Delayed functional expression of neuronal chemokine receptors following focal nerve demyelination in the rat: a mechanism for the development of chronic sensitization of peripheral nociceptors

Sonia Bhangoo, Dongjun Ren, Richard J Miller, Kenneth J Henry, Jayana Lineswala, Chafiq Hamdouchi, Baolin Li, Patrick E Monahan, David M Chan, Matthew S Ripsch, Fletcher A White, Sonia Bhangoo, Dongjun Ren, Richard J Miller, Kenneth J Henry, Jayana Lineswala, Chafiq Hamdouchi, Baolin Li, Patrick E Monahan, David M Chan, Matthew S Ripsch, Fletcher A White

Abstract

Background: Animal and clinical studies have revealed that focal peripheral nerve axon demyelination is accompanied by nociceptive pain behavior. C-C and C-X-C chemokines and their receptors have been strongly implicated in demyelinating polyneuropathies and persistent pain syndromes. Herein, we studied the degree to which chronic nociceptive pain behavior is correlated with the neuronal expression of chemokines and their receptors following unilateral lysophosphatidylcholine (LPC)-induced focal demyelination of the sciatic nerve in rats.

Results: Focal nerve demyelination increased behavioral reflex responsiveness to mechanical stimuli between postoperative day (POD) 3 and POD28 in both the hindpaw ipsilateral and contralateral to the nerve injury. This behavior was accompanied by a bilateral increase in the numbers of primary sensory neurons expressing the chemokine receptors CCR2, CCR5, and CXCR4 by POD14, with no change in the pattern of CXCR3 expression. Significant increases in the numbers of neurons expressing the chemokines monocyte chemoattractant protein-1 (MCP-1/CCL2), Regulated on Activation, Normal T Expressed and Secreted (RANTES/CCL5) and interferon gamma-inducing protein-10 (IP-10/CXCL10) were also evident following nerve injury, although neuronal expression pattern of stromal cell derived factor-1alpha (SDF1/CXCL12) did not change. Functional studies demonstrated that acutely dissociated sensory neurons derived from LPC-injured animals responded with increased [Ca2+]i following exposure to MCP-1, IP-10, SDF1 and RANTES on POD 14 and 28, but these responses were largely absent by POD35. On days 14 and 28, rats received either saline or a CCR2 receptor antagonist isomer (CCR2 RA-[R]) or its inactive enantiomer (CCR2 RA-[S]) by intraperitoneal (i.p.) injection. CCR2 RA-[R] treatment of nerve-injured rats produced stereospecific bilateral reversal of tactile hyperalgesia.

Conclusion: These results suggest that the presence of chemokine signaling by both injured and adjacent, uninjured sensory neurons is correlated with the maintenance phase of a persistent pain state, suggesting that chemokine receptor antagonists may be an important therapeutic intervention for chronic pain.

Figures

Figure 1
Figure 1
Mean threshold force required for paw withdrawal to Von Frey stimulation at 1, 3, 7, 14, 21, 28, 35 and 42 days following LPC-induced focal nerve demyelination. Each data point is the mean threshold (± SE) force on the hindpaw ipsilateral (black circle) or contralateral (white circle) to the focal nerve injury site eliciting a withdrawal response (n = 10). Reduced behavioral thresholds for the hindpaw ipsilateral to the nerve lesion were significantly different from pre-operative baseline on postoperative days 1–28. The threshold force for the hindpaw contralateral to the nerve lesion did not reach significance until postoperative day 3, and significant differences were observed until postoperative day 28. The time course of sham injury (n = 6) is also represented but did not differ from the uninjured animals. Analysis was performed using two-way ANOVA followed by the Bonferroni post-hoc pair-wise comparisons (*p B) LPC-induced focal nerve demyelination did not produce changes in thermal responses as assessed by the Hargreaves test. Each bar is the mean withdrawal latency (± SE) of the hindpaw ipsilateral (white bar) or contralateral (black bar) to the focal nerve demyelination injury at postoperative day 7 and 14 (n = 10).
Figure 2
Figure 2
Expression of CCR2 mRNA and protein immunoreactivity in rat lumbar DRG ipsilateral to focal nerve demyelination. A) Lumbar DRG removed from vehicle-treated animals at POD7 did not exhibit CCR2 mRNA expression (n = 5). B) Many lumbar DRG neurons in vehicle-treated rats sensory neurons were positive for isolectin IB4, a neuronal phenotype that distinguishes some C-fiber nociceptors (green cells). There was no evidence of CCR2 protein expression in sham animals (n = 5). C) Lumbar DRG neurons from nerve-injured rats on POD7 exhibited CCR2 mRNA transcripts in some small and medium diameter neurons (black arrows). Open black arrowhead indicates a neuron without CCR2 mRNA transcripts (n = 4). D) Lumbar DRG neurons from a rat subjected to focal nerve demyelination exhibited few CCR2 immunopositive (white arrows) sensory neurons (n = 4). E) Many lumbar DRG neurons on POD14 exhibited CCR2 mRNA transcripts (black arrows). Open arrowhead indicates non-labeled neuron. F) CCR2 immunoreactivity was present in an increased number of neurons at POD14 (white arrows; n = 5). Scale bar is; 30 μm (A, C), 50 μm (B, D, F), and 100 μm (E).
Figure 3
Figure 3
Percentage of MCP-1, CCR2 and IP-10 immunoreactive neurons with IB4-positive neuronal profiles on POD 14. MCP-1 expression was increased by LPC-induced nerve injury within the IB4-labeled neuronal group in both the DRG ipsilateral and contralateral to the nerve injury. Sham-injury treatment did not produce significant changes in the extent of MCP-1/IB4 colocalization. CCR2 expression was increased by LPC-induced nerve injury within IB4-labeled neuronal group in both the DRG ipsilateral and contralateral to the nerve injury. Like, MCP-1, sham-injury treatment did not produce significant changes in CCR2/IB4 colocalization in either DRG ipsi- or contralateral to the sham injury. IP-10 expression was increased by LPC-induced nerve injury within IB4-labelled neuronal group in both the DRG ipsilateral and contralateral to the nerve injury, while sham-injury treatment did not produce significant changes in IP-10/IB4 colocalization. Comparisons of immunoreactive cell percentages were made between LPC-treatment and sham-treated animals. Data represent means ± SE. Analysis was performed using two-way ANOVA followed by the Bonferroni post-hoc pair-wise comparisons (*p < 0.01).
Figure 4
Figure 4
Expression of CXCR4 mRNA in rat lumbar DRG ipsilateral to focal nerve demyelination. Low (A) and high power (C) photomicrographs of CXCR4 mRNA transcripts present in lumbar DRG removed from vehicle-treated rodents at POD14 (n = 3). Many non-neuronal cells strongly expressed CXCR4. (C) Some presumptive neurons expressed low levels of CXCR4 mRNA (white arrow). Low (B) and high power (D) photomicrographs of CXCR4 mRNA transcripts present in lumbar DRGs derived from injured rats at POD14 (n = 4). The expression level and number of non-neuronal cells exhibiting CXCR4 mRNA transcripts in the lumbar DRG did not change following focal nerve demyelination. However, many neurons upregulated CXCR4 mRNA expression (D; white arrows indicate neurons with low levels of mRNA transcripts; white arrowhead points to a neuron lacking CXCR4 mRNA expression). (Scale bar is 1 mm (A and B); 40 μm (C and D).
Figure 5
Figure 5
Expression of CXCR3 and CCR5 mRNA in rat lumbar DRG following focal nerve demyelination. (A) Many sensory neurons in the lumbar DRG removed from vehicle-treated rats exhibited CXCR3 mRNA transcripts at POD14 (n = 3). (B) CCR5 mRNA expression was absent from the lumbar DRG of vehicle-treated rats at POD14 (n = 3). (C) CXCR3 mRNA expression patterns in sensory neurons subjected to focal nerve demyelination did not differ from vehicle-treated rodents at POD14 (n = 4), but there was an increase in the intensity of CXCR3 mRNA expression. (D) Many neurons in the injured rat lumbar DRG expressed CCR5 transcripts at POD14 (n = 4). Scale bar is 250 μm (A and C); 100 μm (B and D).
Figure 6
Figure 6
Colocalization of MCP-1 immunoreactivity (ir) and isolectin B4(IB4)-binding in the lumbar DRG ipsilateral to LPC-induced demyelination injury at POD7. IB4-binding in rat DRG neurons distinguishes C-fiber nociceptors. (A) Naïve rat lumbar DRG were completely negative for MCP-1ir. (B) Lumbar DRG ipsilateral to LPC-induced sciatic nerve injury exhibited numerous small diameter neurons that are MCP-1 positive at POD7 (red arrows). (C) Numerous small IB4-binding presumptive nociceptors are present in the same DRG tissue section (green arrows). (D) Merging panels B and C demonstrates the extent of colocalization present in lumbar DRG tissue section (yellow arrows). Note not all MCP-1ir neurons were positive for IB4 at POD7 (red arrow). Scale bar is 100 μm (A, B, C and D).
Figure 7
Figure 7
Expression of RANTES mRNA in rat lumbar DRG following focal nerve demyelination. (A) RANTES mRNA expression was absent from the lumbar DRG of vehicle-treated rats at POD14 (n = 3). (B) Many sensory neurons in the DRG ipsilateral to the nerve injury at POD 14 were positive for RANTES mRNA (C) Numerous sensory neurons in the DRG contralateral to the nerve injury displayed expression of RANTES mRNA, albeit at a lower level when compared with the DRG ipsilateral to the nerve injury. Scale bar is 250 μm (A and C); 100 μm (B and D).
Figure 8
Figure 8
Colocalization of IP-10 immunoreactivity (-ir) and isolectin B4 (IB4)-binding neurons in the lumbar DRG of naïve rat and rats subjected to LPC-induced nerve injury. IB4-binding in rat DRG neurons distinguishes a population of C-fiber nociceptors. A) The majority of IP-10-ir cells were limited to medium diameter neurons in the lumbar DRG from vehicle-treated rats (red arrows). D) IB4-binding small diameter presumptive nociceptors (green arrows) did not colocalize with IP-10-ir lumbar DRG neurons from vehicle-treated rodents at POD7 (G, merged images). B) Lumbar DRG ipsilateral to focal nerve demyelination exhibited numerous medium and small diameter IP-10-ir neurons at POD7. Limited numbers of neurons were positive for both IP-10-ir (B, red arrows) and IB4-binding (E, green arrows) on POD7 (H, merged images). C) Many IP-10-ir neurons (red arrows) colocalized with IB4-binding neurons (F, green arrows) at POD14 (I, merged images). Yellow arrows indicate colocalized cells. Scale bar is 100 μm.
Figure 9
Figure 9
Chemokines increased [Ca2+]i levels in acutely isolated rat DRG cells following focal demyelination injury. The figure shows examples of responses of cells acutely isolated from rat DRGs ipsilateral to the nerve injury at various days after a focal demyelination injury. Under normal conditions, cells rarely respond to any chemokine but did respond to other stimuli such as high K or ATP (A). However, there was an increased responsiveness of the cells, the majority of which could be characterized as neurons, between post-operative days 14–28 (B and C, respectively). The frequency of the responses to chemokines returned to approximately the same level as control animals by post-operative day 35 (D). For all experiments, MCP-1 (M), IP-10 (I), RANTES (R), SDF1 (S) were applied at a concentration of 100 nM. Capsaicin (C), high K (K) and ATP (A) were applied at concentrations of 100 nM, 50 mM and 100 uM, respectively.
Figure 10
Figure 10
CCR2 receptor antagonist (CCR2 RA-[R]) administration reversed existing nociceptive behavior. Animals were subjected to a nerve demyelination injury on day 0 and nociceptive behavior was assessed for 28 days. On days 14 and 28 post-surgery, animals received 5 mg/kg CCR2 RA-[R] or 5 mg/kg of its inactive enantiomer, (CCR2 RA-[S], or saline by intraperitoneal injection, and behavioral responses were tested 1 h later. Administration of the CCR2 RA-[R] to focal nerve demyelination injured rats resulted in a significant bilateral increase of mN force required to elicit a paw withdrawal compared with vehicle-treated controls and animals subjected to CCR2 RA-[S]. Nociceptive behavior in vehicle-treated controls and animals subjected to CCR2 RA-[S] differed significantly from day 0 pre-injury baseline responses (*p < 0.01). Data represent means ± SE.

References

    1. Thacker MA, Clark AK, Marchand F, McMahon SB. Pathophysiology of peripheral neuropathic pain: immune cells and molecules. Anesth Analg. 2007;105:838–47. doi: 10.1213/01.ane.0000275190.42912.37.
    1. Verri WA, Jr, Cunha TM, Parada CA, Poole S, Cunha FQ, Ferreira SH. Hypernociceptive role of cytokines and chemokines: targets for analgesic drug development? Pharmacol Ther. 2006;112:116–38. doi: 10.1016/j.pharmthera.2006.04.001.
    1. Bhangoo SK, Ren D, Miller RJ, Chan DM, Ripsch MS, Weiss C, McGinnis C, White FA. CXCR4 chemokine receptor signaling mediates pain hypersensitivity in association with antiretroviral toxic neuropathy. Brain Behav Immun. 2007;21:581–91. doi: 10.1016/j.bbi.2006.12.003.
    1. 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;21:5027–35.
    1. Qin X, Wan Y, Wang X. CCL2 and CXCL1 trigger calcitonin gene-related peptide release by exciting primary nociceptive neurons. J Neurosci Res. 2005;82:51–62. doi: 10.1002/jnr.20612.
    1. 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;96:2189–99. doi: 10.1152/jn.00222.2006.
    1. Tanaka T, Minami M, Nakagawa T, Satoh M. Enhanced production of monocyte chemoattractant protein-1 in the dorsal root ganglia in a rat model of neuropathic pain: possible involvement in the development of neuropathic pain. Neurosci Res. 2004;48:463–9. doi: 10.1016/j.neures.2004.01.004.
    1. White FA, Sun J, Waters SM, Ma C, Ren D, Ripsch M, Steflik J, Cortright DN, Lamotte RH, Miller RJ. 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;102:14092–7. doi: 10.1073/pnas.0503496102.
    1. Jung H, Toth PT, White FA, Miller RJ. Monocyte chemoattractant protein-1 functions as a neuromodulator in dorsal root ganglia neurons. J Neurochem. 2007.
    1. Zhang N, Inan S, Cowan A, Sun R, Wang JM, Rogers TJ, Caterina M, Oppenheim JJ. A proinflammatory chemokine, CCL3, sensitizes the heat- and capsaicin-gated ion channel TRPV1. Proc Natl Acad Sci USA. 2005;102:4536–41. doi: 10.1073/pnas.0406030102.
    1. Abbadie C, Lindia JA, Cumiskey AM, Peterson LB, Mudgett JS, Bayne EK, DeMartino JA, MacIntyre DE, Forrest MJ. Impaired neuropathic pain responses in mice lacking the chemokine receptor CCR2. Proc Natl Acad Sci USA. 2003;100:7947–52. doi: 10.1073/pnas.1331358100.
    1. Wallace VC, Cottrell DF, Brophy PJ, Fleetwood-Walker SM. Focal lysolecithin-induced demyelination of peripheral afferents results in neuropathic pain behavior that is attenuated by cannabinoids. J Neurosci. 2003;23:3221–33.
    1. Cook WJ, Kramer MF, Walker RM, Burwell TJ, Holman HA, Coen DM, Knipe DM. Persistent expression of chemokine and chemokine receptor RNAs at primary and latent sites of herpes simplex virus 1 infection. Virol J. 2004;1:5. doi: 10.1186/1743-422X-1-5.
    1. Melli G, Keswani SC, Fischer A, Chen W, Hoke A. Spatially distinct and functionally independent mechanisms of axonal degeneration in a model of HIV-associated sensory neuropathy. Brain. 2006;129:1330–8. doi: 10.1093/brain/awl058.
    1. Subang MC, Richardson PM. Influence of injury and cytokines on synthesis of monocyte chemoattractant protein-1 mRNA in peripheral nervous tissue. Eur J Neurosci. 2001;13:521–8. doi: 10.1046/j.1460-9568.2001.01425.x.
    1. Taskinen HS, Roytta M. Increased expression of chemokines (MCP-1, MIP-1alpha, RANTES) after peripheral nerve transection. J Peripher Nerv Syst. 2000;5:75–81. doi: 10.1046/j.1529-8027.2000.00009.x.
    1. Bhangoo S, Jung H, Chan DM, Ripsch M, Ren D, Miller RJ, White FA. Abstract Viewer/Itinerary Planner. Atlanta, GA: Society for Neuroscience Annual Meeting; 2006. Peripheral demyelination injury induces upregulation of chemokine/receptor expression and neuronal signaling in a model of neuropathic pain; p. 250.3.
    1. White FA, Ripsch M, Bhangoo S, Ren D, Weiss C, Miller RJ. Abstract Viewer/Itinerary Planner. Washington, DC: Society for Neuroscience Annual Meeting; 2005. Regulation of chemokines/receptors in the dorsal root ganglion following focal demyelination of the sciatic nerve; p. 748.9.
    1. Zou D, Dasse O, Evans J, Higgins P, Kintigh J, Knerr L, Kondru R, Schwartz E, Zhai H-X. Book Pyrrolidinone derivatives. City: NORRIS, Allen; UCB, S.A. Intellectual Property Department Allée de la Recherche, 60 B-1070 Bruxelles (BE); 2003. Pyrrolidinone derivatives.
    1. Ma C, Shu Y, Zheng Z, Chen Y, Yao H, Greenquist KW, White FA, LaMotte RH. Similar electrophysiological changes in axotomized and neighboring intact dorsal root ganglion neurons. J Neurophysiol. 2003;89:1588–602. doi: 10.1152/jn.00855.2002.
    1. Lu M, Grove EA, Miller RJ. Abnormal development of the hippocampal dentate gyrus in mice lacking the CXCR4 chemokine receptor. Proc Natl Acad Sci USA. 2002;99:7090–5. doi: 10.1073/pnas.092013799.
    1. Ma C, LaMotte RH. Enhanced excitability of dissociated primary sensory neurons after chronic compression of the dorsal root ganglion in the rat. Pain. 2005;113:106–12. doi: 10.1016/j.pain.2004.10.001.
    1. Fang X, Djouhri L, McMullan S, Berry C, Waxman SG, Okuse K, Lawson SN. Intense isolectin-B4 binding in rat dorsal root ganglion neurons distinguishes C-fiber nociceptors with broad action potentials and high Nav1.9 expression. J Neurosci. 2006;26:7281–92. doi: 10.1523/JNEUROSCI.1072-06.2006.
    1. Petruska JC, Napaporn J, Johnson RD, Gu JG, Cooper BY. Subclassified acutely dissociated cells of rat DRG: histochemistry and patterns of capsaicin-, proton-, and ATP-activated currents. J Neurophysiol. 2000;84:2365–79.
    1. Streit WJ, Schulte BA, Balentine JD, Spicer SS. Evidence for glycoconjugate in nociceptive primary sensory neurons and its origin from the Golgi complex. Brain Res. 1986;377:1–17. doi: 10.1016/0006-8993(86)91185-6.
    1. Jong EK de, Dijkstra IM, Hensens M, Brouwer N, van Amerongen M, Liem RS, Boddeke HW, Biber K. Vesicle-mediated transport and release of CCL21 in endangered neurons: a possible explanation for microglia activation remote from a primary lesion. J Neurosci. 2005;25:7548–57. doi: 10.1523/JNEUROSCI.1019-05.2005.
    1. Gleichmann M, Gillen C, Czardybon M, Bosse F, Greiner-Petter R, Auer J, Muller HW. Cloning and characterization of SDF-1gamma, a novel SDF-1 chemokine transcript with developmentally regulated expression in the nervous system. Eur J Neurosci. 2000;12:1857–66. doi: 10.1046/j.1460-9568.2000.00048.x.
    1. Kury P, Greiner-Petter R, Cornely C, Jurgens T, Muller HW. Mammalian achaete scute homolog 2 is expressed in the adult sciatic nerve and regulates the expression of Krox24, Mob-1, CXCR4, and p57kip2 in Schwann cells. J Neurosci. 2002;22:7586–95.
    1. Kury P, Koller H, Hamacher M, Cornely C, Hasse B, Muller HW. Cyclic AMP and tumor necrosis factor-alpha regulate CXCR4 gene expression in Schwann cells. Mol Cell Neurosci. 2003;24:1–9. doi: 10.1016/S1044-7431(03)00132-5.
    1. Fujioka T, Purev E, Rostami A. Chemokine mRNA expression in the cauda equina of Lewis rats with experimental allergic neuritis. J Neuroimmunol. 1999;97:51–9. doi: 10.1016/S0165-5728(99)00048-X.
    1. Tran PB, Ren D, Veldhouse TJ, Miller RJ. Chemokine receptors are expressed widely by embryonic and adult neural progenitor cells. J Neurosci Res. 2004;76:20–34. doi: 10.1002/jnr.20001.
    1. White FA, Bhangoo SK, Miller RJ. Chemokines: integrators of pain and inflammation. Nat Rev Drug Discov. 2005;4:834–44. doi: 10.1038/nrd1852.
    1. Ji JF, He BP, Dheen ST, Tay SS. Interactions of chemokines and chemokine receptors mediate the migration of mesenchymal stem cells to the impaired site in the brain after hypoglossal nerve injury. Stem Cells. 2004;22:415–27. doi: 10.1634/stemcells.22-3-415.
    1. Cole KE, Strick CA, Paradis TJ, Ogborne KT, Loetscher M, Gladue RP, Lin W, Boyd JG, Moser B, Wood DE, et al. Interferon-inducible T cell alpha chemoattractant (I-TAC): a novel non-ELR CXC chemokine with potent activity on activated T cells through selective high affinity binding to CXCR3. J Exp Med. 1998;187:2009–21. doi: 10.1084/jem.187.12.2009.
    1. Loetscher M, Gerber B, Loetscher P, Jones SA, Piali L, Clark-Lewis I, Baggiolini M, Moser B. Chemokine receptor specific for IP10 and mig: structure, function, and expression in activated T-lymphocytes. J Exp Med. 1996;184:963–9. doi: 10.1084/jem.184.3.963.
    1. Weng Y, Siciliano SJ, Waldburger KE, Sirotina-Meisher A, Staruch MJ, Daugherty BL, Gould SL, Springer MS, DeMartino JA. Binding and functional properties of recombinant and endogenous CXCR3 chemokine receptors. J Biol Chem. 1998;273:18288–91. doi: 10.1074/jbc.273.29.18288.
    1. Obata K, Yamanaka H, Fukuoka T, Yi D, Tokunaga A, Hashimoto N, Yoshikawa H, Noguchi K. Contribution of injured and uninjured dorsal root ganglion neurons to pain behavior and the changes in gene expression following chronic constriction injury of the sciatic nerve in rats. Pain. 2003;101:65–77. doi: 10.1016/S0304-3959(02)00296-8.
    1. Oh SB, Endoh T, Simen AA, Ren D, Miller RJ. Regulation of calcium currents by chemokines and their receptors. J Neuroimmunol. 2002;123:66–75. doi: 10.1016/S0165-5728(01)00485-4.
    1. Amir R, Michaelis M, Devor M. Burst discharge in primary sensory neurons: triggered by subthreshold oscillations, maintained by depolarizing afterpotentials. J Neurosci. 2002;22:1187–98.
    1. Devor M. Neuropathic pain: what do we do with all these theories? Acta Anaesthesiol Scand. 2001;45:1121–7. doi: 10.1034/j.1399-6576.2001.450912.x.
    1. Kajander KC, Bennett GJ. Onset of a painful peripheral neuropathy in rat: a partial and differential deafferentation and spontaneous discharge in A beta and A delta primary afferent neurons. J Neurophysiol. 1992;68:734–44.
    1. Milligan ED, Zapata V, Chacur M, Schoeniger D, Biedenkapp J, O'Connor KA, Verge GM, Chapman G, Green P, Foster AC, et al. Evidence that exogenous and endogenous fractalkine can induce spinal nociceptive facilitation in rats. Eur J Neurosci. 2004;20:2294–302. doi: 10.1111/j.1460-9568.2004.03709.x.
    1. 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;20:1150–60. doi: 10.1111/j.1460-9568.2004.03593.x.
    1. Zhang J, De Koninck Y. Spatial and temporal relationship between monocyte chemoattractant protein-1 expression and spinal glial activation following peripheral nerve injury. J Neurochem. 2006;97:772–83. doi: 10.1111/j.1471-4159.2006.03746.x.
    1. Raghavendra V, Tanga F, DeLeo JA. Inhibition of microglial activation attenuates the development but not existing hypersensitivity in a rat model of neuropathy. J Pharmacol Exp Ther. 2003;306:624–30. doi: 10.1124/jpet.103.052407.
    1. Saccani A, Saccani S, Orlando S, Sironi M, Bernasconi S, Ghezzi P, Mantovani A, Sica A. Redox regulation of chemokine receptor expression. Proc Natl Acad Sci USA. 2000;97:2761–6. doi: 10.1073/pnas.97.6.2761.
    1. Zhang X, Meister B, Elde R, Verge VM, Hokfelt T. Large calibre primary afferent neurons projecting to the gracile nucleus express neuropeptide Y after sciatic nerve lesions: an immunohistochemical and in situ hybridization study in rats. Eur J Neurosci. 1993;5:1510–9. doi: 10.1111/j.1460-9568.1993.tb00219.x.
    1. Menetski J, Mistry S, Lu M, Mudgett JS, Ransohoff RM, Demartino JA, Macintyre DE, Abbadie C. Mice overexpressing chemokine ligand 2 (CCL2) in astrocytes display enhanced nociceptive responses. Neuroscience. 2007;149:706–14. doi: 10.1016/j.neuroscience.2007.08.014.
    1. Zhang J, Shi XQ, Echeverry S, Mogil JS, De Koninck Y, Rivest S. Expression of CCR2 in both resident and bone marrow-derived microglia plays a critical role in neuropathic pain. J Neurosci. 2007;27:12396–406. doi: 10.1523/JNEUROSCI.3016-07.2007.
    1. Hirakawa H, Okajima S, Nagaoka T, Kubo T, Takamatsu T, Oyamada M. Regional differences in blood-nerve barrier function and tight-junction protein expression within the rat dorsal root ganglion. Neuroreport. 2004;15:405–8. doi: 10.1097/00001756-200403010-00004.
    1. Keswani SC, Polley M, Pardo CA, Griffin JW, McArthur JC, Hoke A. Schwann cell chemokine receptors mediate HIV-1 gp120 toxicity to sensory neurons. Ann Neurol. 2003;54:287–96. doi: 10.1002/ana.10645.
    1. Charo IF, Ransohoff RM. The many roles of chemokines and chemokine receptors in inflammation. N Engl J Med. 2006;354:610–21. doi: 10.1056/NEJMra052723.
    1. Szabo I, Chen XH, Xin L, Adler MW, Howard OM, Oppenheim JJ, Rogers TJ. Heterologous desensitization of opioid receptors by chemokines inhibits chemotaxis and enhances the perception of pain. Proc Natl Acad Sci USA. 2002;99:10276–81. doi: 10.1073/pnas.102327699.
    1. Zhang N, Rogers TJ, Caterina M, Oppenheim JJ. Proinflammatory chemokines, such as C-C chemokine ligand 3, desensitize mu-opioid receptors on dorsal root ganglia neurons. J Immunol. 2004;173:594–9.
    1. 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;6:434–8. doi: 10.1016/j.jpain.2005.02.001.
    1. Biber K, Rappert A, Kettenmann H, Brouwer N, Copray SC, Boddeke HW. Neuronal SLC (CCL21) expression: implications for the neuron-microglial signaling system. Ernst Schering Res Found Workshop. 2002:45–60.
    1. Wang X, Li X, Schmidt DB, Foley JJ, Barone FC, Ames RS, Sarau HM. Identification and molecular characterization of rat CXCR3: receptor expression and interferon-inducible protein-10 binding are increased in focal stroke. Mol Pharmacol. 2000;57:1190–8.
    1. Ahn DK, Kim KH, Jung CY, Choi HS, Lim EJ, Youn DH, Bae YC. Role of peripheral group I and II metabotropic glutamate receptors in IL-1beta-induced mechanical allodynia in the orofacial area of conscious rats. Pain. 2005;118:53–60. doi: 10.1016/j.pain.2005.07.017.
    1. Attal N, Filliatreau G, Perrot S, Jazat F, Di Giamberardino L, Guilbaud G. Behavioural pain-related disorders and contribution of the saphenous nerve in crush and chronic constriction injury of the rat sciatic nerve. Pain. 1994;59:301–12. doi: 10.1016/0304-3959(94)90083-3.
    1. Chacur M, Milligan ED, Gazda LS, Armstrong C, Wang H, Tracey KJ, Maier SF, Watkins LR. A new model of sciatic inflammatory neuritis (SIN): induction of unilateral and bilateral mechanical allodynia following acute unilateral peri-sciatic immune activation in rats. Pain. 2001;94:231–44. doi: 10.1016/S0304-3959(01)00354-2.
    1. Ledeboer A, Mahoney JH, Milligan ED, Martin D, Maier SF, Watkins LR. Spinal cord glia and interleukin-1 do not appear to mediate persistent allodynia induced by intramuscular acidic saline in rats. J Pain. 2006;7:757–67. doi: 10.1016/j.jpain.2006.04.001.
    1. Seltzer Z, Dubner R, Shir Y. A novel behavioral model of neuropathic pain disorders produced in rats by partial sciatic nerve injury. Pain. 1990;43:205–18. doi: 10.1016/0304-3959(90)91074-S.
    1. Sluka KA, Kalra A, Moore SA. Unilateral intramuscular injections of acidic saline produce a bilateral, long-lasting hyperalgesia. Muscle Nerve. 2001;24:37–46. doi: 10.1002/1097-4598(200101)24:1<37::AID-MUS4>;2-8.
    1. Sotgiu ML, Biella G. Differential effects of MK-801, a N-methyl-D-aspartate non-competitive antagonist, on the dorsal horn neuron hyperactivity and hyperexcitability in neuropathic rats. Neurosci Lett. 2000;283:153–6. doi: 10.1016/S0304-3940(00)00941-1.
    1. Xie WR, Deng H, Li H, Bowen TL, Strong JA, Zhang JM. Robust increase of cutaneous sensitivity, cytokine production and sympathetic sprouting in rats with localized inflammatory irritation of the spinal ganglia. Neuroscience. 2006;142:809–22. doi: 10.1016/j.neuroscience.2006.06.045.
    1. Yasuda T, Miki S, Yoshinaga N, Senba E. Effects of amitriptyline and gabapentin on bilateral hyperalgesia observed in an animal model of unilateral axotomy. Pain. 2005;115:161–70. doi: 10.1016/j.pain.2005.02.026.
    1. Yu LC, Hansson P, Lundeberg T. The calcitonin gene-related peptide antagonist CGRP8-37 increases the latency to withdrawal responses bilaterally in rats with unilateral experimental mononeuropathy, an effect reversed by naloxone. Neuroscience. 1996;71:523–31. doi: 10.1016/0306-4522(95)00428-9.
    1. Kleinschnitz C, Brinkhoff J, Sommer C, Stoll G. Contralateral cytokine gene induction after peripheral nerve lesions: dependence on the mode of injury and NMDA receptor signaling. Brain Res Mol Brain Res. 2005;136:23–8. doi: 10.1016/j.molbrainres.2004.12.015.
    1. Milligan ED, Twining C, Chacur M, Biedenkapp J, O'Connor K, Poole S, Tracey K, Martin D, Maier SF, Watkins LR. Spinal glia and proinflammatory cytokines mediate mirror-image neuropathic pain in rats. J Neurosci. 2003;23:1026–40.
    1. Watkins LR, Martin D, Ulrich P, Tracey KJ, Maier SF. Evidence for the involvement of spinal cord glia in subcutaneous formalin induced hyperalgesia in the rat. Pain. 1997;71:225–35. doi: 10.1016/S0304-3959(97)03369-1.
    1. 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. doi: 10.1038/nature04223.
    1. Holmes GM, Hebert SL, Rogers RC, Hermann GE. Immunocytochemical localization of TNF type 1 and type 2 receptors in the rat spinal cord. Brain Res. 2004;1025:210–9. doi: 10.1016/j.brainres.2004.08.020.
    1. Twining CM, Sloane EM, Milligan ED, Chacur M, Martin D, Poole S, Marsh H, Maier SF, Watkins LR. Peri-sciatic proinflammatory cytokines, reactive oxygen species, and complement induce mirror-image neuropathic pain in rats. Pain. 2004;110:299–309. doi: 10.1016/j.pain.2004.04.008.
    1. Spataro LE, Sloane EM, Milligan ED, Wieseler-Frank J, Schoeniger D, Jekich BM, Barrientos RM, Maier SF, Watkins LR. Spinal gap junctions: potential involvement in pain facilitation. J Pain. 2004;5:392–405. doi: 10.1016/j.jpain.2004.06.006.
    1. Jung H, Miller RJ. Activation of the nuclear factor of activated T-cells (NFAT) mediates upregulation of CCR2 chemokine receptors in dorsal root ganglion (DRG) neurons: A possible mechanism for activity-dependent transcription in DRG neurons in association with neuropathic pain. Mol Cell Neurosci. 2007.
    1. Obata K, Katsura H, Sakurai J, Kobayashi K, Yamanaka H, Dai Y, Fukuoka T, Noguchi K. Suppression of the p75 neurotrophin receptor in uninjured sensory neurons reduces neuropathic pain after nerve injury. J Neurosci. 2006;26:11974–86. doi: 10.1523/JNEUROSCI.3188-06.2006.
    1. Obata K, Yamanaka H, Kobayashi K, Dai Y, Mizushima T, Katsura H, Fukuoka T, Tokunaga A, Noguchi K. Role of mitogen-activated protein kinase activation in injured and intact primary afferent neurons for mechanical and heat hypersensitivity after spinal nerve ligation. J Neurosci. 2004;24:10211–22. doi: 10.1523/JNEUROSCI.3388-04.2004.
    1. Chaplan SR, Guo HQ, Lee DH, Luo L, Liu C, Kuei C, Velumian AA, Butler MP, Brown SM, Dubin AE. Neuronal hyperpolarization-activated pacemaker channels drive neuropathic pain. J Neurosci. 2003;23:1169–78.
    1. Liu CN, Devor M, Waxman SG, Kocsis JD. Subthreshold oscillations induced by spinal nerve injury in dissociated muscle and cutaneous afferents of mouse DRG. J Neurophysiol. 2002;87:2009–17.
    1. Liu CN, Wall PD, Ben-Dor E, Michaelis M, Amir R, Devor M. Tactile allodynia in the absence of C-fiber activation: altered firing properties of DRG neurons following spinal nerve injury. Pain. 2000;85:503–21. doi: 10.1016/S0304-3959(00)00251-7.
    1. Wallace VC, Blackbeard J, Segerdahl AR, Hasnie F, Pheby T, McMahon SB, Rice AS. Characterization of rodent models of HIV-gp120 and anti-retroviral-associated neuropathic pain. Brain. 2007;130:2688–702. doi: 10.1093/brain/awm195.
    1. Vogel C, Mossner R, Gerlach M, Heinemann T, Murphy DL, Riederer P, Lesch KP, Sommer C. Absence of thermal hyperalgesia in serotonin transporter-deficient mice. J Neurosci. 2003;23:708–15.

Source: PubMed

3
Subscribe