Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2012

Open Access 01-12-2012 | Research

Pain in experimental autoimmune encephalitis: a comparative study between different mouse models

Authors: Jianning Lu, Martina Kurejova, Laura N Wirotanseng, Ralf A Linker, Rohini Kuner, Anke Tappe-Theodor

Published in: Journal of Neuroinflammation | Issue 1/2012

Login to get access

Abstract

Background

Pain can be one of the most severe symptoms associated with multiple sclerosis (MS) and develops with varying levels and time courses. MS-related pain is difficult to treat, since very little is known about the mechanisms underlying its development. Animal models of experimental autoimmune encephalomyelitis (EAE) mimic many aspects of MS and are well-suited to study underlying pathophysiological mechanisms. Yet, to date very little is known about the sensory abnormalities in different EAE models. We therefore aimed to thoroughly characterize pain behavior of the hindpaw in SJL and C57BL/6 mice immunized with PLP139-151 peptide or MOG35-55 peptide respectively. Moreover, we studied the activity of pain-related molecules and plasticity-related genes in the spinal cord and investigated functional changes in the peripheral nerves using electrophysiology.

Methods

We analyzed thermal and mechanical sensitivity of the hindpaw in both EAE models during the whole disease course. Qualitative and quantitative immunohistochemical analysis of pain-related molecules and plasticity-related genes was performed on spinal cord sections at different timepoints during the disease course. Moreover, we investigated functional changes in the peripheral nerves using electrophysiology.

Results

Mice in both EAE models developed thermal hyperalgesia during the chronic phase of the disease. However, whereas SJL mice developed marked mechanical allodynia over the chronic phase of the disease, C57BL/6 mice developed only minor mechanical allodynia over the onset and peak phase of the disease. Interestingly, the magnitude of glial changes in the spinal cord was stronger in SJL mice than in C57BL/6 mice and their time course matched the temporal profile of mechanical hypersensitivity.

Conclusions

Diverse EAE models bearing genetic, clinical and histopathological heterogeneity, show different profiles of sensory and pathological changes and thereby enable studying the mechanistic basis and the diversity of changes in pain perception that are associated with distinct types of MS.
Literature
2.
go back to reference Stenager E, Knudsen L, Jensen K: Acute and chronic pain syndromes in multiple sclerosis. A 5-year follow-up study. Ital J Neurol Sci 1995, 16:629–632.CrossRefPubMed Stenager E, Knudsen L, Jensen K: Acute and chronic pain syndromes in multiple sclerosis. A 5-year follow-up study. Ital J Neurol Sci 1995, 16:629–632.CrossRefPubMed
3.
go back to reference Svendsen KB, Jensen TS, Overvad K, Hansen HJ, Koch-Henriksen N, Bach FW: Pain in patients with multiple sclerosis: a population-based study. Arch Neurol 2003, 60:1089–1094.CrossRef Svendsen KB, Jensen TS, Overvad K, Hansen HJ, Koch-Henriksen N, Bach FW: Pain in patients with multiple sclerosis: a population-based study. Arch Neurol 2003, 60:1089–1094.CrossRef
4.
go back to reference Stenager E, Knudsen L, Jensen K: Acute and chronic pain syndromes in multiple sclerosis. Acta Neurol Scand 1991, 84:197–200.CrossRefPubMed Stenager E, Knudsen L, Jensen K: Acute and chronic pain syndromes in multiple sclerosis. Acta Neurol Scand 1991, 84:197–200.CrossRefPubMed
5.
go back to reference Pöllmann W, Feneberg W, Erasmus LP: Pain in multiple sclerosis–a still underestimated problem. The 1 year prevalence of pain syndromes, significance and quality of care of multiple sclerosis inpatients. Nervenarzt 2004, 75:135–140.CrossRefPubMed Pöllmann W, Feneberg W, Erasmus LP: Pain in multiple sclerosis–a still underestimated problem. The 1 year prevalence of pain syndromes, significance and quality of care of multiple sclerosis inpatients. Nervenarzt 2004, 75:135–140.CrossRefPubMed
6.
go back to reference Hadjimichael O, Kerns RD, Rizzo MA, Cutter G, Vollmer T: Persistent pain and uncomfortable sensations in persons with multiple sclerosis. Pain 2007, 127:35–41.CrossRefPubMed Hadjimichael O, Kerns RD, Rizzo MA, Cutter G, Vollmer T: Persistent pain and uncomfortable sensations in persons with multiple sclerosis. Pain 2007, 127:35–41.CrossRefPubMed
7.
go back to reference Kenner M, Menon U, Elliott DG: Multiple sclerosis as a painful disease. Int Rev Neurobiol 2007, 79:303–321.CrossRefPubMed Kenner M, Menon U, Elliott DG: Multiple sclerosis as a painful disease. Int Rev Neurobiol 2007, 79:303–321.CrossRefPubMed
8.
go back to reference Osterberg A, Boivie J, Thuomas KA: Central pain in multiple sclerosis–prevalence and clinical characteristics. Eur J Pain 2005, 9:531–542.CrossRefPubMed Osterberg A, Boivie J, Thuomas KA: Central pain in multiple sclerosis–prevalence and clinical characteristics. Eur J Pain 2005, 9:531–542.CrossRefPubMed
9.
go back to reference Svendsen KB, Jensen TS, Hansen HJ, Bach FW: Sensory function and quality of life in patients with multiple sclerosis and pain. Pain 2005, 114:473–481.CrossRefPubMed Svendsen KB, Jensen TS, Hansen HJ, Bach FW: Sensory function and quality of life in patients with multiple sclerosis and pain. Pain 2005, 114:473–481.CrossRefPubMed
10.
go back to reference Wekerle H, Kojima K, Lannes-Vieira J, Lassmann H, Linington C: Animal models. Ann Neurol 1994,36(Suppl):S47–53.CrossRefPubMed Wekerle H, Kojima K, Lannes-Vieira J, Lassmann H, Linington C: Animal models. Ann Neurol 1994,36(Suppl):S47–53.CrossRefPubMed
11.
go back to reference Gold R, Linington C, Lassmann H: Understanding pathogenesis and therapy of multiple sclerosis via animal models: 70 years of merits and culprits in experimental autoimmune encephalomyelitis research. Brain 2006, 129:1953–1971.CrossRefPubMed Gold R, Linington C, Lassmann H: Understanding pathogenesis and therapy of multiple sclerosis via animal models: 70 years of merits and culprits in experimental autoimmune encephalomyelitis research. Brain 2006, 129:1953–1971.CrossRefPubMed
12.
go back to reference Kuerten S, Angelov DN: Comparing the CNS morphology and immunobiology of different EAE models in C57BL/6 mice - a step towards understanding the complexity of multiple sclerosis. Ann Anat 2008, 190:1–15.CrossRefPubMed Kuerten S, Angelov DN: Comparing the CNS morphology and immunobiology of different EAE models in C57BL/6 mice - a step towards understanding the complexity of multiple sclerosis. Ann Anat 2008, 190:1–15.CrossRefPubMed
13.
go back to reference Berger T, Weerth S, Kojima K, Linington C, Wekerle H, Lassmann H: Experimental autoimmune encephalomyelitis: the antigen specificity of T lymphocytes determines the topography of lesions in the central and peripheral nervous system. Lab Invest 1997, 76:355–364.PubMed Berger T, Weerth S, Kojima K, Linington C, Wekerle H, Lassmann H: Experimental autoimmune encephalomyelitis: the antigen specificity of T lymphocytes determines the topography of lesions in the central and peripheral nervous system. Lab Invest 1997, 76:355–364.PubMed
15.
go back to reference Aicher SA, Silverman MB, Winkler CW, Bebo BF Jr: Hyperalgesia in an animal model of multiple sclerosis. Pain 2004, 110:560–570.CrossRefPubMed Aicher SA, Silverman MB, Winkler CW, Bebo BF Jr: Hyperalgesia in an animal model of multiple sclerosis. Pain 2004, 110:560–570.CrossRefPubMed
16.
go back to reference Olechowski CJ, Truong JJ, Kerr BJ: Neuropathic pain behaviours in a chronic-relapsing model of experimental autoimmune encephalomyelitis (EAE). Pain 2009, 141:156–164.CrossRefPubMed Olechowski CJ, Truong JJ, Kerr BJ: Neuropathic pain behaviours in a chronic-relapsing model of experimental autoimmune encephalomyelitis (EAE). Pain 2009, 141:156–164.CrossRefPubMed
17.
go back to reference Rodrigues DH, Sachs D, Teixeira AL: Mechanical hypernociception in experimental autoimmune encephalomyelitis. Arq Neuropsiquiatr 2009, 67:78–81.CrossRefPubMed Rodrigues DH, Sachs D, Teixeira AL: Mechanical hypernociception in experimental autoimmune encephalomyelitis. Arq Neuropsiquiatr 2009, 67:78–81.CrossRefPubMed
18.
go back to reference Lisi L, Navarra P, Cirocchi R, Sharp A, Stigliano E, Feinstein DL, Dello Russo C: Rapamycin reduces clinical signs and neuropathic pain in a chronic model of experimental autoimmune encephalomyelitis. J Neuroimmunol 2012, 243:43–51.CrossRefPubMed Lisi L, Navarra P, Cirocchi R, Sharp A, Stigliano E, Feinstein DL, Dello Russo C: Rapamycin reduces clinical signs and neuropathic pain in a chronic model of experimental autoimmune encephalomyelitis. J Neuroimmunol 2012, 243:43–51.CrossRefPubMed
19.
go back to reference Linker RA, Maurer M, Gaupp S, Martini R, Holtmann B, Giess R, Rieckmann P, Lassmann H, Toyka KV, Sendtner M, Gold R: CNTF is a major protective factor in demyelinating CNS disease: a neurotrophic cytokine as modulator in neuroinflammation. Nat Med 2002, 8:620–624.CrossRefPubMed Linker RA, Maurer M, Gaupp S, Martini R, Holtmann B, Giess R, Rieckmann P, Lassmann H, Toyka KV, Sendtner M, Gold R: CNTF is a major protective factor in demyelinating CNS disease: a neurotrophic cytokine as modulator in neuroinflammation. Nat Med 2002, 8:620–624.CrossRefPubMed
20.
go back to reference Stösser S, Agarwal N, Tappe-Theodor A, Yanagisawa M, Kuner R: Dissecting the functional significance of endothelin A receptors in peripheral nociceptors in vivo via conditional gene deletion. Pain 2010, 148:206–214.CrossRefPubMed Stösser S, Agarwal N, Tappe-Theodor A, Yanagisawa M, Kuner R: Dissecting the functional significance of endothelin A receptors in peripheral nociceptors in vivo via conditional gene deletion. Pain 2010, 148:206–214.CrossRefPubMed
21.
go back to reference Tappe-Theodor A, Constantin CE, Tegeder I, Lechner SG, Langeslag M, Lepcynzsky P, Wirotanseng RI, Kurejova M, Agarwal N, Nagy G, et al.: Galpha(q/11) signaling tonically modulates nociceptor function and contributes to activity-dependent sensitization. Pain 2012, 153:184–196.CrossRef Tappe-Theodor A, Constantin CE, Tegeder I, Lechner SG, Langeslag M, Lepcynzsky P, Wirotanseng RI, Kurejova M, Agarwal N, Nagy G, et al.: Galpha(q/11) signaling tonically modulates nociceptor function and contributes to activity-dependent sensitization. Pain 2012, 153:184–196.CrossRef
22.
go back to reference Jiao Y, Sun Z, Lee T, Fusco FR, Kimble TD, Meade CA, Cuthbertson S, Reiner A: A simple and sensitive antigen retrieval method for free-floating and slide-mounted tissue sections. J Neurosci Methods 1999, 93:149–162.CrossRefPubMed Jiao Y, Sun Z, Lee T, Fusco FR, Kimble TD, Meade CA, Cuthbertson S, Reiner A: A simple and sensitive antigen retrieval method for free-floating and slide-mounted tissue sections. J Neurosci Methods 1999, 93:149–162.CrossRefPubMed
23.
go back to reference Kim KK, Adelstein RS, Kawamoto S: Identification of neuronal nuclei (NeuN) as Fox-3, a new member of the Fox-1 gene family of splicing factors. J Biol Chem 2009, 284:31052–31061.CrossRefPubMedPubMedCentral Kim KK, Adelstein RS, Kawamoto S: Identification of neuronal nuclei (NeuN) as Fox-3, a new member of the Fox-1 gene family of splicing factors. J Biol Chem 2009, 284:31052–31061.CrossRefPubMedPubMedCentral
24.
go back to reference Portiansky EL, Barbeito CG, Gimeno EJ, Zuccolilli GO, Goya RG: Loss of NeuN immunoreactivity in rat spinal cord neurons during aging. Exp Neurol 2006, 202:519–521.CrossRefPubMed Portiansky EL, Barbeito CG, Gimeno EJ, Zuccolilli GO, Goya RG: Loss of NeuN immunoreactivity in rat spinal cord neurons during aging. Exp Neurol 2006, 202:519–521.CrossRefPubMed
25.
go back to reference Unal-Cevik I, Kilinc M, Gursoy-Ozdemir Y, Gurer G, Dalkara T: Loss of NeuN immunoreactivity after cerebral ischemia does not indicate neuronal cell loss: a cautionary note. Brain Res 2004, 1015:169–174.CrossRefPubMed Unal-Cevik I, Kilinc M, Gursoy-Ozdemir Y, Gurer G, Dalkara T: Loss of NeuN immunoreactivity after cerebral ischemia does not indicate neuronal cell loss: a cautionary note. Brain Res 2004, 1015:169–174.CrossRefPubMed
26.
go back to reference Streit WJ, Kreutzberg GW: Lectin binding by resting and reactive microglia. J Neurocytol 1987, 16:249–260.CrossRefPubMed Streit WJ, Kreutzberg GW: Lectin binding by resting and reactive microglia. J Neurocytol 1987, 16:249–260.CrossRefPubMed
27.
go back to reference O'Connor AB, Schwid SR, Herrmann DN, Markman JD, Dworkin RH: Pain associated with multiple sclerosis: systematic review and proposed classification. Pain 2008, 137:96–111.CrossRefPubMed O'Connor AB, Schwid SR, Herrmann DN, Markman JD, Dworkin RH: Pain associated with multiple sclerosis: systematic review and proposed classification. Pain 2008, 137:96–111.CrossRefPubMed
28.
go back to reference Pöllmann W, Feneberg W: Current management of pain associated with multiple sclerosis. CNS Drugs 2008, 22:291–324.CrossRefPubMed Pöllmann W, Feneberg W: Current management of pain associated with multiple sclerosis. CNS Drugs 2008, 22:291–324.CrossRefPubMed
29.
go back to reference Archibald CJ, McGrath PJ, Ritvo PG, Fisk JD, Bhan V, Maxner CE, Murray TJ: Pain prevalence, severity and impact in a clinic sample of multiple sclerosis patients. Pain 1994, 58:89–93.CrossRefPubMed Archibald CJ, McGrath PJ, Ritvo PG, Fisk JD, Bhan V, Maxner CE, Murray TJ: Pain prevalence, severity and impact in a clinic sample of multiple sclerosis patients. Pain 1994, 58:89–93.CrossRefPubMed
30.
go back to reference Kalia LV, O'Connor PW: Severity of chronic pain and its relationship to quality of life in multiple sclerosis. Mult Scler 2005, 11:322–327.CrossRefPubMed Kalia LV, O'Connor PW: Severity of chronic pain and its relationship to quality of life in multiple sclerosis. Mult Scler 2005, 11:322–327.CrossRefPubMed
31.
go back to reference Solaro C, Brichetto G, Amato MP, Cocco E, Colombo B, D'Aleo G, Gasperini C, Ghezzi A, Martinelli V, Milanese C, et al.: The prevalence of pain in multiple sclerosis: a multicenter cross-sectional study. Neurology 2004, 63:919–921.CrossRefPubMed Solaro C, Brichetto G, Amato MP, Cocco E, Colombo B, D'Aleo G, Gasperini C, Ghezzi A, Martinelli V, Milanese C, et al.: The prevalence of pain in multiple sclerosis: a multicenter cross-sectional study. Neurology 2004, 63:919–921.CrossRefPubMed
32.
go back to reference Ehde DM, Osborne TL, Hanley MA, Jensen MP, Kraft GH: The scope and nature of pain in persons with multiple sclerosis. Mult Scler 2006, 12:629–638.CrossRefPubMed Ehde DM, Osborne TL, Hanley MA, Jensen MP, Kraft GH: The scope and nature of pain in persons with multiple sclerosis. Mult Scler 2006, 12:629–638.CrossRefPubMed
33.
go back to reference Grasso MG, Clemenzi A, Tonini A, Pace L, Casillo P, Cuccaro A, Pompa A, Troisi E: Pain in multiple sclerosis: a clinical and instrumental approach. Mult Scler 2008, 14:506–513.CrossRefPubMed Grasso MG, Clemenzi A, Tonini A, Pace L, Casillo P, Cuccaro A, Pompa A, Troisi E: Pain in multiple sclerosis: a clinical and instrumental approach. Mult Scler 2008, 14:506–513.CrossRefPubMed
34.
go back to reference Hirsh AT, Turner AP, Ehde DM, Haselkorn JK: Prevalence and impact of pain in multiple sclerosis: physical and psychologic contributors. Arch Phys Med Rehabil 2009, 90:646–651.CrossRefPubMedPubMedCentral Hirsh AT, Turner AP, Ehde DM, Haselkorn JK: Prevalence and impact of pain in multiple sclerosis: physical and psychologic contributors. Arch Phys Med Rehabil 2009, 90:646–651.CrossRefPubMedPubMedCentral
35.
go back to reference Michalski D, Liebig S, Thomae E, Hinz A, Bergh FT: Pain in patients with multiple sclerosis: a complex assessment including quantitative and qualitative measurements provides for a disease-related biopsychosocial pain model. J Pain Res 2011, 4:219–225.CrossRefPubMedPubMedCentral Michalski D, Liebig S, Thomae E, Hinz A, Bergh FT: Pain in patients with multiple sclerosis: a complex assessment including quantitative and qualitative measurements provides for a disease-related biopsychosocial pain model. J Pain Res 2011, 4:219–225.CrossRefPubMedPubMedCentral
36.
go back to reference Moulin DE, Foley KM, Ebers GC: Pain syndromes in multiple sclerosis. Neurology 1988, 38:1830–1834.CrossRefPubMed Moulin DE, Foley KM, Ebers GC: Pain syndromes in multiple sclerosis. Neurology 1988, 38:1830–1834.CrossRefPubMed
37.
go back to reference Osborne TL, Jensen MP, Ehde DM, Hanley MA, Kraft G: Psychosocial factors associated with pain intensity, pain-related interference, and psychological functioning in persons with multiple sclerosis and pain. Pain 2007, 127:52–62.CrossRefPubMed Osborne TL, Jensen MP, Ehde DM, Hanley MA, Kraft G: Psychosocial factors associated with pain intensity, pain-related interference, and psychological functioning in persons with multiple sclerosis and pain. Pain 2007, 127:52–62.CrossRefPubMed
38.
go back to reference Garbay B, Heape AM, Sargueil F, Cassagne C: Myelin synthesis in the peripheral nervous system. Prog Neurobiol 2000, 61:267–304.CrossRefPubMed Garbay B, Heape AM, Sargueil F, Cassagne C: Myelin synthesis in the peripheral nervous system. Prog Neurobiol 2000, 61:267–304.CrossRefPubMed
39.
go back to reference 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
40.
go back to reference 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.CrossRefPubMed 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.CrossRefPubMed
41.
go back to reference D'Amelio FE, Smith ME, Eng LF: Sequence of tissue responses in the early stages of experimental allergic encephalomyelitis (EAE): immunohistochemical, light microscopic, and ultrastructural observations in the spinal cord. Glia 1990, 3:229–240.CrossRefPubMed D'Amelio FE, Smith ME, Eng LF: Sequence of tissue responses in the early stages of experimental allergic encephalomyelitis (EAE): immunohistochemical, light microscopic, and ultrastructural observations in the spinal cord. Glia 1990, 3:229–240.CrossRefPubMed
42.
go back to reference Gehrmann J, Gold R, Linington C, Lannes-Vieira J, Wekerle H, Kreutzberg GW: Microglial involvement in experimental autoimmune inflammation of the central and peripheral nervous system. Glia 1993, 7:50–59.CrossRefPubMed Gehrmann J, Gold R, Linington C, Lannes-Vieira J, Wekerle H, Kreutzberg GW: Microglial involvement in experimental autoimmune inflammation of the central and peripheral nervous system. Glia 1993, 7:50–59.CrossRefPubMed
43.
go back to reference Gray E, Thomas TL, Betmouni S, Scolding N, Love S: Elevated activity and microglial expression of myeloperoxidase in demyelinated cerebral cortex in multiple sclerosis. Brain Pathol 2008, 18:86–95.CrossRefPubMed Gray E, Thomas TL, Betmouni S, Scolding N, Love S: Elevated activity and microglial expression of myeloperoxidase in demyelinated cerebral cortex in multiple sclerosis. Brain Pathol 2008, 18:86–95.CrossRefPubMed
44.
go back to reference Petzold A, Eikelenboom MJ, Gveric D, Keir G, Chapman M, Lazeron RH, Cuzner ML, Polman CH, Uitdehaag BM, Thompson EJ, Giovannoni G: Markers for different glial cell responses in multiple sclerosis: clinical and pathological correlations. Brain 2002, 125:1462–1473.CrossRefPubMed Petzold A, Eikelenboom MJ, Gveric D, Keir G, Chapman M, Lazeron RH, Cuzner ML, Polman CH, Uitdehaag BM, Thompson EJ, Giovannoni G: Markers for different glial cell responses in multiple sclerosis: clinical and pathological correlations. Brain 2002, 125:1462–1473.CrossRefPubMed
45.
go back to reference Benveniste EN: Role of macrophages/microglia in multiple sclerosis and experimental allergic encephalomyelitis. J Mol Med (Berl) 1997, 75:165–173.CrossRef Benveniste EN: Role of macrophages/microglia in multiple sclerosis and experimental allergic encephalomyelitis. J Mol Med (Berl) 1997, 75:165–173.CrossRef
46.
go back to reference Gonzalez-Scarano F, Baltuch G: Microglia as mediators of inflammatory and degenerative diseases. Annu Rev Neurosci 1999, 22:219–240.CrossRefPubMed Gonzalez-Scarano F, Baltuch G: Microglia as mediators of inflammatory and degenerative diseases. Annu Rev Neurosci 1999, 22:219–240.CrossRefPubMed
47.
48.
go back to reference Milligan ED, O'Connor KA, Nguyen KT, Armstrong CB, Twining C, Gaykema RP, Holguin A, Martin D, Maier SF, Watkins LR: Intrathecal HIV-1 envelope glycoprotein gp120 induces enhanced pain states mediated by spinal cord proinflammatory cytokines. J Neurosci 2001, 21:2808–2819.PubMed Milligan ED, O'Connor KA, Nguyen KT, Armstrong CB, Twining C, Gaykema RP, Holguin A, Martin D, Maier SF, Watkins LR: Intrathecal HIV-1 envelope glycoprotein gp120 induces enhanced pain states mediated by spinal cord proinflammatory cytokines. J Neurosci 2001, 21:2808–2819.PubMed
49.
go back to reference Ozenci V, Kouwenhoven M, Link H: Cytokines in multiple sclerosis: methodological aspects and pathogenic implications. Mult Scler 2002, 8:396–404.CrossRefPubMed Ozenci V, Kouwenhoven M, Link H: Cytokines in multiple sclerosis: methodological aspects and pathogenic implications. Mult Scler 2002, 8:396–404.CrossRefPubMed
50.
go back to reference Szczucinski A, Losy J: Chemokines and chemokine receptors in multiple sclerosis. Potential targets for new therapies. Acta Neurol Scand 2007, 115:137–146.CrossRefPubMed Szczucinski A, Losy J: Chemokines and chemokine receptors in multiple sclerosis. Potential targets for new therapies. Acta Neurol Scand 2007, 115:137–146.CrossRefPubMed
52.
go back to reference 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, 23:1026–1040.PubMed 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, 23:1026–1040.PubMed
54.
go back to reference Watkins LR, Maier SF: Beyond neurons: evidence that immune and glial cells contribute to pathological pain states. Physiol Rev 2002, 82:981–1011.CrossRefPubMed Watkins LR, Maier SF: Beyond neurons: evidence that immune and glial cells contribute to pathological pain states. Physiol Rev 2002, 82:981–1011.CrossRefPubMed
55.
go back to reference Watkins LR, Milligan ED, Maier SF: Glial proinflammatory cytokines mediate exaggerated pain states: implications for clinical pain. Adv Exp Med Biol 2003, 521:1–21.PubMed Watkins LR, Milligan ED, Maier SF: Glial proinflammatory cytokines mediate exaggerated pain states: implications for clinical pain. Adv Exp Med Biol 2003, 521:1–21.PubMed
56.
go back to reference McManus C, Berman JW, Brett FM, Staunton H, Farrell M, Brosnan CF: MCP-1, MCP-2 and MCP-3 expression in multiple sclerosis lesions: an immunohistochemical and in situ hybridization study. J Neuroimmunol 1998, 86:20–29.CrossRefPubMed McManus C, Berman JW, Brett FM, Staunton H, Farrell M, Brosnan CF: MCP-1, MCP-2 and MCP-3 expression in multiple sclerosis lesions: an immunohistochemical and in situ hybridization study. J Neuroimmunol 1998, 86:20–29.CrossRefPubMed
57.
go back to reference Simpson JE, Newcombe J, Cuzner ML, Woodroofe MN: Expression of monocyte chemoattractant protein-1 and other beta-chemokines by resident glia and inflammatory cells in multiple sclerosis lesions. J Neuroimmunol 1998, 84:238–249.CrossRefPubMed Simpson JE, Newcombe J, Cuzner ML, Woodroofe MN: Expression of monocyte chemoattractant protein-1 and other beta-chemokines by resident glia and inflammatory cells in multiple sclerosis lesions. J Neuroimmunol 1998, 84:238–249.CrossRefPubMed
58.
go back to reference Van Der Voorn P, Tekstra J, Beelen RH, Tensen CP, Van Der Valk P, De Groot CJ: Expression of MCP-1 by reactive astrocytes in demyelinating multiple sclerosis lesions. Am J Pathol 1999, 154:45–51.CrossRefPubMedPubMedCentral Van Der Voorn P, Tekstra J, Beelen RH, Tensen CP, Van Der Valk P, De Groot CJ: Expression of MCP-1 by reactive astrocytes in demyelinating multiple sclerosis lesions. Am J Pathol 1999, 154:45–51.CrossRefPubMedPubMedCentral
59.
go back to reference Fischer FR, Santambrogio L, Luo Y, Berman MA, Hancock WW, Dorf ME: Modulation of experimental autoimmune encephalomyelitis: effect of altered peptide ligand on chemokine and chemokine receptor expression. J Neuroimmunol 2000, 110:195–208.CrossRefPubMed Fischer FR, Santambrogio L, Luo Y, Berman MA, Hancock WW, Dorf ME: Modulation of experimental autoimmune encephalomyelitis: effect of altered peptide ligand on chemokine and chemokine receptor expression. J Neuroimmunol 2000, 110:195–208.CrossRefPubMed
60.
go back to reference Fife BT, Huffnagle GB, Kuziel WA, Karpus WJ: CC chemokine receptor 2 is critical for induction of experimental autoimmune encephalomyelitis. J Exp Med 2000, 192:899–905.CrossRefPubMedPubMedCentral Fife BT, Huffnagle GB, Kuziel WA, Karpus WJ: CC chemokine receptor 2 is critical for induction of experimental autoimmune encephalomyelitis. J Exp Med 2000, 192:899–905.CrossRefPubMedPubMedCentral
61.
go back to reference 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 U S A 2003, 100:7947–7952.CrossRefPubMedPubMedCentral 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 U S A 2003, 100:7947–7952.CrossRefPubMedPubMedCentral
62.
go back to reference Dansereau MA, Gosselin RD, Pohl M, Pommier B, Mechighel P, Mauborgne A, Rostene W, Kitabgi P, Beaudet N, Sarret P, Melik-Parsadaniantz S: Spinal CCL2 pronociceptive action is no longer effective in CCR2 receptor antagonist-treated rats. J Neurochem 2008, 106:757–769.CrossRefPubMed Dansereau MA, Gosselin RD, Pohl M, Pommier B, Mechighel P, Mauborgne A, Rostene W, Kitabgi P, Beaudet N, Sarret P, Melik-Parsadaniantz S: Spinal CCL2 pronociceptive action is no longer effective in CCR2 receptor antagonist-treated rats. J Neurochem 2008, 106:757–769.CrossRefPubMed
63.
go back to reference 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, 149:706–714.CrossRefPubMed 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, 149:706–714.CrossRefPubMed
64.
go back to reference 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, 48:463–469.CrossRefPubMed 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, 48:463–469.CrossRefPubMed
65.
go back to reference Gao YJ, Zhang L, Samad OA, Suter MR, Yasuhiko K, Xu ZZ, Park JY, Lind AL, Ma Q, Ji RR: JNK-induced MCP-1 production in spinal cord astrocytes contributes to central sensitization and neuropathic pain. J Neurosci 2009, 29:4096–4108.CrossRefPubMedPubMedCentral Gao YJ, Zhang L, Samad OA, Suter MR, Yasuhiko K, Xu ZZ, Park JY, Lind AL, Ma Q, Ji RR: JNK-induced MCP-1 production in spinal cord astrocytes contributes to central sensitization and neuropathic pain. J Neurosci 2009, 29:4096–4108.CrossRefPubMedPubMedCentral
66.
go back to reference Thacker MA, Clark AK, Bishop T, Grist J, Yip PK, Moon LD, Thompson SW, Marchand F, McMahon SB: CCL2 is a key mediator of microglia activation in neuropathic pain states. Eur J Pain 2009, 13:263–272.CrossRefPubMed Thacker MA, Clark AK, Bishop T, Grist J, Yip PK, Moon LD, Thompson SW, Marchand F, McMahon SB: CCL2 is a key mediator of microglia activation in neuropathic pain states. Eur J Pain 2009, 13:263–272.CrossRefPubMed
67.
go back to reference Shin BA, Yoo HG, Kim HS, Kim MH, Hwang YS, Chay KO, Lee KY, Ahn BW, Jung YD: P38 MAPK pathway is involved in the urokinase plasminogen activator expression in human gastric SNU-638 cells. Oncol Rep 2003, 10:1467–1471.PubMed Shin BA, Yoo HG, Kim HS, Kim MH, Hwang YS, Chay KO, Lee KY, Ahn BW, Jung YD: P38 MAPK pathway is involved in the urokinase plasminogen activator expression in human gastric SNU-638 cells. Oncol Rep 2003, 10:1467–1471.PubMed
68.
go back to reference Ji RR, Baba H, Brenner GJ, Woolf CJ: Nociceptive-specific activation of ERK in spinal neurons contributes to pain hypersensitivity. Nat Neurosci 1999, 2:1114–1119.CrossRefPubMed Ji RR, Baba H, Brenner GJ, Woolf CJ: Nociceptive-specific activation of ERK in spinal neurons contributes to pain hypersensitivity. Nat Neurosci 1999, 2:1114–1119.CrossRefPubMed
69.
go back to reference Ma W, Quirion R: Partial sciatic nerve ligation induces increase in the phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in astrocytes in the lumbar spinal dorsal horn and the gracile nucleus. Pain 2002, 99:175–184.CrossRefPubMed Ma W, Quirion R: Partial sciatic nerve ligation induces increase in the phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in astrocytes in the lumbar spinal dorsal horn and the gracile nucleus. Pain 2002, 99:175–184.CrossRefPubMed
70.
go back to reference Zhuang ZY, Wen YR, Zhang DR, Borsello T, Bonny C, Strichartz GR, Decosterd I, Ji RR: A peptide c-Jun N-terminal kinase (JNK) inhibitor blocks mechanical allodynia after spinal nerve ligation: respective roles of JNK activation in primary sensory neurons and spinal astrocytes for neuropathic pain development and maintenance. J Neurosci 2006, 26:3551–3560.CrossRefPubMed Zhuang ZY, Wen YR, Zhang DR, Borsello T, Bonny C, Strichartz GR, Decosterd I, Ji RR: A peptide c-Jun N-terminal kinase (JNK) inhibitor blocks mechanical allodynia after spinal nerve ligation: respective roles of JNK activation in primary sensory neurons and spinal astrocytes for neuropathic pain development and maintenance. J Neurosci 2006, 26:3551–3560.CrossRefPubMed
71.
go back to reference Gosselin RD, Varela C, Banisadr G, Mechighel P, Rostene W, Kitabgi P, Melik-Parsadaniantz S: Constitutive expression of CCR2 chemokine receptor and inhibition by MCP-1/CCL2 of GABA-induced currents in spinal cord neurones. J Neurochem 2005, 95:1023–1034.CrossRefPubMed Gosselin RD, Varela C, Banisadr G, Mechighel P, Rostene W, Kitabgi P, Melik-Parsadaniantz S: Constitutive expression of CCR2 chemokine receptor and inhibition by MCP-1/CCL2 of GABA-induced currents in spinal cord neurones. J Neurochem 2005, 95:1023–1034.CrossRefPubMed
73.
go back to reference Yong VW, Power C, Forsyth P, Edwards DR: Metalloproteinases in biology and pathology of the nervous system. Nat Rev Neurosci 2001, 2:502–511.CrossRefPubMed Yong VW, Power C, Forsyth P, Edwards DR: Metalloproteinases in biology and pathology of the nervous system. Nat Rev Neurosci 2001, 2:502–511.CrossRefPubMed
74.
go back to reference Kieseier BC, Clements JM, Pischel HB, Wells GM, Miller K, Gearing AJ, Hartung HP: Matrix metalloproteinases MMP-9 and MMP-7 are expressed in experimental autoimmune neuritis and the Guillain-Barre syndrome. Ann Neurol 1998, 43:427–434.CrossRefPubMed Kieseier BC, Clements JM, Pischel HB, Wells GM, Miller K, Gearing AJ, Hartung HP: Matrix metalloproteinases MMP-9 and MMP-7 are expressed in experimental autoimmune neuritis and the Guillain-Barre syndrome. Ann Neurol 1998, 43:427–434.CrossRefPubMed
75.
go back to reference Nygardas PT, Hinkkanen AE: Up-regulation of MMP-8 and MMP-9 activity in the BALB/c mouse spinal cord correlates with the severity of experimental autoimmune encephalomyelitis. Clin Exp Immunol 2002, 128:245–254.CrossRefPubMedPubMedCentral Nygardas PT, Hinkkanen AE: Up-regulation of MMP-8 and MMP-9 activity in the BALB/c mouse spinal cord correlates with the severity of experimental autoimmune encephalomyelitis. Clin Exp Immunol 2002, 128:245–254.CrossRefPubMedPubMedCentral
76.
go back to reference Toft-Hansen H, Nuttall RK, Edwards DR, Owens T: Key metalloproteinases are expressed by specific cell types in experimental autoimmune encephalomyelitis. J Immunol 2004, 173:5209–5218.CrossRefPubMed Toft-Hansen H, Nuttall RK, Edwards DR, Owens T: Key metalloproteinases are expressed by specific cell types in experimental autoimmune encephalomyelitis. J Immunol 2004, 173:5209–5218.CrossRefPubMed
77.
go back to reference Dong M, Liu R, Guo L, Li C, Tan G: Pathological findings in rats with experimental allergic encephalomyelitis. Apmis 2008, 116:972–984.CrossRefPubMed Dong M, Liu R, Guo L, Li C, Tan G: Pathological findings in rats with experimental allergic encephalomyelitis. Apmis 2008, 116:972–984.CrossRefPubMed
79.
go back to reference Kawasaki Y, Xu ZZ, Wang X, Park JY, Zhuang ZY, Tan PH, Gao YJ, Roy K, Corfas G, Lo EH, Ji RR: Distinct roles of matrix metalloproteases in the early- and late-phase development of neuropathic pain. Nat Med 2008, 14:331–336.CrossRefPubMedPubMedCentral Kawasaki Y, Xu ZZ, Wang X, Park JY, Zhuang ZY, Tan PH, Gao YJ, Roy K, Corfas G, Lo EH, Ji RR: Distinct roles of matrix metalloproteases in the early- and late-phase development of neuropathic pain. Nat Med 2008, 14:331–336.CrossRefPubMedPubMedCentral
80.
go back to reference Gijbels K, Masure S, Carton H, Opdenakker G: Gelatinase in the cerebrospinal fluid of patients with multiple sclerosis and other inflammatory neurological disorders. J Neuroimmunol 1992, 41:29–34.CrossRefPubMed Gijbels K, Masure S, Carton H, Opdenakker G: Gelatinase in the cerebrospinal fluid of patients with multiple sclerosis and other inflammatory neurological disorders. J Neuroimmunol 1992, 41:29–34.CrossRefPubMed
81.
go back to reference Lee MA, Palace J, Stabler G, Ford J, Gearing A, Miller K: Serum gelatinase B, TIMP-1 and TIMP-2 levels in multiple sclerosis. A longitudinal clinical and MRI study. Brain 1999,122(Pt 2):191–197.PubMed Lee MA, Palace J, Stabler G, Ford J, Gearing A, Miller K: Serum gelatinase B, TIMP-1 and TIMP-2 levels in multiple sclerosis. A longitudinal clinical and MRI study. Brain 1999,122(Pt 2):191–197.PubMed
82.
go back to reference Leppert D, Ford J, Stabler G, Grygar C, Lienert C, Huber S, Miller KM, Hauser SL, Kappos L: Matrix metalloproteinase-9 (gelatinase B) is selectively elevated in CSF during relapses and stable phases of multiple sclerosis. Brain 1998,121(Pt 12):2327–2334.CrossRefPubMed Leppert D, Ford J, Stabler G, Grygar C, Lienert C, Huber S, Miller KM, Hauser SL, Kappos L: Matrix metalloproteinase-9 (gelatinase B) is selectively elevated in CSF during relapses and stable phases of multiple sclerosis. Brain 1998,121(Pt 12):2327–2334.CrossRefPubMed
83.
go back to reference Lichtinghagen R, Seifert T, Kracke A, Marckmann S, Wurster U, Heidenreich F: Expression of matrix metalloproteinase-9 and its inhibitors in mononuclear blood cells of patients with multiple sclerosis. J Neuroimmunol 1999, 99:19–26.CrossRefPubMed Lichtinghagen R, Seifert T, Kracke A, Marckmann S, Wurster U, Heidenreich F: Expression of matrix metalloproteinase-9 and its inhibitors in mononuclear blood cells of patients with multiple sclerosis. J Neuroimmunol 1999, 99:19–26.CrossRefPubMed
84.
go back to reference Brundula V, Rewcastle NB, Metz LM, Bernard CC, Yong VW: Targeting leukocyte MMPs and transmigration: minocycline as a potential therapy for multiple sclerosis. Brain 2002, 125:1297–1308.CrossRefPubMed Brundula V, Rewcastle NB, Metz LM, Bernard CC, Yong VW: Targeting leukocyte MMPs and transmigration: minocycline as a potential therapy for multiple sclerosis. Brain 2002, 125:1297–1308.CrossRefPubMed
85.
go back to reference Folgueras AR, Fueyo A, Garcia-Suarez O, Cox J, Astudillo A, Tortorella P, Campestre C, Gutierrez-Fernandez A, Fanjul-Fernandez M, Pennington CJ, et al.: Collagenase-2 deficiency or inhibition impairs experimental autoimmune encephalomyelitis in mice. J Biol Chem 2008, 283:9465–9474.CrossRefPubMed Folgueras AR, Fueyo A, Garcia-Suarez O, Cox J, Astudillo A, Tortorella P, Campestre C, Gutierrez-Fernandez A, Fanjul-Fernandez M, Pennington CJ, et al.: Collagenase-2 deficiency or inhibition impairs experimental autoimmune encephalomyelitis in mice. J Biol Chem 2008, 283:9465–9474.CrossRefPubMed
86.
go back to reference Giuliani F, Metz LM, Wilson T, Fan Y, Bar-Or A, Yong VW: Additive effect of the combination of glatiramer acetate and minocycline in a model of MS. J Neuroimmunol 2005, 158:213–221.CrossRefPubMed Giuliani F, Metz LM, Wilson T, Fan Y, Bar-Or A, Yong VW: Additive effect of the combination of glatiramer acetate and minocycline in a model of MS. J Neuroimmunol 2005, 158:213–221.CrossRefPubMed
87.
go back to reference Opdenakker G, Nelissen I, Van Damme J: Functional roles and therapeutic targeting of gelatinase B and chemokines in multiple sclerosis. Lancet Neurol 2003, 2:747–756.CrossRefPubMed Opdenakker G, Nelissen I, Van Damme J: Functional roles and therapeutic targeting of gelatinase B and chemokines in multiple sclerosis. Lancet Neurol 2003, 2:747–756.CrossRefPubMed
88.
go back to reference Sommer C, Kress M: Recent findings on how proinflammatory cytokines cause pain: peripheral mechanisms in inflammatory and neuropathic hyperalgesia. Neurosci Lett 2004, 361:184–187.CrossRefPubMed Sommer C, Kress M: Recent findings on how proinflammatory cytokines cause pain: peripheral mechanisms in inflammatory and neuropathic hyperalgesia. Neurosci Lett 2004, 361:184–187.CrossRefPubMed
89.
go back to reference Sorkin LS, Xiao WH, Wagner R, Myers RR: Tumour necrosis factor-alpha induces ectopic activity in nociceptive primary afferent fibres. Neuroscience 1997, 81:255–262.CrossRefPubMed Sorkin LS, Xiao WH, Wagner R, Myers RR: Tumour necrosis factor-alpha induces ectopic activity in nociceptive primary afferent fibres. Neuroscience 1997, 81:255–262.CrossRefPubMed
90.
go back to reference Woolf CJ, Allchorne A, Safieh-Garabedian B, Poole S: Cytokines, nerve growth factor and inflammatory hyperalgesia: the contribution of tumour necrosis factor alpha. Br J Pharmacol 1997, 121:417–424.CrossRefPubMedPubMedCentral Woolf CJ, Allchorne A, Safieh-Garabedian B, Poole S: Cytokines, nerve growth factor and inflammatory hyperalgesia: the contribution of tumour necrosis factor alpha. Br J Pharmacol 1997, 121:417–424.CrossRefPubMedPubMedCentral
91.
go back to reference Kim H, Moon C, Ahn M, Lee Y, Kim S, Matsumoto Y, Koh CS, Kim MD, Shin T: Increased phosphorylation of cyclic AMP response element-binding protein in the spinal cord of Lewis rats with experimental autoimmune encephalomyelitis. Brain Res 2007, 1162:113–120.CrossRefPubMed Kim H, Moon C, Ahn M, Lee Y, Kim S, Matsumoto Y, Koh CS, Kim MD, Shin T: Increased phosphorylation of cyclic AMP response element-binding protein in the spinal cord of Lewis rats with experimental autoimmune encephalomyelitis. Brain Res 2007, 1162:113–120.CrossRefPubMed
92.
go back to reference Misawa S, Kuwabara S, Mori M, Hayakawa S, Sawai S, Hattori T: Peripheral nerve demyelination in multiple sclerosis. Clin Neurophysiol 2008, 119:1829–1833.CrossRefPubMed Misawa S, Kuwabara S, Mori M, Hayakawa S, Sawai S, Hattori T: Peripheral nerve demyelination in multiple sclerosis. Clin Neurophysiol 2008, 119:1829–1833.CrossRefPubMed
93.
go back to reference Sarova-Pinhas I, Achiron A, Gilad R, Lampl Y: Peripheral neuropathy in multiple sclerosis: a clinical and electrophysiologic study. Acta Neurol Scand 1995, 91:234–238.CrossRefPubMed Sarova-Pinhas I, Achiron A, Gilad R, Lampl Y: Peripheral neuropathy in multiple sclerosis: a clinical and electrophysiologic study. Acta Neurol Scand 1995, 91:234–238.CrossRefPubMed
94.
go back to reference Pender MP, Sears TA: Involvement of the dorsal root ganglion in acute experimental allergic encephalomyelitis in the Lewis rat. A histological and electrophysiological study. J Neurol Sci 1986, 72:231–242.CrossRefPubMed Pender MP, Sears TA: Involvement of the dorsal root ganglion in acute experimental allergic encephalomyelitis in the Lewis rat. A histological and electrophysiological study. J Neurol Sci 1986, 72:231–242.CrossRefPubMed
95.
go back to reference Pender MP, Sears TA: Vulnerability of the dorsal root ganglion in experimental allergic encephalomyelitis. Clin Exp Neurol 1985, 21:211–223.PubMed Pender MP, Sears TA: Vulnerability of the dorsal root ganglion in experimental allergic encephalomyelitis. Clin Exp Neurol 1985, 21:211–223.PubMed
96.
go back to reference Pender MP, Sears TA: The pathophysiology of acute experimental allergic encephalomyelitis in the rabbit. Brain 1984,107(Pt 3):699–726.CrossRefPubMed Pender MP, Sears TA: The pathophysiology of acute experimental allergic encephalomyelitis in the rabbit. Brain 1984,107(Pt 3):699–726.CrossRefPubMed
Metadata
Title
Pain in experimental autoimmune encephalitis: a comparative study between different mouse models
Authors
Jianning Lu
Martina Kurejova
Laura N Wirotanseng
Ralf A Linker
Rohini Kuner
Anke Tappe-Theodor
Publication date
01-12-2012
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2012
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/1742-2094-9-233

Other articles of this Issue 1/2012

Journal of Neuroinflammation 1/2012 Go to the issue