Mini-Review Article

The Therapeutic Potential of Chemokines in the Treatment of Chemotherapy- Induced Peripheral Neuropathy

Author(s): Lin Zhou, Luyao Ao, Yunyi Yan, Wanting Li, Anqi Ye, Yahui Hu*, Weirong Fang* and Yunman Li*

Volume 21, Issue 3, 2020

Page: [288 - 301] Pages: 14

DOI: 10.2174/1389450120666190906153652

Price: $65

Abstract

Background: Some of the current challenges and complications of cancer therapy are chemotherapy- induced peripheral neuropathy (CIPN) and the neuropathic pain that are associated with this condition. Many major chemotherapeutic agents can cause neurotoxicity, significantly modulate the immune system and are always accompanied by various adverse effects. Recent evidence suggests that cross-talk occurs between the nervous system and the immune system during treatment with chemotherapeutic agents; thus, an emerging concept is that neuroinflammation is one of the major mechanisms underlying CIPN, as demonstrated by the upregulation of chemokines. Chemokines were originally identified as regulators of peripheral immune cell trafficking, and chemokines are also expressed on neurons and glial cells in the central nervous system.

Objective: In this review, we collected evidence demonstrating that chemokines are potential mediators and contributors to pain signalling in CIPN. The expression of chemokines and their receptors, such as CX3CL1/CX3CR1, CCL2/CCR2, CXCL1/CXCR2, CXCL12/CXCR4 and CCL3/CCR5, is altered in the pathological conditions of CIPN, and chemokine receptor antagonists attenuate neuropathic pain behaviour.

Conclusion: By understanding the mechanisms of chemokine-mediated communication, we may reveal chemokine targets that can be used as novel therapeutic strategies for the treatment of CIPN.

Keywords: Chemotherapy-induced peripheral neuropathy, chemotherapeutic agents, chemokines, neuroinflammation, CCL2, CX3CL1, CXCL1.

Graphical Abstract
[1]
Marmiroli P, Scuteri A, Cornblath DR, Cavaletti G. Pain in chemotherapy-induced peripheral neurotoxicity. J Peripher Nerv Syst 2017; 22(3): 156-61.
[http://dx.doi.org/10.1111/jns.12226] [PMID: 28600844]
[2]
Brewer JR, Morrison G, Dolan ME, Fleming GF. Chemotherapy-induced peripheral neuropathy: Current status and progress. Gynecol Oncol 2016; 140(1): 176-83.
[http://dx.doi.org/10.1016/j.ygyno.2015.11.011] [PMID: 26556766]
[3]
Fallon MT. Neuropathic pain in cancer. Br J Anaesth 2013; 111(1): 105-11.
[http://dx.doi.org/10.1093/bja/aet208] [PMID: 23794652]
[4]
Ma J, Kavelaars A, Dougherty PM, Heijnen CJ. Beyond symptomatic relief for chemotherapy-induced peripheral neuropathy: Targeting the source. Cancer 2018; 124(11): 2289-98.
[http://dx.doi.org/10.1002/cncr.31248] [PMID: 29461625]
[5]
Seretny M, Currie GL, Sena ES, et al. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: A systematic review and meta-analysis. Pain 2014; 155(12): 2461-70.
[http://dx.doi.org/10.1016/j.pain.2014.09.020] [PMID: 25261162]
[6]
Miltenburg NC, Boogerd W. Chemotherapy-induced neuropathy: A comprehensive survey. Cancer Treat Rev 2014; 40(7): 872-82.
[http://dx.doi.org/10.1016/j.ctrv.2014.04.004] [PMID: 24830939]
[7]
Hama A, Takamatsu H. Chemotherapy-Induced Peripheral Neuropathic Pain and Rodent Models. CNS Neurol Disord Drug Targets 2016; 15(1): 7-19.
[http://dx.doi.org/10.2174/1871527315666151110125325] [PMID: 26553161]
[8]
Banach M, Juranek JK, Zygulska AL. Chemotherapy-induced neuropathies-a growing problem for patients and health care providers. Brain Behav 2016; 7(1)e00558
[http://dx.doi.org/10.1002/brb3.558] [PMID: 28127506]
[9]
Starobova H, Vetter I. Pathophysiology of chemotherapy-induced peripheral neuropathy. Front Mol Neurosci 2017; 10: 174.
[http://dx.doi.org/10.3389/fnmol.2017.00174] [PMID: 28620280]
[10]
Zajączkowska R, Kocot-Kępska M, Leppert W, Wrzosek A, Mika J, Wordliczek J. Mechanisms of chemotherapy-induced peripheral neuropathy. Int J Mol Sci 2019; 20(6)E1451
[http://dx.doi.org/10.3390/ijms20061451] [PMID: 30909387]
[11]
Waseem M, Kaushik P, Tabassum H, Parvez S. Role of mitochondrial mechanism in chemotherapy-induced peripheral neuropathy. Curr Drug Metab 2018; 19(1): 47-54.
[http://dx.doi.org/10.2174/1389200219666171207121313] [PMID: 29219049]
[12]
Han Y, Smith MT. Pathobiology of cancer chemotherapy-induced peripheral neuropathy (CIPN). Front Pharmacol 2013; 4: 156.
[http://dx.doi.org/10.3389/fphar.2013.00156] [PMID: 24385965]
[13]
Jaggi AS, Singh N. Mechanisms in cancer-chemotherapeutic drugs-induced peripheral neuropathy. Toxicology 2012; 291(1-3): 1-9.
[http://dx.doi.org/10.1016/j.tox.2011.10.019] [PMID: 22079234]
[14]
Authier N, Balayssac D, Marchand F, et al. Animal models of chemotherapy-evoked painful peripheral neuropathies. Neurotherapeutics 2009; 6(4): 620-9.
[http://dx.doi.org/10.1016/j.nurt.2009.07.003] [PMID: 19789067]
[15]
Boyle FM, Beatson C, Monk R, Grant SL, Kurek JB. The experimental neuroprotectant leukaemia inhibitory factor (LIF) does not compromise antitumour activity of paclitaxel, cisplatin and carboplatin. Cancer Chemother Pharmacol 2001; 48(6): 429-34.
[http://dx.doi.org/10.1007/s00280-001-0382-6] [PMID: 11800022]
[16]
Boyle FM, Wheeler HR, Shenfield GM. Amelioration of experimental cisplatin and paclitaxel neuropathy with glutamate. J Neurooncol 1999; 41(2): 107-16.
[http://dx.doi.org/10.1023/A:1006124917643] [PMID: 10222430]
[17]
Flatters SJL, Dougherty PM, Colvin LA. clinical and preclinical perspectives on chemotherapy-induced peripheral neuropathy (CIPN): a narrative review. Br J Anaesth 2017; 119(4): 737-49.
[http://dx.doi.org/10.1093/bja/aex229] [PMID: 29121279]
[18]
Cioroiu C, Weimer LH. Update on Chemotherapy-Induced Peripheral Neuropathy. Curr Neurol Neurosci Rep 2017; 17(6): 47.
[http://dx.doi.org/10.1007/s11910-017-0757-7] [PMID: 28421360]
[19]
Di Cesare Mannelli L, Zanardelli M, Landini I, et al. Effect of the SOD mimetic MnL4 on in vitro and in vivo oxaliplatin toxicity: Possible aid in chemotherapy induced neuropathy. Free Radic Biol Med 2016; 93: 67-76.
[http://dx.doi.org/10.1016/j.freeradbiomed.2016.01.023] [PMID: 26828020]
[20]
Storey DJ, Sakala M, McLean CM, et al. Capecitabine combined with oxaliplatin (CapOx) in clinical practice: how significant is peripheral neuropathy? Ann Oncol 2010; 21(8): 1657-61.
[http://dx.doi.org/10.1093/annonc/mdp594] [PMID: 20089559]
[21]
Carvalho LF, Silva AMF, Carvalho AA. The use of antioxidant agents for chemotherapy-induced peripheral neuropathy treatment in animal models. Clin Exp Pharmacol Physiol 2017; 44(10): 971-9.
[http://dx.doi.org/10.1111/1440-1681.12803] [PMID: 28649767]
[22]
Babu A, Prasanth KG, Balaji B. Effect of curcumin in mice model of vincristine-induced neuropathy. Pharm Biol 2015; 53(6): 838-48.
[http://dx.doi.org/10.3109/13880209.2014.943247] [PMID: 25429779]
[23]
Topp KS, Tanner KD, Levine JD. Damage to the cytoskeleton of large diameter sensory neurons and myelinated axons in vincristine-induced painful peripheral neuropathy in the rat. J Comp Neurol 2000; 424(4): 563-76.
[http://dx.doi.org/10.1002/1096-9861(20000904)424:4<563::AID-CNE1>3.0.CO;2-U] [PMID: 10931481]
[24]
Lopus M, Smiyun G, Miller H, Oroudjev E, Wilson L, Jordan MA. Mechanism of action of ixabepilone and its interactions with the βIII-tubulin isotype. Cancer Chemother Pharmacol 2015; 76(5): 1013-24.
[http://dx.doi.org/10.1007/s00280-015-2863-z] [PMID: 26416565]
[25]
Vahdat LT, Thomas ES, Roché HH, et al. Ixabepilone-associated peripheral neuropathy: data from across the phase II and III clinical trials. Support Care Cancer 2012; 20(11): 2661-8.
[http://dx.doi.org/10.1007/s00520-012-1384-0] [PMID: 22382588]
[26]
Thawani SP, Tanji K, De Sousa EA, Weimer LH, Brannagan TH III. Bortezomib-associated demyelinating neuropathy--clinical and pathologic features. J Clin Neuromuscul Dis 2015; 16(4): 202-9.
[http://dx.doi.org/10.1097/CND.0000000000000077] [PMID: 25996966]
[27]
Farquhar-Smith P. Chemotherapy-induced neuropathic pain. Curr Opin Support Palliat Care 2011; 5(1): 1-7.
[http://dx.doi.org/10.1097/SPC.0b013e328342f9cc] [PMID: 21192267]
[28]
Yared JA, Tkaczuk KH. Update on taxane development: new analogs and new formulations. Drug Des Devel Ther 2012; 6: 371-84.
[PMID: 23251087]
[29]
De Iuliis F, Taglieri L, Salerno G, Lanza R, Scarpa S. Taxane induced neuropathy in patients affected by breast cancer: Literature review. Crit Rev Oncol Hematol 2015; 96(1): 34-45.
[http://dx.doi.org/10.1016/j.critrevonc.2015.04.011] [PMID: 26004917]
[30]
Richardson P, Hideshima T, Anderson K. Thalidomide in multiple myeloma. Biomed Pharmacother 2002; 56(3): 115-28.
[http://dx.doi.org/10.1016/S0753-3322(02)00168-3] [PMID: 12046682]
[31]
Morawska M, Grzasko N, Kostyra M, Wojciechowicz J, Hus M. Therapy-related peripheral neuropathy in multiple myeloma patients. Hematol Oncol 2015; 33(4): 113-9.
[http://dx.doi.org/10.1002/hon.2149] [PMID: 25399783]
[32]
Gao YJ, Zhang L, Samad OA, et al. JNK-induced MCP-1 production in spinal cord astrocytes contributes to central sensitization and neuropathic pain. J Neurosci 2009; 29(13): 4096-108.
[http://dx.doi.org/10.1523/JNEUROSCI.3623-08.2009] [PMID: 19339605]
[33]
White FA, Jung H, Miller RJ. Chemokines and the pathophysiology of neuropathic pain. Proc Natl Acad Sci USA 2007; 104(51): 20151-8.
[http://dx.doi.org/10.1073/pnas.0709250104] [PMID: 18083844]
[34]
Moser B, Wolf M, Walz A, Loetscher P. Chemokines: multiple levels of leukocyte migration control. Trends Immunol 2004; 25(2): 75-84.
[http://dx.doi.org/10.1016/j.it.2003.12.005] [PMID: 15102366]
[35]
Bajetto A, Bonavia R, Barbero S, Schettini G. Characterization of chemokines and their receptors in the central nervous system: physiopathological implications. J Neurochem 2002; 82(6): 1311-29.
[http://dx.doi.org/10.1046/j.1471-4159.2002.01091.x] [PMID: 12354279]
[36]
Zlotnik A, Yoshie O. Chemokines: a new classification system and their role in immunity. Immunity 2000; 12(2): 121-7.
[http://dx.doi.org/10.1016/S1074-7613(00)80165-X] [PMID: 10714678]
[37]
Gao YJ, Ji RR. Chemokines, neuronal-glial interactions, and central processing of neuropathic pain. Pharmacol Ther 2010; 126(1): 56-68.
[http://dx.doi.org/10.1016/j.pharmthera.2010.01.002] [PMID: 20117131]
[38]
Zhang ZJ, Jiang BC, Gao YJ. Chemokines in neuron-glial cell interaction and pathogenesis of neuropathic pain. Cell Mol Life Sci 2017; 74(18): 3275-91.
[http://dx.doi.org/10.1007/s00018-017-2513-1] [PMID: 28389721]
[39]
Murphy PM, Baggiolini M, Charo IF, et al. International union of pharmacology. XXII. Nomenclature for chemokine receptors. Pharmacol Rev 2000; 52(1): 145-76.
[PMID: 10699158]
[40]
Schall TJ, Proudfoot AE. Overcoming hurdles in developing successful drugs targeting chemokine receptors. Nat Rev Immunol 2011; 11(5): 355-63.
[http://dx.doi.org/10.1038/nri2972] [PMID: 21494268]
[41]
Old EA, Malcangio M. Chemokine mediated neuron-glia communication and aberrant signalling in neuropathic pain states. Curr Opin Pharmacol 2012; 12(1): 67-73.
[http://dx.doi.org/10.1016/j.coph.2011.10.015] [PMID: 22056024]
[42]
Rossi D, Zlotnik A. The biology of chemokines and their receptors. Annu Rev Immunol 2000; 18: 217-42.
[http://dx.doi.org/10.1146/annurev.immunol.18.1.217] [PMID: 10837058]
[43]
Limatola C, Giovannelli A, Maggi L, et al. SDF-1alpha-mediated modulation of synaptic transmission in rat cerebellum. Eur J Neurosci 2000; 12(7): 2497-504.
[http://dx.doi.org/10.1046/j.1460-9568.2000.00139.x] [PMID: 10947825]
[44]
Bonecchi R, Galliera E, Borroni EM, Corsi MM, Locati M, Mantovani A. Chemokines and chemokine receptors: an overview. Front Biosci 2009; 14: 540-51.
[http://dx.doi.org/10.2741/3261] [PMID: 19273084]
[45]
Oh SB, Tran PB, Gillard SE, Hurley RW, Hammond DL, Miller RJ. Chemokines and glycoprotein120 produce pain hypersensitivity by directly exciting primary nociceptive neurons. J Neurosci 2001; 21(14): 5027-35.
[http://dx.doi.org/10.1523/JNEUROSCI.21-14-05027.2001] [PMID: 11438578]
[46]
Abbadie C, Lindia JA, Cumiskey AM, et al. Impaired neuropathic pain responses in mice lacking the chemokine receptor CCR2. Proc Natl Acad Sci USA 2003; 100(13): 7947-52.
[http://dx.doi.org/10.1073/pnas.1331358100] [PMID: 12808141]
[47]
Abbadie C. Chemokines, chemokine receptors and pain. Trends Immunol 2005; 26(10): 529-34.
[http://dx.doi.org/10.1016/j.it.2005.08.001] [PMID: 16099720]
[48]
Kiguchi N, Kobayashi Y, Maeda T, Saika F, Kishioka S. CC-chemokine MIP-1α in the spinal cord contributes to nerve injury-induced neuropathic pain. Neurosci Lett 2010; 484(1): 17-21.
[http://dx.doi.org/10.1016/j.neulet.2010.07.085] [PMID: 20692319]
[49]
White FA, Bhangoo SK, Miller RJ. Chemokines: integrators of pain and inflammation. Nat Rev Drug Discov 2005; 4(10): 834-44.
[http://dx.doi.org/10.1038/nrd1852] [PMID: 16224455]
[50]
Manjavachi MN, Quintão NL, Campos MM, et al. The effects of the selective and non-peptide CXCR2 receptor antagonist SB225002 on acute and long-lasting models of nociception in mice. Eur J Pain 2010; 14(1): 23-31.
[http://dx.doi.org/10.1016/j.ejpain.2009.01.007] [PMID: 19264522]
[51]
Rittner HL, Labuz D, Schaefer M, et al. Pain control by CXCR2 ligands through Ca2+-regulated release of opioid peptides from polymorphonuclear cells. FASEB J 2006; 20(14): 2627-9.
[http://dx.doi.org/10.1096/fj.06-6077fje] [PMID: 17060402]
[52]
Bennett GJ, Xie YK. A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 1988; 33(1): 87-107.
[http://dx.doi.org/10.1016/0304-3959(88)90209-6] [PMID: 2837713]
[53]
Decosterd I, Woolf CJ. Spared nerve injury: an animal model of persistent peripheral neuropathic pain. Pain 2000; 87(2): 149-58.
[http://dx.doi.org/10.1016/S0304-3959(00)00276-1] [PMID: 10924808]
[54]
Kim SH, Chung JM. An experimental model for peripheral neuropathy produced by segmental spinal nerve ligation in the rat. Pain 1992; 50(3): 355-63.
[http://dx.doi.org/10.1016/0304-3959(92)90041-9] [PMID: 1333581]
[55]
Seltzer Z, Dubner R, Shir Y. A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain 1990; 43(2): 205-18.
[http://dx.doi.org/10.1016/0304-3959(90)91074-S] [PMID: 1982347]
[56]
Hu SJ, Xing JL. An experimental model for chronic compression of dorsal root ganglion produced by intervertebral foramen stenosis in the rat. Pain 1998; 77(1): 15-23.
[http://dx.doi.org/10.1016/S0304-3959(98)00067-0] [PMID: 9755014]
[57]
Van Coillie E, Van Damme J, Opdenakker G. The MCP/eotaxin subfamily of CC chemokines. Cytokine Growth Factor Rev 1999; 10(1): 61-86.
[http://dx.doi.org/10.1016/S1359-6101(99)00005-2] [PMID: 10379912]
[58]
Kurihara T, Bravo R. Cloning and functional expression of mCCR2, a murine receptor for the C-C chemokines JE and FIC. J Biol Chem 1996; 271(20): 11603-7.
[http://dx.doi.org/10.1074/jbc.271.20.11603] [PMID: 8662823]
[59]
Abbadie C, Bhangoo S, De Koninck Y, Malcangio M, Melik-Parsadaniantz S, White FA. Chemokines and pain mechanisms. Brain Res Brain Res Rev 2009; 60(1): 125-34.
[http://dx.doi.org/10.1016/j.brainresrev.2008.12.002] [PMID: 19146875]
[60]
Bhangoo SK, Ripsch MS, Buchanan DJ, Miller RJ, White FA. Increased chemokine signaling in a model of HIV1-associated peripheral neuropathy. Mol Pain 2009; 5: 48.
[http://dx.doi.org/10.1186/1744-8069-5-48] [PMID: 19674450]
[61]
Guo W, Wang H, Zou S, Dubner R, Ren K. Chemokine signaling involving chemokine (C-C motif) ligand 2 plays a role in descending pain facilitation. Neurosci Bull 2012; 28(2): 193-207.
[http://dx.doi.org/10.1007/s12264-012-1218-6] [PMID: 22466130]
[62]
Van Steenwinckel J, Reaux-Le Goazigo A, Pommier B, et al. CCL2 released from neuronal synaptic vesicles in the spinal cord is a major mediator of local inflammation and pain after peripheral nerve injury. J Neurosci 2011; 31(15): 5865-75.
[http://dx.doi.org/10.1523/JNEUROSCI.5986-10.2011] [PMID: 21490228]
[63]
Zhang ZJ, Dong YL, Lu Y, Cao S, Zhao ZQ, Gao YJ. Chemokine CCL2 and its receptor CCR2 in the medullary dorsal horn are involved in trigeminal neuropathic pain. J Neuroinflammation 2012; 9: 136.
[http://dx.doi.org/10.1186/1742-2094-9-136] [PMID: 22721162]
[64]
Gornstein E, Schwarz TL. The paradox of paclitaxel neurotoxicity: Mechanisms and unanswered questions. Neuropharmacology 2014; 76(Pt A): 175-83.
[65]
Park SB, Goldstein D, Krishnan AV, et al. Chemotherapy-induced peripheral neurotoxicity: a critical analysis. CA Cancer J Clin 2013; 63(6): 419-37.
[http://dx.doi.org/10.3322/caac.21204] [PMID: 24590861]
[66]
McWhinney SR, Goldberg RM, McLeod HL. Platinum neurotoxicity pharmacogenetics. Mol Cancer Ther 2009; 8(1): 10-6.
[http://dx.doi.org/10.1158/1535-7163.MCT-08-0840] [PMID: 19139108]
[67]
Warwick RA, Hanani M. The contribution of satellite glial cells to chemotherapy-induced neuropathic pain. Eur J Pain 2013; 17(4): 571-80.
[http://dx.doi.org/10.1002/j.1532-2149.2012.00219.x] [PMID: 23065831]
[68]
Milligan ED, Watkins LR. Pathological and protective roles of glia in chronic pain. Nat Rev Neurosci 2009; 10(1): 23-36.
[http://dx.doi.org/10.1038/nrn2533] [PMID: 19096368]
[69]
Zhang H, Boyette-Davis JA, Kosturakis AK, et al. Induction of monocyte chemoattractant protein-1 (MCP-1) and its receptor CCR2 in primary sensory neurons contributes to paclitaxel-induced peripheral neuropathy. J Pain 2013; 14(10): 1031-44.
[http://dx.doi.org/10.1016/j.jpain.2013.03.012] [PMID: 23726937]
[70]
Sun JH, Yang B, Donnelly DF, Ma C, LaMotte RH. MCP-1 enhances excitability of nociceptive neurons in chronically compressed dorsal root ganglia. J Neurophysiol 2006; 96(5): 2189-99.
[http://dx.doi.org/10.1152/jn.00222.2006] [PMID: 16775210]
[71]
White FA, Sun J, Waters SM, 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 2005; 102(39): 14092-7.
[http://dx.doi.org/10.1073/pnas.0503496102] [PMID: 16174730]
[72]
Jung H, Bhangoo S, Banisadr G, et al. Visualization of chemokine receptor activation in transgenic mice reveals peripheral activation of CCR2 receptors in states of neuropathic pain. J Neurosci 2009; 29(25): 8051-62.
[http://dx.doi.org/10.1523/JNEUROSCI.0485-09.2009] [PMID: 19553445]
[73]
Devor M, Wall PD. Cross-excitation in dorsal root ganglia of nerve-injured and intact rats. J Neurophysiol 1990; 64(6): 1733-46.
[http://dx.doi.org/10.1152/jn.1990.64.6.1733] [PMID: 2074461]
[74]
Oh EJ, Weinreich D. Chemical communication between vagal afferent somata in nodose Ganglia of the rat and the Guinea pig in vitro. J Neurophysiol 2002; 87(6): 2801-7.
[http://dx.doi.org/10.1152/jn.2002.87.6.2801] [PMID: 12037182]
[75]
Boyette-Davis JA, Cata JP, Zhang H, et al. Follow-up psychophysical studies in bortezomib-related chemoneuropathy patients. J Pain 2011; 12(9): 1017-24.
[http://dx.doi.org/10.1016/j.jpain.2011.04.008] [PMID: 21703938]
[76]
Richardson PG, Xie W, Mitsiades C, et al. Single-agent bortezomib in previously untreated multiple myeloma: efficacy, characterization of peripheral neuropathy, and molecular correlations with response and neuropathy. J Clin Oncol 2009; 27(21): 3518-25.
[http://dx.doi.org/10.1200/JCO.2008.18.3087] [PMID: 19528374]
[77]
Cook DN. The role of MIP-1 alpha in inflammation and hematopoiesis. J Leukoc Biol 1996; 59(1): 61-6.
[http://dx.doi.org/10.1002/jlb.59.1.61] [PMID: 8558069]
[78]
Menten P, Wuyts A, Van Damme J. Macrophage inflammatory protein-1. Cytokine Growth Factor Rev 2002; 13(6): 455-81.
[http://dx.doi.org/10.1016/S1359-6101(02)00045-X] [PMID: 12401480]
[79]
Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res 2009; 29(6): 313-26.
[http://dx.doi.org/10.1089/jir.2008.0027] [PMID: 19441883]
[80]
Austin PJ, Moalem-Taylor G. The neuro-immune balance in neuropathic pain: involvement of inflammatory immune cells, immune-like glial cells and cytokines. J Neuroimmunol 2010; 229(1-2): 26-50.
[http://dx.doi.org/10.1016/j.jneuroim.2010.08.013] [PMID: 20870295]
[81]
Elson K, Speck P, Simmons A. Herpes simplex virus infection of murine sensory ganglia induces proliferation of neuronal satellite cells. J Gen Virol 2003; 84(Pt 5): 1079-84.
[http://dx.doi.org/10.1099/vir.0.19035-0] [PMID: 12692271]
[82]
Takeda M, Tanimoto T, Kadoi J, et al. Enhanced excitability of nociceptive trigeminal ganglion neurons by satellite glial cytokine following peripheral inflammation. Pain 2007; 129(1-2): 155-66.
[http://dx.doi.org/10.1016/j.pain.2006.10.007] [PMID: 17127002]
[83]
Li Y, Zhang H, Kosturakis AK, et al. MAPK signaling downstream to TLR4 contributes to paclitaxel-induced peripheral neuropathy. Brain Behav Immun 2015; 49: 255-66.
[http://dx.doi.org/10.1016/j.bbi.2015.06.003] [PMID: 26065826]
[84]
Li Y, Zhang H, Zhang H, Kosturakis AK, Jawad AB, Dougherty PM. Toll-like receptor 4 signaling contributes to Paclitaxel-induced peripheral neuropathy. J Pain 2014; 15(7): 712-25.
[http://dx.doi.org/10.1016/j.jpain.2014.04.001] [PMID: 24755282]
[85]
Makker PG, Duffy SS, Lees JG, et al. Characterisation of Immune and Neuroinflammatory Changes Associated with Chemotherapy-Induced Peripheral Neuropathy. PLoS One 2017; 12(1)e0170814
[http://dx.doi.org/10.1371/journal.pone.0170814] [PMID: 28125674]
[86]
Al-Mazidi S, Alotaibi M, Nedjadi T, Chaudhary A, Alzoghaibi M, Djouhri L. Blocking of cytokines signalling attenuates evoked and spontaneous neuropathic pain behaviours in the paclitaxel rat model of chemotherapy-induced neuropathy. Eur J Pain 2018; 22(4): 810-21.
[http://dx.doi.org/10.1002/ejp.1169] [PMID: 29282807]
[87]
Zhang H, Li Y, de Carvalho-Barbosa M, et al. Dorsal Root Ganglion Infiltration by Macrophages Contributes to Paclitaxel Chemotherapy-Induced Peripheral Neuropathy. J Pain 2016; 17(7): 775-86.
[http://dx.doi.org/10.1016/j.jpain.2016.02.011] [PMID: 26979998]
[88]
Curry ZA, Wilkerson JL, Bagdas D, et al. Monoacylglycerol lipase inhibitors reverse paclitaxel-induced nociceptive behavior and Proinflammatory markers in a mouse model of chemotherapy-induced neuropathy. J Pharmacol Exp Ther 2018; 366(1): 169-83.
[http://dx.doi.org/10.1124/jpet.117.245704] [PMID: 29540562]
[89]
Illias AM, Gist AC, Zhang H, Kosturakis AK, Dougherty PM. Chemokine CCL2 and its receptor CCR2 in the dorsal root ganglion contribute to oxaliplatin-induced mechanical hypersensitivity. Pain 2018; 159(7): 1308-16.
[http://dx.doi.org/10.1097/j.pain.0000000000001212] [PMID: 29554018]
[90]
Ju G, Hökfelt T, Brodin E, et al. Primary sensory neurons of the rat showing calcitonin gene-related peptide immunoreactivity and their relation to substance P-, somatostatin-, galanin-, vasoactive intestinal polypeptide- and cholecystokinin-immunoreactive ganglion cells. Cell Tissue Res 1987; 247(2): 417-31.
[http://dx.doi.org/10.1007/BF00218323] [PMID: 2434236]
[91]
Li L, Rutlin M, Abraira VE, et al. The functional organization of cutaneous low-threshold mechanosensory neurons. Cell 2011; 147(7): 1615-27.
[http://dx.doi.org/10.1016/j.cell.2011.11.027] [PMID: 22196735]
[92]
Wang H, Rivero-Melián C, Robertson B, Grant G. Transganglionic transport and binding of the isolectin B4 from Griffonia simplicifolia I in rat primary sensory neurons. Neuroscience 1994; 62(2): 539-51.
[http://dx.doi.org/10.1016/0306-4522(94)90387-5] [PMID: 7530347]
[93]
Montague K, Simeoli R, Valente J, Malcangio M. A novel interaction between CX3CR1 and CCR2 signalling in monocytes constitutes an underlying mechanism for persistent vincristine-induced pain. J Neuroinflammation 2018; 15(1): 101.
[http://dx.doi.org/10.1186/s12974-018-1116-6] [PMID: 29625610]
[94]
Curran MP, McKeage K. Bortezomib: a review of its use in patients with multiple myeloma. Drugs 2009; 69(7): 859-88.
[http://dx.doi.org/10.2165/00003495-200969070-00006] [PMID: 19441872]
[95]
Kumar SK, Rajkumar SV, Dispenzieri A, et al. Improved survival in multiple myeloma and the impact of novel therapies. Blood 2008; 111(5): 2516-20.
[http://dx.doi.org/10.1182/blood-2007-10-116129] [PMID: 17975015]
[96]
Liu C, Luan S, OuYang H, et al. Upregulation of CCL2 via ATF3/c-Jun interaction mediated the Bortezomib-induced peripheral neuropathy. Brain Behav Immun 2016; 53: 96-104.
[http://dx.doi.org/10.1016/j.bbi.2015.11.004] [PMID: 26554515]
[97]
Zhang H, Yoon SY, Zhang H, Dougherty PM. Evidence that spinal astrocytes but not microglia contribute to the pathogenesis of Paclitaxel-induced painful neuropathy. J Pain 2012; 13(3): 293-303.
[http://dx.doi.org/10.1016/j.jpain.2011.12.002] [PMID: 22285612]
[98]
Wang YS, Li YY, Cui W, et al. Melatonin Attenuates Pain Hypersensitivity and Decreases Astrocyte-Mediated Spinal Neuroinflammation in a Rat Model of Oxaliplatin-Induced Pain. Inflammation 2017; 40(6): 2052-61.
[http://dx.doi.org/10.1007/s10753-017-0645-y] [PMID: 28812173]
[99]
Pan Y, Lloyd C, Zhou H, et al. Neurotactin, a membrane-anchored chemokine upregulated in brain inflammation. Nature 1997; 387(6633): 611-7.
[http://dx.doi.org/10.1038/42491] [PMID: 9177350]
[100]
Verge GM, Milligan ED, Maier SF, Watkins LR, Naeve GS, Foster AC. Fractalkine (CX3CL1) and fractalkine receptor (CX3CR1) distribution in spinal cord and dorsal root ganglia under basal and neuropathic pain conditions. Eur J Neurosci 2004; 20(5): 1150-60.
[http://dx.doi.org/10.1111/j.1460-9568.2004.03593.x] [PMID: 15341587]
[101]
Chapman GA, Moores K, Harrison D, Campbell CA, Stewart BR, Strijbos PJ. Fractalkine cleavage from neuronal membranes represents an acute event in the inflammatory response to excitotoxic brain damage. J Neurosci 2000; 20(15): RC87.
[http://dx.doi.org/10.1523/JNEUROSCI.20-15-j0004.2000] [PMID: 10899174]
[102]
Kim KW, Vallon-Eberhard A, Zigmond E, et al. In vivo structure/function and expression analysis of the CX3C chemokine fractalkine. Blood 2011; 118(22): e156-67.
[http://dx.doi.org/10.1182/blood-2011-04-348946] [PMID: 21951685]
[103]
Lindia JA, McGowan E, Jochnowitz N, Abbadie C. Induction of CX3CL1 expression in astrocytes and CX3CR1 in microglia in the spinal cord of a rat model of neuropathic pain. J Pain 2005; 6(7): 434-8.
[http://dx.doi.org/10.1016/j.jpain.2005.02.001] [PMID: 15993821]
[104]
Bazan JF, Bacon KB, Hardiman G, et al. A new class of membrane-bound chemokine with a CX3C motif. Nature 1997; 385(6617): 640-4.
[http://dx.doi.org/10.1038/385640a0] [PMID: 9024663]
[105]
Clark AK, Malcangio M. Microglial signalling mechanisms: Cathepsin S and Fractalkine. Exp Neurol 2012; 234(2): 283-92.
[http://dx.doi.org/10.1016/j.expneurol.2011.09.012] [PMID: 21946268]
[106]
Clark AK, Staniland AA, Malcangio M. Fractalkine/CX3CR1 signalling in chronic pain and inflammation. Curr Pharm Biotechnol 2011; 12(10): 1707-14.
[http://dx.doi.org/10.2174/138920111798357465] [PMID: 21466443]
[107]
Huang ZZ, Li D, Ou-Yang HD, et al. Cerebrospinal fluid oxaliplatin contributes to the acute pain induced by systemic administration of oxaliplatin. Anesthesiology 2016; 124(5): 1109-21.
[http://dx.doi.org/10.1097/ALN.0000000000001084] [PMID: 26978408]
[108]
Li D, Huang ZZ, Ling YZ, et al. Up-regulation of cx3cl1 via nuclear factor-κb-dependent histone acetylation is involved in paclitaxel-induced peripheral neuropathy. Anesthesiology 2015; 122(5): 1142-51.
[http://dx.doi.org/10.1097/ALN.0000000000000560] [PMID: 25494456]
[109]
Huang ZZ, Li D, Liu CC, et al. CX3CL1-mediated macrophage activation contributed to paclitaxel-induced DRG neuronal apoptosis and painful peripheral neuropathy. Brain Behav Immun 2014; 40: 155-65.
[http://dx.doi.org/10.1016/j.bbi.2014.03.014] [PMID: 24681252]
[110]
Montague K, Malcangio M. The therapeutic potential of monocyte/macrophage manipulation in the treatment of chemotherapy-induced painful neuropathy. Front Mol Neurosci 2017; 10: 397.
[http://dx.doi.org/10.3389/fnmol.2017.00397] [PMID: 29230166]
[111]
Carlin LM, Stamatiades EG, Auffray C, et al. Nr4a1-dependent Ly6C(low) monocytes monitor endothelial cells and orchestrate their disposal. Cell 2013; 153(2): 362-75.
[http://dx.doi.org/10.1016/j.cell.2013.03.010] [PMID: 23582326]
[112]
Schwarz N, Pruessmeyer J, Hess FM, et al. Requirements for leukocyte transmigration via the transmembrane chemokine CX3CL1. Cell Mol Life Sci 2010; 67(24): 4233-48.
[http://dx.doi.org/10.1007/s00018-010-0433-4] [PMID: 20559678]
[113]
Old EA, Nadkarni S, Grist J, et al. Monocytes expressing CX3CR1 orchestrate the development of vincristine-induced pain. J Clin Invest 2014; 124(5): 2023-36.
[http://dx.doi.org/10.1172/JCI71389] [PMID: 24743146]
[114]
Zhuang ZY, Kawasaki Y, Tan PH, Wen YR, Huang J, Ji RR. Role of the CX3CR1/p38 MAPK pathway in spinal microglia for the development of neuropathic pain following nerve injury-induced cleavage of fractalkine. Brain Behav Immun 2007; 21(5): 642-51.
[http://dx.doi.org/10.1016/j.bbi.2006.11.003] [PMID: 17174525]
[115]
Fonović UP, Jevnikar Z, Kos J. Cathepsin S generates soluble CX3CL1 (fractalkine) in vascular smooth muscle cells. Biol Chem 2013; 394(10): 1349-52.
[http://dx.doi.org/10.1515/hsz-2013-0189] [PMID: 23893684]
[116]
Clark AK, Yip PK, Grist J, et al. Inhibition of spinal microglial cathepsin S for the reversal of neuropathic pain. Proc Natl Acad Sci USA 2007; 104(25): 10655-60.
[http://dx.doi.org/10.1073/pnas.0610811104] [PMID: 17551020]
[117]
Wang J, Zhang XS, Tao R, et al. Upregulation of CX3CL1 mediated by NF-κB activation in dorsal root ganglion contributes to peripheral sensitization and chronic pain induced by oxaliplatin administration. Mol Pain 2017.131744806917726256
[http://dx.doi.org/10.1177/1744806917726256] [PMID: 28849713]
[118]
Salem ML, Al-Khami AA, El-Naggar SA, Díaz-Montero CM, Chen Y, Cole DJ. Cyclophosphamide induces dynamic alterations in the host microenvironments resulting in a Flt3 ligand-dependent expansion of dendritic cells. J Immunol 2010; 184(4): 1737-47.
[http://dx.doi.org/10.4049/jimmunol.0902309] [PMID: 20083664]
[119]
Nakahara T, Uchi H, Lesokhin AM, et al. Cyclophosphamide enhances immunity by modulating the balance of dendritic cell subsets in lymphoid organs. Blood 2010; 115(22): 4384-92.
[http://dx.doi.org/10.1182/blood-2009-11-251231] [PMID: 20154220]
[120]
Medina-Echeverz J, Fioravanti J, Zabala M, Ardaiz N, Prieto J, Berraondo P. Successful colon cancer eradication after chemoimmunotherapy is associated with profound phenotypic change of intratumoral myeloid cells. J Immunol 2011; 186(2): 807-15.
[http://dx.doi.org/10.4049/jimmunol.1001483] [PMID: 21148040]
[121]
Jacquelin S, Licata F, Dorgham K, et al. CX3CR1 reduces Ly6Chigh-monocyte motility within and release from the bone marrow after chemotherapy in mice. Blood 2013; 122(5): 674-83.
[http://dx.doi.org/10.1182/blood-2013-01-480749] [PMID: 23775714]
[122]
Schall T. Fractalkine--a strange attractor in the chemokine landscape. Immunol Today 1997; 18(4): 147.
[http://dx.doi.org/10.1016/S0167-5699(97)84655-5] [PMID: 9136448]
[123]
Hesselgesser J, Horuk R. Chemokine and chemokine receptor expression in the central nervous system. J Neurovirol 1999; 5(1): 13-26.
[http://dx.doi.org/10.3109/13550289909029741] [PMID: 10190686]
[124]
Nishiyori A, Minami M, Ohtani Y, et al. Localization of fractalkine and CX3CR1 mRNAs in rat brain: does fractalkine play a role in signaling from neuron to microglia? FEBS Lett 1998; 429(2): 167-72.
[http://dx.doi.org/10.1016/S0014-5793(98)00583-3] [PMID: 9650583]
[125]
Hughes PM, Botham MS, Frentzel S, Mir A, Perry VH. Expression of fractalkine (CX3CL1) and its receptor, CX3CR1, during acute and chronic inflammation in the rodent CNS. Glia 2002; 37(4): 314-27.
[http://dx.doi.org/10.1002/glia.10037] [PMID: 11870871]
[126]
Harrison JK, Jiang Y, Chen S, et al. Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia. Proc Natl Acad Sci USA 1998; 95(18): 10896-901.
[http://dx.doi.org/10.1073/pnas.95.18.10896] [PMID: 9724801]
[127]
Milligan ED, Zapata V, Chacur M, et al. Evidence that exogenous and endogenous fractalkine can induce spinal nociceptive facilitation in rats. Eur J Neurosci 2004; 20(9): 2294-302.
[http://dx.doi.org/10.1111/j.1460-9568.2004.03709.x] [PMID: 15525271]
[128]
Wolf Y, Yona S, Kim KW, Jung S. Microglia, seen from the CX3CR1 angle. Front Cell Neurosci 2013; 7: 26.
[http://dx.doi.org/10.3389/fncel.2013.00026] [PMID: 23507975]
[129]
Lo HM, Lai TH, Li CH, Wu WB. TNF-α induces CXCL1 chemokine expression and release in human vascular endothelial cells in vitro via two distinct signaling pathways. Acta Pharmacol Sin 2014; 35(3): 339-50.
[http://dx.doi.org/10.1038/aps.2013.182] [PMID: 24487964]
[130]
Rittner HL, Mousa SA, Labuz D, et al. Selective local PMN recruitment by CXCL1 or CXCL2/3 injection does not cause inflammatory pain. J Leukoc Biol 2006; 79(5): 1022-32.
[http://dx.doi.org/10.1189/jlb.0805452] [PMID: 16522746]
[131]
Acharyya S, Oskarsson T, Vanharanta S, et al. A CXCL1 paracrine network links cancer chemoresistance and metastasis. Cell 2012; 150(1): 165-78.
[http://dx.doi.org/10.1016/j.cell.2012.04.042] [PMID: 22770218]
[132]
Zou A, Lambert D, Yeh H, et al. Elevated CXCL1 expression in breast cancer stroma predicts poor prognosis and is inversely associated with expression of TGF-β signaling proteins. BMC Cancer 2014; 14: 781.
[http://dx.doi.org/10.1186/1471-2407-14-781] [PMID: 25344051]
[133]
Dong F, Du YR, Xie W, Strong JA, He XJ, Zhang JM. Increased function of the TRPV1 channel in small sensory neurons after local inflammation or in vitro exposure to the pro-inflammatory cytokine GRO/KC. Neurosci Bull 2012; 28(2): 155-64.
[http://dx.doi.org/10.1007/s12264-012-1208-8] [PMID: 22466126]
[134]
Vallès A, Grijpink-Ongering L, de Bree FM, Tuinstra T, Ronken E. Differential regulation of the CXCR2 chemokine network in rat brain trauma: implications for neuroimmune interactions and neuronal survival. Neurobiol Dis 2006; 22(2): 312-22.
[http://dx.doi.org/10.1016/j.nbd.2005.11.015] [PMID: 16472549]
[135]
Ragozzino D. CXC chemokine receptors in the central nervous system: Role in cerebellar neuromodulation and development. J Neurovirol 2002; 8(6): 559-72.
[http://dx.doi.org/10.1080/13550280290100932] [PMID: 12476350]
[136]
Yang RH, Strong JA, Zhang JM. NF-kappaB mediated enhancement of potassium currents by the chemokine CXCL1/growth related oncogene in small diameter rat sensory neurons. Mol Pain 2009; 5: 26.
[http://dx.doi.org/10.1186/1744-8069-5-26] [PMID: 19476648]
[137]
Zhang ZJ, Cao DL, Zhang X, Ji RR, Gao YJ. Chemokine contribution to neuropathic pain: respective induction of CXCL1 and CXCR2 in spinal cord astrocytes and neurons. Pain 2013; 154(10): 2185-97.
[http://dx.doi.org/10.1016/j.pain.2013.07.002] [PMID: 23831863]
[138]
Manjavachi MN, Costa R, Quintão NL, Calixto JB. The role of keratinocyte-derived chemokine (KC) on hyperalgesia caused by peripheral nerve injury in mice. Neuropharmacology 2014; 79: 17-27.
[http://dx.doi.org/10.1016/j.neuropharm.2013.10.026] [PMID: 24184386]
[139]
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(Pt 8): 2193-209.
[http://dx.doi.org/10.1093/brain/awu140] [PMID: 24919967]
[140]
Orchard TS, Gaudier-Diaz MM, Phuwamongkolwiwat-Chu P, et al. Low sucrose, omega-3 enriched diet has region-specific effects on neuroinflammation and synaptic function markers in a mouse model of doxorubicin-based chemotherapy. Nutrients 2018; 10(12)E2004
[http://dx.doi.org/10.3390/nu10122004] [PMID: 30567351]
[141]
Hsu YL, Hung JY, Tsai EM, et al. Benzyl butyl phthalate increases the chemoresistance to doxorubicin/cyclophosphamide by increasing breast cancer-associated dendritic cell-derived CXCL1/GROα and S100A8/A9. Oncol Rep 2015; 34(6): 2889-900.
[http://dx.doi.org/10.3892/or.2015.4307] [PMID: 26397389]
[142]
Chen L, Pan XW, Huang H, et al. Epithelial-mesenchymal transition induced by GRO-α-CXCR2 promotes bladder cancer recurrence after intravesical chemotherapy. Oncotarget 2017; 8(28): 45274-85.
[http://dx.doi.org/10.18632/oncotarget.16786] [PMID: 28423359]
[143]
Wong J, Tran LT, Magun EA, Magun BE, Wood LJ. Production of IL-1β by bone marrow-derived macrophages in response to chemotherapeutic drugs: synergistic effects of doxorubicin and vincristine. Cancer Biol Ther 2014; 15(10): 1395-403.
[http://dx.doi.org/10.4161/cbt.29922] [PMID: 25046000]
[144]
Zhou L, Hu Y, Li C, et al. Levo-corydalmine alleviates vincristine-induced neuropathic pain in mice by inhibiting an NF-kappa B-dependent CXCL1/CXCR2 signaling pathway. Neuropharmacology 2018; 135: 34-47.
[http://dx.doi.org/10.1016/j.neuropharm.2018.03.004] [PMID: 29518397]
[145]
Luo X, Wang X, Xia Z, Chung SK, Cheung CW. CXCL12/CXCR4 axis: an emerging neuromodulator in pathological pain. Rev Neurosci 2016; 27(1): 83-92.
[http://dx.doi.org/10.1515/revneuro-2015-0016] [PMID: 26353174]
[146]
Li M, Ransohoff RM. Multiple roles of chemokine CXCL12 in the central nervous system: a migration from immunology to neurobiology. Prog Neurobiol 2008; 84(2): 116-31.
[http://dx.doi.org/10.1016/j.pneurobio.2007.11.003] [PMID: 18177992]
[147]
Guyon A. CXCL12 chemokine and its receptors as major players in the interactions between immune and nervous systems. Front Cell Neurosci 2014; 8: 65.
[http://dx.doi.org/10.3389/fncel.2014.00065] [PMID: 24639628]
[148]
White FA, Wilson NM. Chemokines as pain mediators and modulators. Curr Opin Anaesthesiol 2008; 21(5): 580-5.
[http://dx.doi.org/10.1097/ACO.0b013e32830eb69d] [PMID: 18784482]
[149]
Memi F, Abe P, Cariboni A, MacKay F, Parnavelas JG, Stumm R. CXC chemokine receptor 7 (CXCR7) affects the migration of GnRH neurons by regulating CXCL12 availability. J Neurosci 2013; 33(44): 17527-37.
[http://dx.doi.org/10.1523/JNEUROSCI.0857-13.2013] [PMID: 24174685]
[150]
Dubový P, Klusáková I, Svízenská I, Brázda V. Spatio-temporal changes of SDF1 and its CXCR4 receptor in the dorsal root ganglia following unilateral sciatic nerve injury as a model of neuropathic pain. Histochem Cell Biol 2010; 133(3): 323-37.
[http://dx.doi.org/10.1007/s00418-010-0675-0] [PMID: 20127490]
[151]
Bai L, Wang X, Li Z, et al. Upregulation of chemokine cxcl12 in the dorsal root ganglia and spinal cord contributes to the development and maintenance of neuropathic pain following spared nerve injury in rats. Neurosci Bull 2016; 32(1): 27-40.
[http://dx.doi.org/10.1007/s12264-015-0007-4] [PMID: 26781879]
[152]
Reaux-Le Goazigo A, Rivat C, Kitabgi P, Pohl M, Melik Parsadaniantz S. Cellular and subcellular localization of CXCL12 and CXCR4 in rat nociceptive structures: physiological relevance. Eur J Neurosci 2012; 36(5): 2619-31.
[http://dx.doi.org/10.1111/j.1460-9568.2012.08179.x] [PMID: 22694179]
[153]
Xu T, Zhang XL, Ou-Yang HD, et al. Epigenetic upregulation of CXCL12 expression mediates antitubulin chemotherapeutics-induced neuropathic pain. Pain 2017; 158(4): 637-48.
[http://dx.doi.org/10.1097/j.pain.0000000000000805] [PMID: 28072604]
[154]
Li YY, Li H, Liu ZL, et al. Activation of STAT3-mediated CXCL12 up-regulation in the dorsal root ganglion contributes to oxaliplatin-induced chronic pain. Mol Pain 2017.131744806917747425
[http://dx.doi.org/10.1177/1744806917747425] [PMID: 29166835]
[155]
Matsushita K, Tozaki-Saitoh H, Kojima C, et al. Chemokine (C-C motif) receptor 5 is an important pathological regulator in the development and maintenance of neuropathic pain. Anesthesiology 2014; 120(6): 1491-503.
[http://dx.doi.org/10.1097/ALN.0000000000000190] [PMID: 24589480]
[156]
Gamo K, Kiryu-Seo S, Konishi H, et al. G-protein-coupled receptor screen reveals a role for chemokine receptor CCR5 in suppressing microglial neurotoxicity. J Neurosci 2008; 28(46): 11980-8.
[http://dx.doi.org/10.1523/JNEUROSCI.2920-08.2008] [PMID: 19005063]
[157]
Jo YH, Schlichter R. Synaptic corelease of ATP and GABA in cultured spinal neurons. Nat Neurosci 1999; 2(3): 241-5.
[http://dx.doi.org/10.1038/6344] [PMID: 10195216]
[158]
Fam SR, Gallagher CJ, Salter MW. P2Y(1) purinoceptor-mediated Ca(2+) signaling and Ca(2+) wave propagation in dorsal spinal cord astrocytes. J Neurosci 2000; 20(8): 2800-8.
[http://dx.doi.org/10.1523/JNEUROSCI.20-08-02800.2000] [PMID: 10751431]
[159]
Ochi-ishi R, Nagata K, Inoue T, Tozaki-Saitoh H, Tsuda M, Inoue K. Involvement of the chemokine CCL3 and the purinoceptor P2X7 in the spinal cord in paclitaxel-induced mechanical allodynia. Mol Pain 2014; 10: 53.
[http://dx.doi.org/10.1186/1744-8069-10-53] [PMID: 25127716]
[160]
Cataldo G, Erb SJ, Lunzer MM, et al. The bivalent ligand MCC22 potently attenuates hyperalgesia in a mouse model of cisplatin-evoked neuropathic pain without tolerance or reward. Neuropharmacology 2019. 107598.
[http://dx.doi.org/10.1016/j.neuropharm.2019.04.004] [PMID: 30970233]
[161]
Carr FB, Géranton SM, Hunt SP. Descending controls modulate inflammatory joint pain and regulate CXC chemokine and iNOS expression in the dorsal horn. Mol Pain 2014; 10: 39.
[http://dx.doi.org/10.1186/1744-8069-10-39] [PMID: 24947159]
[162]
Liu W, Ye J, Yan H. Investigation of key genes and pathways in inhibition of oxycodone on vincristine-induced microglia activation by using bioinformatics analysis. Dis Markers 2019.20193521746
[http://dx.doi.org/10.1155/2019/3521746] [PMID: 30881521]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy