期刊论文详细信息
Journal of Neuroinflammation
Active immunization with myelin-derived altered peptide ligand reduces mechanical pain hypersensitivity following peripheral nerve injury
Gila Moalem-Taylor1  Vasso Apostolopoulos3  Barbara Cameron2  Cristina F Kim1  Justin G Lees1  Samuel S Duffy1  Chamini J Perera1 
[1] School of Medical Sciences, University of New South Wales, UNSW Medicine, Sydney 2052, NSW, Australia;Centre for Infection and Inflammation Research, School of Medical Sciences, University of New South Wales, Sydney 2052, NSW, Australia;College of Health and Biomedicine, Centre for Chronic Disease Prevention and Management, Victoria University, Melbourne, VIC, Australia
关键词: M1 and M2 macrophages;    Anti-inflammatory cytokines;    Pro-inflammatory cytokines;    Mechanical allodynia;    Altered peptide ligand;    Myelin basic protein;    Neuropathic pain;   
Others  :  1137613
DOI  :  10.1186/s12974-015-0253-4
 received in 2014-10-31, accepted in 2015-01-22,  发布年份 2015
PDF
【 摘 要 】

Background

T cells have been implicated in neuropathic pain that is caused by peripheral nerve injury. Immunogenic myelin basic protein (MBP) peptides have been shown to initiate mechanical allodynia in a T cell-dependent manner. Antagonistic altered peptide ligands (APLs) are peptides with substitutions in amino acid residues at T cell receptor contact sites and can inhibit T cell function and modulate inflammatory responses. In the present study, we studied the effects of immunization with MBP-derived APL on pain behavior and neuroinflammation in an animal model of peripheral nerve injury.

Methods

Lewis rats were immunized subcutaneously at the base of the tail with either a weakly encephalitogenic peptide of MBP (cyclo-MBP87-99) or APL (cyclo-(87-99)[A91,A96]MBP87-99) in complete Freund’s adjuvant (CFA) or CFA only (control), following chronic constriction injury (CCI) of the left sciatic nerve. Pain hypersensitivity was tested by measurements of paw withdrawal threshold to mechanical stimuli, regulatory T cells in spleen and lymph nodes were analyzed by flow cytometry, and immune cell infiltration into the nervous system was assessed by immunohistochemistry (days 10 and 30 post-CCI). Cytokines were measured in serum and nervous tissue of nerve-injured rats (day 10 post-CCI).

Results

Rats immunized with the APL cyclo-(87-99)[A91,A96]MBP87-99 had significantly reduced mechanical pain hypersensitivity in the ipsilateral hindpaw compared to cyclo-MBP87-99-treated and control rats. This was associated with significantly decreased infiltration of T cells and ED1+ macrophages in the injured nerve of APL-treated animals. The percentage of anti-inflammatory (M2) macrophages was significantly upregulated in the APL-treated rats on day 30 post-CCI. Compared to the control rats, microglial activation in the ipsilateral lumbar spinal cord was significantly increased in the MBP-treated rats, but was not altered in the rats immunized with the MBP-derived APL. In addition, immunization with the APL significantly increased splenic regulatory T cells. Several cytokines were significantly altered after CCI, but no significant difference was observed between the APL-treated and control rats.

Conclusions

These results suggest that immune deviation by active immunization with a non-encephalitogenic MBP-derived APL mediates an analgesic effect in animals with peripheral nerve injury. Thus, T cell immunomodulation warrants further investigation as a possible therapeutic strategy for the treatment of peripheral neuropathic pain.

【 授权许可】

   
2015 Perera et al.; licensee BioMed Central.

【 预 览 】
附件列表
Files Size Format View
20150317105253422.pdf 2097KB PDF download
Figure 10. 20KB Image download
Figure 9. 283KB Image download
Figure 8. 110KB Image download
Figure 7. 158KB Image download
Figure 6. 20KB Image download
Figure 5. 96KB Image download
Figure 4. 65KB Image download
Figure 3. 101KB Image download
Figure 2. 68KB Image download
Figure 1. 38KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 4.

Figure 5.

Figure 6.

Figure 7.

Figure 8.

Figure 9.

Figure 10.

【 参考文献 】
  • [1]Dworkin RH, Backonja M, Rowbotham MC, Allen RR, Argoff CR, Bennett GJ, Bushnell MC, Farrar JT, Galer BS, Haythornthwaite JA, Hewitt DJ, Loeser JD, Max MB, Saltarelli M, Schmader KE, Stein C, Thompson D, Turk DC, Wallace MS, Watkins LR, Weinstein SM: Advances in neuropathic pain: diagnosis, mechanisms, and treatment recommendations. Arch Neurol 2003, 60:1524-34.
  • [2]Myers RR, Campana WM, Shubayev VI: The role of neuroinflammation in neuropathic pain: mechanisms and therapeutic targets. Drug Discov Today 2006, 11:8-20.
  • [3]Gao YJ, Ji RR: Chemokines, neuronal-glial interactions, and central processing of neuropathic pain. Pharmacol Ther 2010, 126:56-68.
  • [4]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, 229:26-50.
  • [5]Costigan M, Moss A, Latremoliere A, Johnston C, Verma-Gandhu M, Herbert TA, Barrett L, Brenner GJ, Vardeh D, Woolf CJ, Fitzgerald M: T-cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. J Neurosci 2009, 29:14415-22.
  • [6]Moalem G, Xu K, Yu L: T lymphocytes play a role in neuropathic pain following peripheral nerve injury in rats. Neuroscience 2004, 129:767-77.
  • [7]Cui JG, Holmin S, Mathiesen T, Meyerson BA, Linderoth B: Possible role of inflammatory mediators in tactile hypersensitivity in rat models of mononeuropathy. Pain 2000, 88:239-48.
  • [8]Hu P, Bembrick AL, Keay KA, McLachlan EM: Immune cell involvement in dorsal root ganglia and spinal cord after chronic constriction or transection of the rat sciatic nerve. Brain Behav Immun 2007, 21:599-616.
  • [9]Hu P, McLachlan EM: Macrophage and lymphocyte invasion of dorsal root ganglia after peripheral nerve lesions in the rat. Neuroscience 2002, 112:23-38.
  • [10]Cao L, DeLeo JA: CNS-infiltrating CD4+ T lymphocytes contribute to murine spinal nerve transection-induced neuropathic pain. Eur J Immunol 2008, 38:448-58.
  • [11]Draleau K, Maddula S, Slaiby A, Nutile-McMenemy N, De Leo J, Cao L: Phenotypic Identification of Spinal Cord-Infiltrating CD4 T Lymphocytes in a Murine Model of Neuropathic Pain. J Pain Relief Suppl 2014, 3:003.
  • [12]Kim CF, Moalem-Taylor G: Detailed characterization of neuro-immune responses following neuropathic injury in mice. Brain Res 2011, 1405:95-108.
  • [13]Austin PJ, Kim CF, Perera CJ, Moalem-Taylor G: Regulatory T cells attenuate neuropathic pain following peripheral nerve injury and experimental autoimmune neuritis. Pain 2012, 153:1916-31.
  • [14]Kalergis AM, Nathenson SG: Altered peptide ligand-mediated TCR antagonism can be modulated by a change in a single amino acid residue within the CDR3 beta of an MHC class I-restricted TCR. J Immunol 2000, 165:280-5.
  • [15]Katsara M, Minigo G, Plebanski M, Apostolopoulos V: The good, the bad and the ugly: how altered peptide ligands modulate immunity. Expert Opin Biol Ther 2008, 8:1873-84.
  • [16]Gaur A, Boehme SA, Chalmers D, Crowe PD, Pahuja A, Ling N, Brocke S, Steinman L, Conlon PJ: Amelioration of relapsing experimental autoimmune encephalomyelitis with altered myelin basic protein peptides involves different cellular mechanisms. J Neuroimmunol 1997, 74:149-58.
  • [17]Katsara M, Yuriev E, Ramsland PA, Deraos G, Tselios T, Matsoukas J, Apostolopoulos V: Mannosylation of mutated MBP83-99 peptides diverts immune responses from Th1 to Th2. Mol Immunol 2008, 45:3661-70.
  • [18]Karin N, Mitchell DJ, Brocke S, Ling N, Steinman L: Reversal of experimental autoimmune encephalomyelitis by a soluble peptide variant of a myelin basic protein epitope: T cell receptor antagonism and reduction of interferon gamma and tumor necrosis factor alpha production. J Exp Med 1994, 180:2227-37.
  • [19]Young DA, Lowe LD, Booth SS, Whitters MJ, Nicholson L, Kuchroo VK, Collins M: IL-4, IL-10, IL-13, and TGF-beta from an altered peptide ligand-specific Th2 cell clone down-regulate adoptive transfer of experimental autoimmune encephalomyelitis. J Immunol 2000, 164:3563-72.
  • [20]Nicholson LB, Murtaza A, Hafler BP, Sette A, Kuchroo VK: A T cell receptor antagonist peptide induces T cells that mediate bystander suppression and prevent autoimmune encephalomyelitis induced with multiple myelin antigens. Proc Natl Acad Sci U S A 1997, 94:9279-84.
  • [21]Nicholson LB, Greer JM, Sobel RA, Lees MB, Kuchroo VK: An altered peptide ligand mediates immune deviation and prevents autoimmune encephalomyelitis. Immunity 1995, 3:397-405.
  • [22]Mantzourani ED, Tselios TV, Grdadolnik SG, Platts JA, Brancale A, Deraos GN, Matsoukas JM, Mavromoustakos TM: Comparison of Proposed Putative Active Conformations of Myelin Basic Protein Epitope 87–99 Linear Altered Peptide Ligands by Spectroscopic and Modelling Studies: The Role of Positions 91 and 96 in T-Cell Receptor Activation. J Med Chem 2006, 49:6683-91.
  • [23]Matsoukas J, Apostolopoulos V, Kalbacher H, Papini A-M, Tselios T, Chatzantoni K, Biagioli T, Lolli F, Deraos S, Papathanassopoulos P, Troganis A, Mantzourani E, Mavromoustakos T, Mouzaki A: Design And Synthesis of a Novel Potent Myelin Basic Protein Epitope 87–99 Cyclic Analogue: Enhanced Stability and Biological Properties of Mimics Render Them a Potentially New Class of Immunomodulators†. J Med Chem 2005, 48:1470-80.
  • [24]Tian DH, Perera CJ, Apostolopoulos V, Moalem-Taylor G: Effects of vaccination with altered peptide ligand on chronic pain in experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. Front Neurol 2013, 4:168.
  • [25]Hauben E, Agranov E, Gothilf A, Nevo U, Cohen A, Smirnov I, Steinman L, Schwartz M: Posttraumatic therapeutic vaccination with modified myelin self-antigen prevents complete paralysis while avoiding autoimmune disease. J Clin Invest 2001, 108:591-9.
  • [26]Lewitus GM, Wilf-Yarkoni A, Ziv Y, Shabat-Simon M, Gersner R, Zangen A, Schwartz M: Vaccination as a novel approach for treating depressive behavior. Biol Psychiatry 2009, 65:283-8.
  • [27]Kobayashi H, Chattopadhyay S, Kato K, Dolkas J, Kikuchi S-i, Myers RR, Shubayev VI: MMPs initiate Schwann cell-mediated MBP degradation and mechanical nociception after nerve damage. MolCell Neurosci 2008, 39:619-27.
  • [28]Liu H, Shiryaev SA, Chernov AV, Kim Y, Shubayev I, Remacle AG, Baranovskaya S, Golubkov VS, Strongin AY, Shubayev VI: Immunodominant fragments of myelin basic protein initiate T cell-dependent pain. J Neuroinflammation 2012, 9:119. BioMed Central Full Text
  • [29]Bennett GJ, Xie YK: A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 1988, 33:87-107.
  • [30]Raghavendra V, Tanga FY, DeLeo JA: Complete Freunds adjuvant-induced peripheral inflammation evokes glial activation and proinflammatory cytokine expression in the CNS. Eur J Neurosci 2004, 20:467-73.
  • [31]Gould KE, Swanborg RH: T and B cell responses to myelin basic protein and encephalitogenic epitopes. J Neuroimmunol 1993, 46:193-8.
  • [32]Katsara M, Yuriev E, Ramsland PA, Tselios T, Deraos G, Lourbopoulos A, Grigoriadis N, Matsoukas J, Apostolopoulos V: Altered peptide ligands of myelin basic protein (MBP87-99) conjugated to reduced mannan modulate immune responses in mice. Immunology 2009, 128:521-33.
  • [33]Moalem-Taylor G, Allbutt HN, Iordanova MD, Tracey DJ: Pain hypersensitivity in rats with experimental autoimmune neuritis, an animal model of human inflammatory demyelinating neuropathy. Brain Behav Immun 2007, 21:699-710.
  • [34]Liu T, van Rooijen N, Tracey DJ: Depletion of macrophages reduces axonal degeneration and hyperalgesia following nerve injury. Pain 2000, 86:25-32.
  • [35]Barclay J, Clark AK, Ganju P, Gentry C, Patel S, Wotherspoon G, Buxton F, Song C, Ullah J, Winter J, Fox A, Bevan S, Malcangio M: Role of the cysteine protease cathepsin S in neuropathic hyperalgesia. Pain 2007, 130:225-34.
  • [36]Mert T, Gunay I, Ocal I, Guzel AI, Inal TC, Sencar L, Polat S: Macrophage depletion delays progression of neuropathic pain in diabetic animals. Naunyn Schmiedebergs Arch Pharmacol 2009, 379:445-52.
  • [37]Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popovich PG: Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci 2009, 29:13435-44.
  • [38]Ibarra A, Sosa M, García E, Flores A, Cruz Y, Mestre H, Martiñón S, Pineda-Rodríguez BA, Gutiérrez-Ospina G: Prophylactic neuroprotection with A91 improves the outcome of spinal cord injured rats. Neurosci Lett 2013, 554:59-63.
  • [39]Martiñon S, García E, Flores N, Gonzalez I, Ortega T, Buenrostro M, Reyes R, Fernandez-Presas AM, Guizar-Sahagún G, Correa D, Ibarra A: Vaccination with a neural-derived peptide plus administration of glutathione improves the performance of paraplegic rats. Eur J Neurosci 2007, 26:403-12.
  • [40]Sloan-Lancaster J, Shaw AS, Rothbard JB, Allen PM: Partial T cell signaling: altered phospho-zeta and lack of zap70 recruitment in APL-induced T cell anergy. Cell 1994, 79:913-22.
  • [41]von Herrath MG, Harrison LC: Antigen-induced regulatory T cells in autoimmunity. Nat Rev Immunol 2003, 3:223-32.
  • [42]McGuirk P, Mills KH: Pathogen-specific regulatory T cells provoke a shift in the Th1/Th2 paradigm in immunity to infectious diseases. Trends Immunol 2002, 23:450-5.
  • [43]Zhang X, Wu Z, Hayashi Y, Okada R, Nakanishi H: Peripheral role of cathepsin S in Th1 cell-dependent transition of nerve injury-induced acute pain to a chronic pain state. J Neurosci 2014, 34:3013-22.
  • [44]Cao X, Cai SF, Fehniger TA, Song J, Collins LI, Piwnica-Worms DR, Ley TJ: Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity 2007, 27:635-46.
  • [45]Ydens E, Cauwels A, Asselbergh B, Goethals S, Peeraer L, Lornet G, Almeida-Souza L, Van Ginderachter JA, Timmerman V, Janssens S: Acute injury in the peripheral nervous system triggers an alternative macrophage response. J Neuroinflammation 2012, 9:176. BioMed Central Full Text
  • [46]Turtzo LC, Lescher J, Janes L, Dean DD, Budde MD, Frank JA: Macrophagic and microglial responses after focal traumatic brain injury in the female rat. J Neuroinflammation 2014, 11:82. BioMed Central Full Text
  • [47]Hsieh CL, Kim CC, Ryba BE, Niemi EC, Bando JK, Locksley RM, Liu J, Nakamura MC, Seaman WE: Traumatic brain injury induces macrophage subsets in the brain. Eur J Immunol 2013, 43:2010-22.
  • [48]Wang G, Zhang J, Hu X, Zhang L, Mao L, Jiang X, Liou AK, Leak RK, Gao Y, Chen J: Microglia/macrophage polarization dynamics in white matter after traumatic brain injury. J Cereb Blood Flow Metab 2013, 33:1864-74.
  • [49]Shechter R, London A, Varol C, Raposo C, Cusimano M, Yovel G, Rolls A, Mack M, Pluchino S, Martino G, Jung S, Schwartz M: Infiltrating blood-derived macrophages are vital cells playing an anti-inflammatory role in recovery from spinal cord injury in mice. PLoS Med 2009, 6:e1000113.
  • [50]Martinez FO, Helming L, Gordon S: Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol 2009, 27:451-83.
  • [51]Weber MS, Prod’homme T, Youssef S, Dunn SE, Rundle CD, Lee L, Patarroyo JC, Stuve O, Sobel RA, Steinman L, Zamvil SS: Type II monocytes modulate T cell-mediated central nervous system autoimmune disease. Nat Med 2007, 13:935-43.
  • [52]Qin H, Yeh W-I, De Sarno P, Holdbrooks AT, Liu Y, Muldowney MT, Reynolds SL, Yanagisawa LL, Fox TH, Park K, Harrington LE, Raman C, Benveniste EN: Signal transducer and activator of transcription-3/suppressor of cytokine signaling-3 (STAT3/SOCS3) axis in myeloid cells regulates neuroinflammation. Proc Natl Acad Sci 2012, 109:5004-9.
  • [53]Clark AK, Gentry C, Bradbury EJ, McMahon SB, Malcangio M: Role of spinal microglia in rat models of peripheral nerve injury and inflammation. Eur J Pain 2007, 11:223-30.
  • [54]Ledeboer A, Sloane EM, Milligan ED, Frank MG, Mahony JH, Maier SF, Watkins LR: Minocycline attenuates mechanical allodynia and proinflammatory cytokine expression in rat models of pain facilitation. Pain 2005, 115:71-83.
  • [55]Colburn RW, DeLeo JA, Rickman AJ, Yeager MP, Kwon P, Hickey WF: Dissociation of microglial activation and neuropathic pain behaviors following peripheral nerve injury in the rat. J Neuroimmunol 1997, 79:163-75.
  • [56]Zheng FY, Xiao WH, Bennett GJ: The response of spinal microglia to chemotherapy-evoked painful peripheral neuropathies is distinct from that evoked by traumatic nerve injuries. Neuroscience 2011, 176:447-54.
  • [57]Gritsch S, Lu J, Thilemann S, Wortge S, Mobius W, Bruttger J, Karram K, Ruhwedel T, Blanfeld M, Vardeh D, Waisman A, Nave KA, Kuner R: Oligodendrocyte ablation triggers central pain independently of innate or adaptive immune responses in mice. Nat Commun 2014, 5:5472.
  • [58]Ledeboer A, Jekich BM, Sloane EM, Mahoney JH, Langer SJ, Milligan ED, Martin D, Maier SF, Johnson KW, Leinwand LA, Chavez RA, Watkins LR: Intrathecal interleukin-10 gene therapy attenuates paclitaxel-induced mechanical allodynia and proinflammatory cytokine expression in dorsal root ganglia in rats. Brain Behav Immun 2007, 21:686-98.
  • [59]Mika J, Osikowicz M, Rojewska E, Korostynski M, Wawrzczak-Bargiela A, Przewlocki R, Przewlocka B: Differential activation of spinal microglial and astroglial cells in a mouse model of peripheral neuropathic pain. Eur J Pharmacol 2009, 623:65-72.
  • [60]Moalem-Taylor G, Li M, Allbutt H, Wu A, Tracey D: A preconditioning nerve lesion inhibits mechanical pain hypersensitivity following subsequent neuropathic injury. Molecular Pain 2011, 7:1. BioMed Central Full Text
  • [61]Bileviciute-Ljungar I, Biella G, Bellomi P, Sotgiu ML: Contralateral treatment with lidocaine reduces spinal neuronal activity in mononeuropathic rats. Neurosci Lett 2001, 311:157-60.
  • [62]Koltzenburg M, Wall PD, McMahon SB: Does the right side know what the left is doing? Trends Neurosci 1999, 22:122-7.
  • [63]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.
  • [64]Parkitny L, McAuley JH, Kelly PJ, Di Pietro F, Cameron B, Moseley GL: Multiplex cytokine concentration measurement: how much do the medium and handling matter? Mediators Inflamm 2013, 2013:890706.
  • [65]Mika J, Korostynski M, Kaminska D, Wawrzczak-Bargiela A, Osikowicz M, Makuch W, Przewlocki R, Przewlocka B: Interleukin-1alpha has antiallodynic and antihyperalgesic activities in a rat neuropathic pain model. Pain 2008, 138:587-97.
  • [66]Hopkins SJ, Rothwell NJ: Cytokines and the nervous system. I: expression and recognition. Trends Neurosci 1995, 18:83-8.
  • [67]Giulian D, Baker TJ, Shih LC, Lachman LB: Interleukin 1 of the central nervous system is produced by ameboid microglia. J Exp Med 1986, 164:594-604.
  • [68]Okamoto K, Martin DP, Schmelzer JD, Mitsui Y, Low PA: Pro- and anti-inflammatory cytokine gene expression in rat sciatic nerve chronic constriction injury model of neuropathic pain. Exp Neurol 2001, 169:386-91.
  • [69]Gillen C, Jander S, Stoll G: Sequential expression of mRNA for proinflammatory cytokines and interleukin-10 in the rat peripheral nervous system: comparison between immune-mediated demyelination and Wallerian degeneration. J Neurosci Res 1998, 51:489-96.
  • [70]Liu L, Yang TM, Liedtke W, Simon SA: Chronic IL-1beta signaling potentiates voltage-dependent sodium currents in trigeminal nociceptive neurons. J Neurophysiol 2006, 95:1478-90.
  • [71]Reeve AJ, Patel S, Fox A, Walker K, Urban L: Intrathecally administered endotoxin or cytokines produce allodynia, hyperalgesia and changes in spinal cord neuronal responses to nociceptive stimuli in the rat. Eur J Pain 2000, 4:247-57.
  • [72]Sung CS, Wen ZH, Chang WK, Ho ST, Tsai SK, Chang YC, Wong CS: Intrathecal interleukin-1beta administration induces thermal hyperalgesia by activating inducible nitric oxide synthase expression in the rat spinal cord. Brain Res 2004, 1015:145-53.
  • [73]Zhang RX, Li A, Liu B, Wang L, Ren K, Zhang H, Berman BM, Lao L: IL-1ra alleviates inflammatory hyperalgesia through preventing phosphorylation of NMDA receptor NR-1 subunit in rats. Pain 2008, 135:232-9.
  • [74]Wolf G, Gabay E, Tal M, Yirmiya R, Shavit Y: Genetic impairment of interleukin-1 signaling attenuates neuropathic pain, autotomy, and spontaneous ectopic neuronal activity, following nerve injury in mice. Pain 2006, 120:315-24.
  文献评价指标  
  下载次数:48次 浏览次数:14次