric oxide production in microglia with novel imidazodiazepines for nonsedative pain treatment. ACS Chem Neurosci. 2020;11:2019–30.PubMedCrossRef Nieman AN, Li G, Zahn NM, Mian MY, Mikulsky BN, Hoffman DA, Wilcox TM, Kehoe AS, Luecke IW, Poe MM, et al. Targeting nitric oxide production in microglia with novel imidazodiazepines for nonsedative pain treatment. ACS Chem Neurosci. 2020;11:2019–30.PubMedCrossRef
13.
go back to reference Morioka N, Tanabe H, Inoue A, Dohi T, Nakata Y. Noradrenaline reduces the ATP-stimulated phosphorylation of p38 MAP kinase via beta-adrenergic receptors-cAMP-protein kinase A-dependent mechanism in cultured rat spinal microglia. Neurochem Int. 2009;55:226–34.PubMedCrossRef Morioka N, Tanabe H, Inoue A, Dohi T, Nakata Y. Noradrenaline reduces the ATP-stimulated phosphorylation of p38 MAP kinase via beta-adrenergic receptors-cAMP-protein kinase A-dependent mechanism in cultured rat spinal microglia. Neurochem Int. 2009;55:226–34.PubMedCrossRef
14.
go back to reference Caraci F, Merlo S, Drago F, Caruso G, Parenti C, Sortino MA. Rescue of noradrenergic system as a novel pharmacological strategy in the treatment of chronic pain: focus on microglia activation. Front Pharmacol. 2019;10:1024.PubMedPubMedCentralCrossRef Caraci F, Merlo S, Drago F, Caruso G, Parenti C, Sortino MA. Rescue of noradrenergic system as a novel pharmacological strategy in the treatment of chronic pain: focus on microglia activation. Front Pharmacol. 2019;10:1024.PubMedPubMedCentralCrossRef
15.
go back to reference Hinojosa AE, Caso JR, García-Bueno B, Leza JC, Madrigal JL. Dual effects of noradrenaline on astroglial production of chemokines and pro-inflammatory mediators. J Neuroinflammation. 2013;10:81.PubMedPubMedCentralCrossRef Hinojosa AE, Caso JR, García-Bueno B, Leza JC, Madrigal JL. Dual effects of noradrenaline on astroglial production of chemokines and pro-inflammatory mediators. J Neuroinflammation. 2013;10:81.PubMedPubMedCentralCrossRef
16.
go back to reference Kohro Y, Matsuda T, Yoshihara K, Kohno K, Koga K, Katsuragi R, Oka T, Tashima R, Muneta S, Yamane T, et al. Spinal astrocytes in superficial laminae gate brainstem descending control of mechanosensory hypersensitivity. Nat Neurosci. 2020;23:1376–87.PubMedCrossRef Kohro Y, Matsuda T, Yoshihara K, Kohno K, Koga K, Katsuragi R, Oka T, Tashima R, Muneta S, Yamane T, et al. Spinal astrocytes in superficial laminae gate brainstem descending control of mechanosensory hypersensitivity. Nat Neurosci. 2020;23:1376–87.PubMedCrossRef
18.
go back to reference Tsuda M. Microglia-mediated regulation of neuropathic pain: molecular and cellular mechanisms. Biol Pharm Bull. 2019;42:1959–68.PubMedCrossRef Tsuda M. Microglia-mediated regulation of neuropathic pain: molecular and cellular mechanisms. Biol Pharm Bull. 2019;42:1959–68.PubMedCrossRef
19.
go back to reference Sideris-Lampretsas G, Malcangio M. Microglial heterogeneity in chronic pain. Brain Behav Immun. 2021;96:279–89.PubMedCrossRef Sideris-Lampretsas G, Malcangio M. Microglial heterogeneity in chronic pain. Brain Behav Immun. 2021;96:279–89.PubMedCrossRef
20.
go back to reference Xiong XY, Liu L, Yang QW. Functions and mechanisms of microglia/macrophages in neuroinflammation and neurogenesis after stroke. Prog Neurobiol. 2016;142:23–44.PubMedCrossRef Xiong XY, Liu L, Yang QW. Functions and mechanisms of microglia/macrophages in neuroinflammation and neurogenesis after stroke. Prog Neurobiol. 2016;142:23–44.PubMedCrossRef
22.
go back to reference Cao S, Fisher DW, Rodriguez G, Yu T, Dong H. Comparisons of neuroinflammation, microglial activation, and degeneration of the locus coeruleus-norepinephrine system in APP/PS1 and aging mice. J Neuroinflammation. 2021;18:10.PubMedPubMedCentralCrossRef Cao S, Fisher DW, Rodriguez G, Yu T, Dong H. Comparisons of neuroinflammation, microglial activation, and degeneration of the locus coeruleus-norepinephrine system in APP/PS1 and aging mice. J Neuroinflammation. 2021;18:10.PubMedPubMedCentralCrossRef
23.
go back to reference National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animals. Guide for the Care and Use of Laboratory Animals. 8th ed. Washington (DC): National Academies Press (US); 2011. National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animals. Guide for the Care and Use of Laboratory Animals. 8th ed. Washington (DC): National Academies Press (US); 2011.
24.
go back to reference Cao S, Deng W, Li Y, Qin B, Zhang L, Yu S, Xie P, Xiao Z, Yu T. Chronic constriction injury of sciatic nerve changes circular RNA expression in rat spinal dorsal horn. J Pain Res. 2017;10:1687–96.PubMedPubMedCentralCrossRef Cao S, Deng W, Li Y, Qin B, Zhang L, Yu S, Xie P, Xiao Z, Yu T. Chronic constriction injury of sciatic nerve changes circular RNA expression in rat spinal dorsal horn. J Pain Res. 2017;10:1687–96.PubMedPubMedCentralCrossRef
25.
go back to reference Tervo DG, Hwang BY, Viswanathan S, Gaj T, Lavzin M, Ritola KD, Lindo S, Michael S, Kuleshova E, Ojala D, et al. A Designer AAV variant permits efficient retrograde access to projection neurons. Neuron. 2016;92:372–82.PubMedPubMedCentralCrossRef Tervo DG, Hwang BY, Viswanathan S, Gaj T, Lavzin M, Ritola KD, Lindo S, Michael S, Kuleshova E, Ojala D, et al. A Designer AAV variant permits efficient retrograde access to projection neurons. Neuron. 2016;92:372–82.PubMedPubMedCentralCrossRef
26.
go back to reference Zhang Q, Hu DX, He F, Li CY, Qi GJ, Cai HW, Li TX, Ming J, Zhang P, Chen XQ, Tian B. Locus coeruleus-CA1 projections are involved in chronic depressive stress-induced hippocampal vulnerability to transient global ischaemia. Nat Commun. 2019;10:2942.PubMedPubMedCentralCrossRef Zhang Q, Hu DX, He F, Li CY, Qi GJ, Cai HW, Li TX, Ming J, Zhang P, Chen XQ, Tian B. Locus coeruleus-CA1 projections are involved in chronic depressive stress-induced hippocampal vulnerability to transient global ischaemia. Nat Commun. 2019;10:2942.PubMedPubMedCentralCrossRef
27.
go back to reference François A, Low SA, Sypek EI, Christensen AJ, Sotoudeh C, Beier KT, Ramakrishnan C, Ritola KD, Sharif-Naeini R, Deisseroth K, et al. A brainstem-spinal cord inhibitory circuit for mechanical pain modulation by GABA and enkephalins. Neuron. 2017;93:822-839.e826.PubMedPubMedCentralCrossRef François A, Low SA, Sypek EI, Christensen AJ, Sotoudeh C, Beier KT, Ramakrishnan C, Ritola KD, Sharif-Naeini R, Deisseroth K, et al. A brainstem-spinal cord inhibitory circuit for mechanical pain modulation by GABA and enkephalins. Neuron. 2017;93:822-839.e826.PubMedPubMedCentralCrossRef
28.
go back to reference Cho C, Michailidis V, Lecker I, Collymore C, Hanwell D, Loka M, Danesh M, Pham C, Urban P, Bonin RP, Martin LJ. Evaluating analgesic efficacy and administration route following craniotomy in mice using the grimace scale. Sci Rep. 2019;9:359.PubMedPubMedCentralCrossRef Cho C, Michailidis V, Lecker I, Collymore C, Hanwell D, Loka M, Danesh M, Pham C, Urban P, Bonin RP, Martin LJ. Evaluating analgesic efficacy and administration route following craniotomy in mice using the grimace scale. Sci Rep. 2019;9:359.PubMedPubMedCentralCrossRef
29.
go back to reference Hylden JL, Wilcox GL. Intrathecal morphine in mice: a new technique. Eur J Pharmacol. 1980;67:313–6.PubMedCrossRef Hylden JL, Wilcox GL. Intrathecal morphine in mice: a new technique. Eur J Pharmacol. 1980;67:313–6.PubMedCrossRef
30.
go back to reference Huang Q, Sun ML, Chen Y, Li XY, Wang YX. Concurrent bullatine A enhances morphine antinociception and inhibits morphine antinociceptive tolerance by indirect activation of spinal κ-opioid receptors. J Ethnopharmacol. 2017;196:151–9.PubMedCrossRef Huang Q, Sun ML, Chen Y, Li XY, Wang YX. Concurrent bullatine A enhances morphine antinociception and inhibits morphine antinociceptive tolerance by indirect activation of spinal κ-opioid receptors. J Ethnopharmacol. 2017;196:151–9.PubMedCrossRef
31.
go back to reference Luo YJ, Li YD, Wang L, Yang SR, Yuan XS, Wang J, Cherasse Y, Lazarus M, Chen JF, Qu WM, Huang ZL. Nucleus accumbens controls wakefulness by a subpopulation of neurons expressing dopamine D(1) receptors. Nat Commun. 2018;9:1576.PubMedPubMedCentralCrossRef Luo YJ, Li YD, Wang L, Yang SR, Yuan XS, Wang J, Cherasse Y, Lazarus M, Chen JF, Qu WM, Huang ZL. Nucleus accumbens controls wakefulness by a subpopulation of neurons expressing dopamine D(1) receptors. Nat Commun. 2018;9:1576.PubMedPubMedCentralCrossRef
32.
go back to reference Ahsan MZ, Zhao MJ, Shoaib RM, Zhang Y, Wang YX. Comparative study of dezocine, pentazocine and tapentadol on antinociception and physical dependence. Life Sci. 2021;285: 119996.PubMedCrossRef Ahsan MZ, Zhao MJ, Shoaib RM, Zhang Y, Wang YX. Comparative study of dezocine, pentazocine and tapentadol on antinociception and physical dependence. Life Sci. 2021;285: 119996.PubMedCrossRef
33.
go back to reference Wang H, Huang M, Wang W, Zhang Y, Ma X, Luo L, Xu X, Xu L, Shi H, Xu Y, et al. Microglial TLR4-induced TAK1 phosphorylation and NLRP3 activation mediates neuroinflammation and contributes to chronic morphine-induced antinociceptive tolerance. Pharmacol Res. 2021;165: 105482.PubMedCrossRef Wang H, Huang M, Wang W, Zhang Y, Ma X, Luo L, Xu X, Xu L, Shi H, Xu Y, et al. Microglial TLR4-induced TAK1 phosphorylation and NLRP3 activation mediates neuroinflammation and contributes to chronic morphine-induced antinociceptive tolerance. Pharmacol Res. 2021;165: 105482.PubMedCrossRef
35.
go back to reference Otsubo Y, Satoh Y, Kodama M, Araki Y, Satomoto M, Sakamoto E, Pagès G, Pouysségur J, Endo S, Kazama T. Mechanical allodynia but not thermal hyperalgesia is impaired in mice deficient for ERK2 in the central nervous system. Pain. 2012;153:2241–52.PubMedCrossRef Otsubo Y, Satoh Y, Kodama M, Araki Y, Satomoto M, Sakamoto E, Pagès G, Pouysségur J, Endo S, Kazama T. Mechanical allodynia but not thermal hyperalgesia is impaired in mice deficient for ERK2 in the central nervous system. Pain. 2012;153:2241–52.PubMedCrossRef
36.
go back to reference Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods. 1994;53:55–63.PubMedCrossRef Chaplan SR, Bach FW, Pogrel JW, Chung JM, Yaksh TL. Quantitative assessment of tactile allodynia in the rat paw. J Neurosci Methods. 1994;53:55–63.PubMedCrossRef
37.
go back to reference Yuan SB, Ji G, Li B, Andersson T, Neugebauer V, Tang SJ. A Wnt5a signaling pathway in the pathogenesis of HIV-1 gp120-induced pain. Pain. 2015;156:1311–9.PubMedPubMedCentralCrossRef Yuan SB, Ji G, Li B, Andersson T, Neugebauer V, Tang SJ. A Wnt5a signaling pathway in the pathogenesis of HIV-1 gp120-induced pain. Pain. 2015;156:1311–9.PubMedPubMedCentralCrossRef
38.
go back to reference Caputi FF, Nicora M, Simeone R, Candeletti S, Romualdi P. Tapentadol: an analgesic that differs from classic opioids due to its noradrenergic mechanism of action. Minerva Med. 2019;110:62–78.PubMedCrossRef Caputi FF, Nicora M, Simeone R, Candeletti S, Romualdi P. Tapentadol: an analgesic that differs from classic opioids due to its noradrenergic mechanism of action. Minerva Med. 2019;110:62–78.PubMedCrossRef
40.
go back to reference Pertovaara A. The noradrenergic pain regulation system: a potential target for pain therapy. Eur J Pharmacol. 2013;716:2–7.PubMedCrossRef Pertovaara A. The noradrenergic pain regulation system: a potential target for pain therapy. Eur J Pharmacol. 2013;716:2–7.PubMedCrossRef
41.
go back to reference Maeda M, Tsuruoka M, Hayashi B, Nagasawa I, Inoue T. Descending pathways from activated locus coeruleus/subcoeruleus following unilateral hindpaw inflammation in the rat. Brain Res Bull. 2009;78:170–4.PubMedCrossRef Maeda M, Tsuruoka M, Hayashi B, Nagasawa I, Inoue T. Descending pathways from activated locus coeruleus/subcoeruleus following unilateral hindpaw inflammation in the rat. Brain Res Bull. 2009;78:170–4.PubMedCrossRef
42.
go back to reference Tsuruoka M, Hitoto T, Hiruma Y, Matsui Y. Neurochemical evidence for inflammation-induced activation of the coeruleospinal modulation system in the rat. Brain Res. 1999;821:236–40.PubMedCrossRef Tsuruoka M, Hitoto T, Hiruma Y, Matsui Y. Neurochemical evidence for inflammation-induced activation of the coeruleospinal modulation system in the rat. Brain Res. 1999;821:236–40.PubMedCrossRef
43.
go back to reference Hughes SW, Hickey L, Hulse RP, Lumb BM, Pickering AE. Endogenous analgesic action of the pontospinal noradrenergic system spatially restricts and temporally delays the progression of neuropathic pain following tibial nerve injury. Pain. 2013;154:1680–90.PubMedPubMedCentralCrossRef Hughes SW, Hickey L, Hulse RP, Lumb BM, Pickering AE. Endogenous analgesic action of the pontospinal noradrenergic system spatially restricts and temporally delays the progression of neuropathic pain following tibial nerve injury. Pain. 2013;154:1680–90.PubMedPubMedCentralCrossRef
44.
go back to reference Parent AJ, Tétreault P, Roux M, Belleville K, Longpré JM, Beaudet N, Goffaux P, Sarret P. Descending nociceptive inhibition is modulated in a time-dependent manner in a double-hit model of chronic/tonic pain. Neuroscience. 2016;315:70–8.PubMedCrossRef Parent AJ, Tétreault P, Roux M, Belleville K, Longpré JM, Beaudet N, Goffaux P, Sarret P. Descending nociceptive inhibition is modulated in a time-dependent manner in a double-hit model of chronic/tonic pain. Neuroscience. 2016;315:70–8.PubMedCrossRef
45.
go back to reference Wei H, Jin CY, Viisanen H, You HJ, Pertovaara A. Histamine in the locus coeruleus promotes descending noradrenergic inhibition of neuropathic hypersensitivity. Pharmacol Res. 2014;90:58–66.PubMedCrossRef Wei H, Jin CY, Viisanen H, You HJ, Pertovaara A. Histamine in the locus coeruleus promotes descending noradrenergic inhibition of neuropathic hypersensitivity. Pharmacol Res. 2014;90:58–66.PubMedCrossRef
46.
go back to reference Li N, Li C, Han R, Wang Y, Yang M, Wang H, Tian J. LPM580098, a novel triple reuptake inhibitor of serotonin, noradrenaline, and dopamine attenuates neuropathic pain. Front Pharmacol. 2019;10:53.PubMedPubMedCentralCrossRef Li N, Li C, Han R, Wang Y, Yang M, Wang H, Tian J. LPM580098, a novel triple reuptake inhibitor of serotonin, noradrenaline, and dopamine attenuates neuropathic pain. Front Pharmacol. 2019;10:53.PubMedPubMedCentralCrossRef
47.
48.
go back to reference Oladosu FA, Maixner W, Nackley AG. Alternative splicing of G protein-coupled receptors: relevance to pain management. Mayo Clin Proc. 2015;90:1135–51.PubMedCrossRef Oladosu FA, Maixner W, Nackley AG. Alternative splicing of G protein-coupled receptors: relevance to pain management. Mayo Clin Proc. 2015;90:1135–51.PubMedCrossRef
49.
go back to reference Coull JA, Beggs S, Boudreau D, Boivin D, Tsuda M, Inoue K, Gravel C, Salter MW, De Koninck Y. BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain. Nature. 2005;438:1017–21.PubMedCrossRef Coull JA, Beggs S, Boudreau D, Boivin D, Tsuda M, Inoue K, Gravel C, Salter MW, De Koninck Y. BDNF from microglia causes the shift in neuronal anion gradient underlying neuropathic pain. Nature. 2005;438:1017–21.PubMedCrossRef
50.
go back to reference Hayashida K, Eisenach JC. A tropomyosine receptor kinase inhibitor blocks spinal neuroplasticity essential for the anti-hypersensitivity effects of gabapentin and clonidine in rats with peripheral nerve injury. J Pain. 2011;12:94–100.PubMedPubMedCentralCrossRef Hayashida K, Eisenach JC. A tropomyosine receptor kinase inhibitor blocks spinal neuroplasticity essential for the anti-hypersensitivity effects of gabapentin and clonidine in rats with peripheral nerve injury. J Pain. 2011;12:94–100.PubMedPubMedCentralCrossRef
51.
go back to reference Kawanabe R, Yoshihara K, Hatada I, Tsuda M. Activation of spinal dorsal horn astrocytes by noxious stimuli involves descending noradrenergic signaling. Mol Brain. 2021;14:79.PubMedPubMedCentralCrossRef Kawanabe R, Yoshihara K, Hatada I, Tsuda M. Activation of spinal dorsal horn astrocytes by noxious stimuli involves descending noradrenergic signaling. Mol Brain. 2021;14:79.PubMedPubMedCentralCrossRef
52.
go back to reference Chen Z, Doyle TM, Luongo L, Largent-Milnes TM, Giancotti LA, Kolar G, Squillace S, Boccella S, Walker JK, Pendleton A, et al. Sphingosine-1-phosphate receptor 1 activation in astrocytes contributes to neuropathic pain. Proc Natl Acad Sci U S A. 2019;116:10557–62.PubMedPubMedCentralCrossRef Chen Z, Doyle TM, Luongo L, Largent-Milnes TM, Giancotti LA, Kolar G, Squillace S, Boccella S, Walker JK, Pendleton A, et al. Sphingosine-1-phosphate receptor 1 activation in astrocytes contributes to neuropathic pain. Proc Natl Acad Sci U S A. 2019;116:10557–62.PubMedPubMedCentralCrossRef
53.
go back to reference Arora V, Morado-Urbina CE, Aschenbrenner CA, Hayashida K, Wang F, Martin TJ, Eisenach JC, Peters CM. Disruption of spinal noradrenergic activation delays recovery of acute incision-induced hypersensitivity and increases spinal glial activation in the rat. J Pain. 2016;17:190–202.PubMedCrossRef Arora V, Morado-Urbina CE, Aschenbrenner CA, Hayashida K, Wang F, Martin TJ, Eisenach JC, Peters CM. Disruption of spinal noradrenergic activation delays recovery of acute incision-induced hypersensitivity and increases spinal glial activation in the rat. J Pain. 2016;17:190–202.PubMedCrossRef
54.
go back to reference Chen G, Park CK, Xie RG, Berta T, Nedergaard M, Ji RR. Connexin-43 induces chemokine release from spinal cord astrocytes to maintain late-phase neuropathic pain in mice. Brain. 2014;137:2193–209.PubMedPubMedCentralCrossRef Chen G, Park CK, Xie RG, Berta T, Nedergaard M, Ji RR. Connexin-43 induces chemokine release from spinal cord astrocytes to maintain late-phase neuropathic pain in mice. Brain. 2014;137:2193–209.PubMedPubMedCentralCrossRef
55.
go back to reference Ma L, Li J, Zhou J, Zhang D, Xiao Z, Yu T, Li Y, Cao S. Intravenous lidocaine alleviates postherpetic neuralgia in rats via regulation of neuroinflammation of microglia and astrocytes. iScience. 2021;24:102108.PubMedPubMedCentralCrossRef Ma L, Li J, Zhou J, Zhang D, Xiao Z, Yu T, Li Y, Cao S. Intravenous lidocaine alleviates postherpetic neuralgia in rats via regulation of neuroinflammation of microglia and astrocytes. iScience. 2021;24:102108.PubMedPubMedCentralCrossRef
56.
go back to reference Zhang ZJ, Jiang BC, Gao YJ. Chemokines in neuron-glial cell interaction and pathogenesis of neuropathic pain. Cell Mol Life Sci. 2017;74:3275–91.PubMedCrossRef Zhang ZJ, Jiang BC, Gao YJ. Chemokines in neuron-glial cell interaction and pathogenesis of neuropathic pain. Cell Mol Life Sci. 2017;74:3275–91.PubMedCrossRef
57.
go back to reference Tsuda M. P2 receptors, microglial cytokines and chemokines, and neuropathic pain. J Neurosci Res. 2017;95:1319–29.PubMedCrossRef Tsuda M. P2 receptors, microglial cytokines and chemokines, and neuropathic pain. J Neurosci Res. 2017;95:1319–29.PubMedCrossRef
58.
go back to reference Li SS, Zhang WS, Ji D, Zhou YL, Li H, Yang JL, Xiong YC, Zhang YQ, Xu H. Involvement of spinal microglia and interleukin-18 in the anti-nociceptive effect of dexmedetomidine in rats subjected to CCI. Neurosci Lett. 2014;560:21–5.PubMedCrossRef Li SS, Zhang WS, Ji D, Zhou YL, Li H, Yang JL, Xiong YC, Zhang YQ, Xu H. Involvement of spinal microglia and interleukin-18 in the anti-nociceptive effect of dexmedetomidine in rats subjected to CCI. Neurosci Lett. 2014;560:21–5.PubMedCrossRef
59.
go back to reference Xu B, Zhang WS, Yang JL, Lû N, Deng XM, Xu H, Zhang YQ. Evidence for suppression of spinal glial activation by dexmedetomidine in a rat model of monoarthritis. Clin Exp Pharmacol Physiol. 2010;37:e158-166.PubMedCrossRef Xu B, Zhang WS, Yang JL, Lû N, Deng XM, Xu H, Zhang YQ. Evidence for suppression of spinal glial activation by dexmedetomidine in a rat model of monoarthritis. Clin Exp Pharmacol Physiol. 2010;37:e158-166.PubMedCrossRef
60.
go back to reference Choi S, Yamada A, Kim W, Kim SK, Furue H. Noradrenergic inhibition of spinal hyperexcitation elicited by cutaneous cold stimuli in rats with oxaliplatin-induced allodynia: electrophysiological and behavioral assessments. J Physiol Sci. 2017;67:431–8.PubMedCrossRef Choi S, Yamada A, Kim W, Kim SK, Furue H. Noradrenergic inhibition of spinal hyperexcitation elicited by cutaneous cold stimuli in rats with oxaliplatin-induced allodynia: electrophysiological and behavioral assessments. J Physiol Sci. 2017;67:431–8.PubMedCrossRef
Metadata
Title
Activation of locus coeruleus-spinal cord noradrenergic neurons alleviates neuropathic pain in mice via reducing neuroinflammation from astrocytes and microglia in spinal dorsal horn
Authors
Juan Li
Yiyong Wei
Junli Zhou
Helin Zou
Lulin Ma
Chengxi Liu
Zhi Xiao
Xingfeng Liu
Xinran Tan
Tian Yu
Song Cao
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2022
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-022-02489-9

Other articles of this Issue 1/2022

Journal of Neuroinflammation 1/2022 Go to the issue