Skip to main content
Top
Published in: Arthritis Research & Therapy 5/2014

Open Access 01-10-2014 | Review

Nociceptive neurons detect cytokines in arthritis

Author: Hans-Georg Schaible

Published in: Arthritis Research & Therapy | Issue 5/2014

Login to get access

Abstract

Proinflammatory cytokines are major mediators in the pathogenesis of diseases of joints such as rheumatoid arthritis and osteoarthritis. This review emphasizes that proinflammatory cytokines such as tumor necrosis factor-alpha, interleukin-1beta, interleukin-6 and interleukin-17 are also mediators of pain by directly acting on the nociceptive system. Proportions of nociceptive sensory neurons express receptors for these cytokines, and the application of cytokines rapidly changes the excitability, ion currents and second messenger systems of these neurons. By inducing persistent sensitization of nociceptive sensory neurons (C- and a proportion of Aδ-fibers) for mechanical stimuli in the joint (a process called peripheral sensitization), these cytokines significantly contribute to the persistent hyperalgesia typical for many disease states of the joint. In addition, the disease-associated release of cytokines in the spinal cord supports the generation of central sensitization. The therapeutic neutralization of proinflammatory cytokines thus not only reduces the process of inflammation but may directly reduce hyperalgesia and pain by reversing the neuronal effects of cytokines. It is emerging that different cytokines have different actions on neurons. The neutralization of tumor necrosis factor-alpha reduces both mechanical and thermal hyperalgesia of the joint. The neutralization of interleukin-1beta attenuates thermal hyperalgesia whereas the neutralization of interleukin-6 and interleukin-17 mainly reduces mechanical hyperalgesia. These different effects are partly explained by influencing different target molecules in sensory neurons. For example, in cultured sensory neurons tumor necrosis factor-alpha and interleukin-1beta upregulate the TRPV1 ion channel, which is involved in the transduction of heat stimuli, consistent with an effect of these cytokines in thermal hyperalgesia. By contrast, interleukin-17 upregulates the TRPV4 ion channel, which has a role in the transduction of mechanical stimuli. Thus, the analgesic potential of neutralizing cytokines seems to depend on which cytokine is mainly involved in the particular pain state.
Appendix
Available only for authorised users
Literature
1.
go back to reference McInnes IB, Schett G: The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011, 365: 2205-2219. 10.1056/NEJMra1004965.CrossRefPubMed McInnes IB, Schett G: The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011, 365: 2205-2219. 10.1056/NEJMra1004965.CrossRefPubMed
2.
go back to reference Bondeson J, Blom AB, Wainwright S, Hughes C, Caterson B, van den Berg WB: The role of synovial macrophages and macrophage-produced mediators in driving inflammatory and destructive responses in osteoarthritis. Arthritis Rheum. 2010, 62: 647-657. 10.1002/art.27290.CrossRefPubMed Bondeson J, Blom AB, Wainwright S, Hughes C, Caterson B, van den Berg WB: The role of synovial macrophages and macrophage-produced mediators in driving inflammatory and destructive responses in osteoarthritis. Arthritis Rheum. 2010, 62: 647-657. 10.1002/art.27290.CrossRefPubMed
3.
go back to reference Boettger MK, Hensellek S, Richter F, Gajda M, Stöckigt R, Segond von Banchet G, Bräuer R, Schaible HG: Antinociceptive effects of tumor necrosis factor alpha neutralization in a rat model of antigen-induced arthritis: evidence of a neuronal target. Arthritis Rheum. 2008, 58: 2368-2378. 10.1002/art.23608.CrossRefPubMed Boettger MK, Hensellek S, Richter F, Gajda M, Stöckigt R, Segond von Banchet G, Bräuer R, Schaible HG: Antinociceptive effects of tumor necrosis factor alpha neutralization in a rat model of antigen-induced arthritis: evidence of a neuronal target. Arthritis Rheum. 2008, 58: 2368-2378. 10.1002/art.23608.CrossRefPubMed
4.
go back to reference Inglis JJ, Nissim A, Less DM, Hunt S, Chernajovyky Y, Kidd BL: The differential contribution of tumor necrosis factor to thermal and mechanical hyperalgesia during chronic inflammation. Arthritis Res Ther. 2005, 7: R807-10.1186/ar1743.CrossRefPubMedCentralPubMed Inglis JJ, Nissim A, Less DM, Hunt S, Chernajovyky Y, Kidd BL: The differential contribution of tumor necrosis factor to thermal and mechanical hyperalgesia during chronic inflammation. Arthritis Res Ther. 2005, 7: R807-10.1186/ar1743.CrossRefPubMedCentralPubMed
5.
go back to reference Hess A, Axmann R, Rech J, Finzel S, Heindl C, Kreitz S, Sergeeva M, Saake M, Garcia M, Kollias G, Straub RH, Sporns O, Doerfler A, Brune K, Schett G: Blockade of TNF-α rapidly inhibits pain responses in the central nervous system. Proc Natl Acad Sci U S A. 2011, 108: 3731-3736. 10.1073/pnas.1011774108.CrossRefPubMedCentralPubMed Hess A, Axmann R, Rech J, Finzel S, Heindl C, Kreitz S, Sergeeva M, Saake M, Garcia M, Kollias G, Straub RH, Sporns O, Doerfler A, Brune K, Schett G: Blockade of TNF-α rapidly inhibits pain responses in the central nervous system. Proc Natl Acad Sci U S A. 2011, 108: 3731-3736. 10.1073/pnas.1011774108.CrossRefPubMedCentralPubMed
6.
go back to reference DeLeo JA, Colburn RW, Nichols M, Malhotra A: Interleukin-6-mediated hyperalgesia/allodynia and increased spinal IL-6 expression in a rat mononeuropathy model. J Interferon Cytokine Res. 1996, 16: 695-700. 10.1089/jir.1996.16.695.CrossRefPubMed DeLeo JA, Colburn RW, Nichols M, Malhotra A: Interleukin-6-mediated hyperalgesia/allodynia and increased spinal IL-6 expression in a rat mononeuropathy model. J Interferon Cytokine Res. 1996, 16: 695-700. 10.1089/jir.1996.16.695.CrossRefPubMed
7.
go back to reference Xu XJ, Hao JX, Andell-Jonsson S, Poli V, Bartfai T, Wiesenfeld-Hallin Z: Nociceptive responses in interleukin-6-deficient mice to peripheral inflammation and peripheral nerve section. Cytokine. 1997, 12: 1028-1033. 10.1006/cyto.1997.0243.CrossRef Xu XJ, Hao JX, Andell-Jonsson S, Poli V, Bartfai T, Wiesenfeld-Hallin Z: Nociceptive responses in interleukin-6-deficient mice to peripheral inflammation and peripheral nerve section. Cytokine. 1997, 12: 1028-1033. 10.1006/cyto.1997.0243.CrossRef
8.
go back to reference Sommer C, Schmidt C, George A: Hyperalgesia in experimental neuropathy is dependent on the TNF receptor 1. Exp Neurol. 1998, 151: 138-142. 10.1006/exnr.1998.6797.CrossRefPubMed Sommer C, Schmidt C, George A: Hyperalgesia in experimental neuropathy is dependent on the TNF receptor 1. Exp Neurol. 1998, 151: 138-142. 10.1006/exnr.1998.6797.CrossRefPubMed
10.
go back to reference Segond von Banchet G, Boettger MK, Fischer N, Gajda M, Bräuer R, Schaible HG: Experimental arthritis causes tumor necrosis factor-alpha-dependent infiltration of macrophages into rat dorsal root ganglia which correlates with pain-related behavior. Pain. 2009, 145: 151-159. 10.1016/j.pain.2009.06.002.CrossRefPubMed Segond von Banchet G, Boettger MK, Fischer N, Gajda M, Bräuer R, Schaible HG: Experimental arthritis causes tumor necrosis factor-alpha-dependent infiltration of macrophages into rat dorsal root ganglia which correlates with pain-related behavior. Pain. 2009, 145: 151-159. 10.1016/j.pain.2009.06.002.CrossRefPubMed
11.
go back to reference McMahon SB, Malcangio M: Current challenges in glia-pain biology. Neuron. 2009, 64: 46-54. 10.1016/j.neuron.2009.09.033.CrossRefPubMed McMahon SB, Malcangio M: Current challenges in glia-pain biology. Neuron. 2009, 64: 46-54. 10.1016/j.neuron.2009.09.033.CrossRefPubMed
12.
go back to reference Boyle DL, Jones TL, Hammaker D, Svensson CI, Rosengren S, Albani S, Sorkin L, Firestein GS: Regulation of peripheral inflammation by spinal p38 MAP kinase in rats. PLoS Med. 2006, 3: e338-10.1371/journal.pmed.0030338.CrossRefPubMedCentralPubMed Boyle DL, Jones TL, Hammaker D, Svensson CI, Rosengren S, Albani S, Sorkin L, Firestein GS: Regulation of peripheral inflammation by spinal p38 MAP kinase in rats. PLoS Med. 2006, 3: e338-10.1371/journal.pmed.0030338.CrossRefPubMedCentralPubMed
13.
go back to reference Boettger MK, Weber K, Grossmann D, Gajda M, Bauer R, Bär KJ, Schulz S, Voss A, Geis C, Bräuer R, Schaible H-G: Spinal TNF-alpha neutralization reduces peripheral inflammation and hyperalgesia and suppresses autonomic responses in experimental arthritis. Arthritis Rheum. 2010, 62: 1308-1318. 10.1002/art.27380.CrossRefPubMed Boettger MK, Weber K, Grossmann D, Gajda M, Bauer R, Bär KJ, Schulz S, Voss A, Geis C, Bräuer R, Schaible H-G: Spinal TNF-alpha neutralization reduces peripheral inflammation and hyperalgesia and suppresses autonomic responses in experimental arthritis. Arthritis Rheum. 2010, 62: 1308-1318. 10.1002/art.27380.CrossRefPubMed
14.
go back to reference Richter F, Natura G, Loeser S, Schmidt K, Viisanen H, Schaible H-G: Tumor necrosis factor (TNF) causes persistent sensitization of joint nociceptors for mechanical stimuli. Arthritis Rheum. 2010, 62: 3806-3814. 10.1002/art.27715.CrossRefPubMed Richter F, Natura G, Loeser S, Schmidt K, Viisanen H, Schaible H-G: Tumor necrosis factor (TNF) causes persistent sensitization of joint nociceptors for mechanical stimuli. Arthritis Rheum. 2010, 62: 3806-3814. 10.1002/art.27715.CrossRefPubMed
15.
go back to reference Cunha FQ, Poole S, Lorenzetti BB, Ferreira SH: The pivotal role of tumour necrosis factor α in the development of inflammatory hyperalgesia. Br J Pharmacol. 1992, 107: 660-664. 10.1111/j.1476-5381.1992.tb14503.x.CrossRefPubMedCentralPubMed Cunha FQ, Poole S, Lorenzetti BB, Ferreira SH: The pivotal role of tumour necrosis factor α in the development of inflammatory hyperalgesia. Br J Pharmacol. 1992, 107: 660-664. 10.1111/j.1476-5381.1992.tb14503.x.CrossRefPubMedCentralPubMed
16.
go back to reference Watkins LR, Wiertelak EP, Goehler LE, Smith KP, Martin D, Maier SF: Characterization of cytokine-induced hyperalgesia. Brain Res. 1994, 654: 15-26. 10.1016/0006-8993(94)91566-0.CrossRefPubMed Watkins LR, Wiertelak EP, Goehler LE, Smith KP, Martin D, Maier SF: Characterization of cytokine-induced hyperalgesia. Brain Res. 1994, 654: 15-26. 10.1016/0006-8993(94)91566-0.CrossRefPubMed
17.
go back to reference Jin X, Gereau RW: Acute p38-mediated modulation of tetrodotoxin-resistant sodium channels in mouse sensory neurons by tumor necrosis factor-α. J Neurosci. 2006, 26: 246-255. 10.1523/JNEUROSCI.3858-05.2006.CrossRefPubMed Jin X, Gereau RW: Acute p38-mediated modulation of tetrodotoxin-resistant sodium channels in mouse sensory neurons by tumor necrosis factor-α. J Neurosci. 2006, 26: 246-255. 10.1523/JNEUROSCI.3858-05.2006.CrossRefPubMed
18.
go back to reference Opree A, Kress M: Involvement of the proinflammatory cytokines tumor necrosis factor-alpha, IL-1 beta, and IL-6 but not IL-8 in the development of heat hyperalgesia: effects on heat-evoked calcitonin gene-related peptide release from rat skin. J Neurosci. 2000, 20: 6289-6293.PubMed Opree A, Kress M: Involvement of the proinflammatory cytokines tumor necrosis factor-alpha, IL-1 beta, and IL-6 but not IL-8 in the development of heat hyperalgesia: effects on heat-evoked calcitonin gene-related peptide release from rat skin. J Neurosci. 2000, 20: 6289-6293.PubMed
19.
go back to reference Hensellek S, Brell P, Schaible H-G, Bräuer R, Segond von Banchet G: The cytokine TNFα increases the proportion of DRG neurones expressing the TRPV1 receptor via the TNFR1 receptor and ERK activation. Mol Cell Neurosci. 2007, 36: 381-391. 10.1016/j.mcn.2007.07.010.CrossRefPubMed Hensellek S, Brell P, Schaible H-G, Bräuer R, Segond von Banchet G: The cytokine TNFα increases the proportion of DRG neurones expressing the TRPV1 receptor via the TNFR1 receptor and ERK activation. Mol Cell Neurosci. 2007, 36: 381-391. 10.1016/j.mcn.2007.07.010.CrossRefPubMed
20.
go back to reference Clark AK, Gentry C, Bradbury EJ, McMahon SC, Malcangio M: Role of spinal microglia in rat models of peripheral nerve injury and inflammation. Eur J Pain. 2007, 11: 223-230. 10.1016/j.ejpain.2006.02.003.CrossRefPubMed Clark AK, Gentry C, Bradbury EJ, McMahon SC, Malcangio M: Role of spinal microglia in rat models of peripheral nerve injury and inflammation. Eur J Pain. 2007, 11: 223-230. 10.1016/j.ejpain.2006.02.003.CrossRefPubMed
21.
go back to reference Christianson CA, Corr M, Firestein GS, Mobargha A, Yaksh TL, Svensson CI: Characterization of the acute and persistent pain state present in K/BxN serum transfer arthritis. Pain. 2010, 151: 394-403. 10.1016/j.pain.2010.07.030.CrossRefPubMedCentralPubMed Christianson CA, Corr M, Firestein GS, Mobargha A, Yaksh TL, Svensson CI: Characterization of the acute and persistent pain state present in K/BxN serum transfer arthritis. Pain. 2010, 151: 394-403. 10.1016/j.pain.2010.07.030.CrossRefPubMedCentralPubMed
22.
go back to reference Sagar DR, Burston JJ, Hatway GJ, Woodhams SG, Pearson RG, Bennett AJ: The contribution of spinal glial cells to chronic pain behaviour in the monosodium iodoacetate model of osteoarthritic pain. Mol Pain. 2011, 7: 88-100. 10.1186/1744-8069-7-88.CrossRefPubMedCentralPubMed Sagar DR, Burston JJ, Hatway GJ, Woodhams SG, Pearson RG, Bennett AJ: The contribution of spinal glial cells to chronic pain behaviour in the monosodium iodoacetate model of osteoarthritic pain. Mol Pain. 2011, 7: 88-100. 10.1186/1744-8069-7-88.CrossRefPubMedCentralPubMed
23.
go back to reference König C, Zharsky M, Möller C, Schaible H-G, Ebersberger A: Involvement of peripheral and spinal tumor necrosis factor α (TNFα) in spinal cord hyperexcitability during knee joint inflammation in rat. Arthritis Rheum. 2014, 66: 599-609. 10.1002/art.38271.CrossRef König C, Zharsky M, Möller C, Schaible H-G, Ebersberger A: Involvement of peripheral and spinal tumor necrosis factor α (TNFα) in spinal cord hyperexcitability during knee joint inflammation in rat. Arthritis Rheum. 2014, 66: 599-609. 10.1002/art.38271.CrossRef
24.
go back to reference Inglis JJ, Notley CA, Essex D, Wilson AW, Feldmann M, Anand P, Williams R: Collagen-induced arthritis as a model of hyperalgesia. Functional and cellular analysis of the analgesic actions of tumor necrosis factor blockade. Arthritis Rheum. 2007, 56: 4015-4023. 10.1002/art.23063.CrossRefPubMed Inglis JJ, Notley CA, Essex D, Wilson AW, Feldmann M, Anand P, Williams R: Collagen-induced arthritis as a model of hyperalgesia. Functional and cellular analysis of the analgesic actions of tumor necrosis factor blockade. Arthritis Rheum. 2007, 56: 4015-4023. 10.1002/art.23063.CrossRefPubMed
25.
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. 10.1016/j.neulet.2003.12.007.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. 10.1016/j.neulet.2003.12.007.CrossRefPubMed
26.
go back to reference Hakim AW, Dong X-D, Svensson P, Kumar U, Cairns BE: TNFα mechanically sensitizes masseter muscle afferent fibers of male rats. J Neurophysiol. 2009, 102: 1551-1559. 10.1152/jn.00326.2009.CrossRefPubMed Hakim AW, Dong X-D, Svensson P, Kumar U, Cairns BE: TNFα mechanically sensitizes masseter muscle afferent fibers of male rats. J Neurophysiol. 2009, 102: 1551-1559. 10.1152/jn.00326.2009.CrossRefPubMed
27.
go back to reference Fonseca JE, Santos MJ, Canhao H, Choy E: Interleukin-6 as a key player in systemic inflammation and joint destruction. Autoimmun Rev. 2009, 8: 538-542. 10.1016/j.autrev.2009.01.012.CrossRefPubMed Fonseca JE, Santos MJ, Canhao H, Choy E: Interleukin-6 as a key player in systemic inflammation and joint destruction. Autoimmun Rev. 2009, 8: 538-542. 10.1016/j.autrev.2009.01.012.CrossRefPubMed
28.
go back to reference Nowell MA, Richards PJ, Horiuchi S, Yamamoto N, Rose-John S, Topley N, Williams AS, Jones SA: Soluble IL-6 receptor governs IL-6 activity in experimental arthritis: blockade of arthritis severity by soluble glycoprotein 130. J Immunol. 2003, 171: 3202-3209. 10.4049/jimmunol.171.6.3202.CrossRefPubMed Nowell MA, Richards PJ, Horiuchi S, Yamamoto N, Rose-John S, Topley N, Williams AS, Jones SA: Soluble IL-6 receptor governs IL-6 activity in experimental arthritis: blockade of arthritis severity by soluble glycoprotein 130. J Immunol. 2003, 171: 3202-3209. 10.4049/jimmunol.171.6.3202.CrossRefPubMed
29.
go back to reference Axmann R, Bohm C, Kronke G, Zwerina J, Smolen J, Schett G: Inhibition of interleukin-6 receptor directly blocks osteoclast formation in vitro and in vivo. Arthritis Rheum. 2009, 60: 2747-2756. 10.1002/art.24781.CrossRefPubMed Axmann R, Bohm C, Kronke G, Zwerina J, Smolen J, Schett G: Inhibition of interleukin-6 receptor directly blocks osteoclast formation in vitro and in vivo. Arthritis Rheum. 2009, 60: 2747-2756. 10.1002/art.24781.CrossRefPubMed
30.
go back to reference Jones SA, Richards PJ, Scheller J, Rose-John S: IL-6 transsignaling: the in vivo consequences. J Interferon Cytokine Res. 2005, 25: 241-253. 10.1089/jir.2005.25.241.CrossRefPubMed Jones SA, Richards PJ, Scheller J, Rose-John S: IL-6 transsignaling: the in vivo consequences. J Interferon Cytokine Res. 2005, 25: 241-253. 10.1089/jir.2005.25.241.CrossRefPubMed
31.
go back to reference Boettger MK, Leuchtweis J, Kümmel D, Gajda M, Bräuer R, Schaible H-G: Differential effects of locally and systemically administered soluble glycoprotein 130 on pain and inflammation in experimental arthritis. Arthritis Res Ther. 2010, 12: R140-10.1186/ar3079.CrossRefPubMedCentralPubMed Boettger MK, Leuchtweis J, Kümmel D, Gajda M, Bräuer R, Schaible H-G: Differential effects of locally and systemically administered soluble glycoprotein 130 on pain and inflammation in experimental arthritis. Arthritis Res Ther. 2010, 12: R140-10.1186/ar3079.CrossRefPubMedCentralPubMed
32.
go back to reference Obreja O, Biasio W, Andratsch M, Lips KS, Rathee PK, Ludwig A, Rose-John S, Kress M: Fast modulation of heat-activated ionic current by proinflammatory interleukin 6 in rat sensory neurons. Brain. 2005, 128: 1634-1641. 10.1093/brain/awh490.CrossRefPubMed Obreja O, Biasio W, Andratsch M, Lips KS, Rathee PK, Ludwig A, Rose-John S, Kress M: Fast modulation of heat-activated ionic current by proinflammatory interleukin 6 in rat sensory neurons. Brain. 2005, 128: 1634-1641. 10.1093/brain/awh490.CrossRefPubMed
33.
go back to reference Segond von Banchet G, Kiehl M, Schaible HG: Acute and long-term effects of IL-6 on cultured dorsal root ganglion neurones from adult rat. J Neurochem. 2005, 94: 238-248. 10.1111/j.1471-4159.2005.03185.x.CrossRef Segond von Banchet G, Kiehl M, Schaible HG: Acute and long-term effects of IL-6 on cultured dorsal root ganglion neurones from adult rat. J Neurochem. 2005, 94: 238-248. 10.1111/j.1471-4159.2005.03185.x.CrossRef
34.
go back to reference Brenn D, Richter F, Schaible HG: Sensitization of unmyelinated sensory fibers of the joint nerve to mechanical stimuli by interleukin-6 in the rat: an inflammatory mechanism of joint pain. Arthritis Rheum. 2007, 56: 351-359. 10.1002/art.22282.CrossRefPubMed Brenn D, Richter F, Schaible HG: Sensitization of unmyelinated sensory fibers of the joint nerve to mechanical stimuli by interleukin-6 in the rat: an inflammatory mechanism of joint pain. Arthritis Rheum. 2007, 56: 351-359. 10.1002/art.22282.CrossRefPubMed
35.
go back to reference Dina OA, Green PG, Levine JD: Role of interleukin-6 in chronic muscle hyperalgesic priming. Neuroscience. 2008, 152: 521-525. 10.1016/j.neuroscience.2008.01.006.CrossRefPubMedCentralPubMed Dina OA, Green PG, Levine JD: Role of interleukin-6 in chronic muscle hyperalgesic priming. Neuroscience. 2008, 152: 521-525. 10.1016/j.neuroscience.2008.01.006.CrossRefPubMedCentralPubMed
36.
go back to reference Andratsch M, Mair N, Constantin CE, Scherbakov N, Benetti C, Quarta S, Vogl C, Sailer CA, Uceyler N, Brockhaus J, Martini R, Sommer C, Zeilhofer HU, Möller W, Kuner R, Davis JB, Rose-John S, Kress M: A key role for gp130 expressed on peripheral sensory nerves in pathological pain. J Neurosci. 2009, 29: 13473-13483. 10.1523/JNEUROSCI.1822-09.2009.CrossRefPubMed Andratsch M, Mair N, Constantin CE, Scherbakov N, Benetti C, Quarta S, Vogl C, Sailer CA, Uceyler N, Brockhaus J, Martini R, Sommer C, Zeilhofer HU, Möller W, Kuner R, Davis JB, Rose-John S, Kress M: A key role for gp130 expressed on peripheral sensory nerves in pathological pain. J Neurosci. 2009, 29: 13473-13483. 10.1523/JNEUROSCI.1822-09.2009.CrossRefPubMed
37.
go back to reference Quarta S, Vogl C, Constantin CE, Üçeyler N, Sommer C, Kress M: Genetic evidence for an essential role of neuronally expressed IL-6 signal transducer gp130 in the induction and maintenance of experimentally induced mechanical hypersensitivity in vivo and in vitro. Mol Pain. 2011, 7: 73-10.1186/1744-8069-7-73.CrossRefPubMedCentralPubMed Quarta S, Vogl C, Constantin CE, Üçeyler N, Sommer C, Kress M: Genetic evidence for an essential role of neuronally expressed IL-6 signal transducer gp130 in the induction and maintenance of experimentally induced mechanical hypersensitivity in vivo and in vitro. Mol Pain. 2011, 7: 73-10.1186/1744-8069-7-73.CrossRefPubMedCentralPubMed
38.
go back to reference Vazquez E, Kahlenbach J, Segond von Banchet G, König C, Schaible H-G, Ebersberger A: Spinal interleukin-6 is an amplifier of arthritic pain. Arthritis Rheum. 2012, 64: 2233-2242. 10.1002/art.34384.CrossRefPubMed Vazquez E, Kahlenbach J, Segond von Banchet G, König C, Schaible H-G, Ebersberger A: Spinal interleukin-6 is an amplifier of arthritic pain. Arthritis Rheum. 2012, 64: 2233-2242. 10.1002/art.34384.CrossRefPubMed
39.
go back to reference Ebbinghaus M, Uhlig B, Richter F, Segond von Banchet G, Gajda M, Bräuer R, Schaible H-G: The role of interleukin-1β in arthritic pain: main involvement in thermal but not in mechanical hyperalgesia in rat antigen-induced arthritis. Arthritis Rheum. 2012, 64: 3897-3907. 10.1002/art.34675.CrossRefPubMed Ebbinghaus M, Uhlig B, Richter F, Segond von Banchet G, Gajda M, Bräuer R, Schaible H-G: The role of interleukin-1β in arthritic pain: main involvement in thermal but not in mechanical hyperalgesia in rat antigen-induced arthritis. Arthritis Rheum. 2012, 64: 3897-3907. 10.1002/art.34675.CrossRefPubMed
40.
go back to reference Copray JC, Mantingh I, Brouwer N, Biber K, Kust BM, Liem RS, Huitinga I, Tilders FJ, Van Dam AM: Expression of interleukin-1beta in rat dorsal root ganglia. J Neuroimmunol. 2001, 118: 203-211. 10.1016/S0165-5728(01)00324-1.CrossRefPubMed Copray JC, Mantingh I, Brouwer N, Biber K, Kust BM, Liem RS, Huitinga I, Tilders FJ, Van Dam AM: Expression of interleukin-1beta in rat dorsal root ganglia. J Neuroimmunol. 2001, 118: 203-211. 10.1016/S0165-5728(01)00324-1.CrossRefPubMed
41.
go back to reference Obreja O, Rathee PK, Lips KS, Distler C, Kress M: IL-1 beta potentiates heat-activated currents in rat sensory neurons: involvement of IL-1RI, tyrosine kinase, and protein kinase C. FASEB J. 2002, 16: 497-503. 10.1096/fj.02-0101com.CrossRef Obreja O, Rathee PK, Lips KS, Distler C, Kress M: IL-1 beta potentiates heat-activated currents in rat sensory neurons: involvement of IL-1RI, tyrosine kinase, and protein kinase C. FASEB J. 2002, 16: 497-503. 10.1096/fj.02-0101com.CrossRef
42.
go back to reference Schäfers M, Sorkin L: Effect of cytokines on neuronal excitability. Neurosci Lett. 2008, 437: 188-193. 10.1016/j.neulet.2008.03.052.CrossRefPubMed Schäfers M, Sorkin L: Effect of cytokines on neuronal excitability. Neurosci Lett. 2008, 437: 188-193. 10.1016/j.neulet.2008.03.052.CrossRefPubMed
43.
go back to reference Fukuoka H, Kawatani M, Hisamitsu T, Takeshige C: Cutaneous hyperalgesia induced by peripheral injection of interleukin-1 beta in the rat. Brain Res. 1994, 657: 133-140. 10.1016/0006-8993(94)90960-1.CrossRefPubMed Fukuoka H, Kawatani M, Hisamitsu T, Takeshige C: Cutaneous hyperalgesia induced by peripheral injection of interleukin-1 beta in the rat. Brain Res. 1994, 657: 133-140. 10.1016/0006-8993(94)90960-1.CrossRefPubMed
44.
go back to reference Binshtok AM, Wang H, Zimmermann K, Amaya F, Vardeh D, Shi L, Brenner GJ, Ji RR, Bean BP, Woolf CJ, Samad TA: Nociceptors are interleukin-1 beta sensors. J Neurosci. 2008, 28: 14062-14073. 10.1523/JNEUROSCI.3795-08.2008.CrossRefPubMedCentralPubMed Binshtok AM, Wang H, Zimmermann K, Amaya F, Vardeh D, Shi L, Brenner GJ, Ji RR, Bean BP, Woolf CJ, Samad TA: Nociceptors are interleukin-1 beta sensors. J Neurosci. 2008, 28: 14062-14073. 10.1523/JNEUROSCI.3795-08.2008.CrossRefPubMedCentralPubMed
45.
go back to reference Takeda M, Kitagawa J, Takahashi M, Matsumoto S: Activation of interleukin-1 beta receptor suppresses the voltage-gated potassium currents in the small-diameter trigeminal ganglion neurons following peripheral inflammation. Pain. 2008, 139: 594-602. 10.1016/j.pain.2008.06.015.CrossRefPubMed Takeda M, Kitagawa J, Takahashi M, Matsumoto S: Activation of interleukin-1 beta receptor suppresses the voltage-gated potassium currents in the small-diameter trigeminal ganglion neurons following peripheral inflammation. Pain. 2008, 139: 594-602. 10.1016/j.pain.2008.06.015.CrossRefPubMed
46.
go back to reference Kleibeuker W, Ledeboer A, Eijkelkamp N, Watkins LR, Maier SF, Zijlstra J, Heijnen CJ, Kavelaars A: A role for G protein-coupled receptor kinase 2 in mechanical allodynia. Eur J Neurosci. 2007, 25: 1696-1704. 10.1111/j.1460-9568.2007.05423.x.CrossRefPubMed Kleibeuker W, Ledeboer A, Eijkelkamp N, Watkins LR, Maier SF, Zijlstra J, Heijnen CJ, Kavelaars A: A role for G protein-coupled receptor kinase 2 in mechanical allodynia. Eur J Neurosci. 2007, 25: 1696-1704. 10.1111/j.1460-9568.2007.05423.x.CrossRefPubMed
47.
48.
go back to reference Lubberts E: IL-17/Th17 targeting: on the road to prevent chronic destructive arthritis?. Cytokine. 2008, 41: 84-91. 10.1016/j.cyto.2007.09.014.CrossRefPubMed Lubberts E: IL-17/Th17 targeting: on the road to prevent chronic destructive arthritis?. Cytokine. 2008, 41: 84-91. 10.1016/j.cyto.2007.09.014.CrossRefPubMed
49.
go back to reference Pappu R, Ramirez-Carrozzi V, Ota N, Ouyang W, Hu Y: The IL-17 family cytokines in immunity and disease. J Clin Immunol. 2010, 30: 185-195. 10.1007/s10875-010-9369-6.CrossRefPubMed Pappu R, Ramirez-Carrozzi V, Ota N, Ouyang W, Hu Y: The IL-17 family cytokines in immunity and disease. J Clin Immunol. 2010, 30: 185-195. 10.1007/s10875-010-9369-6.CrossRefPubMed
50.
go back to reference Hueber W, Patel DD, Dryja T, Wright AM, Koroleva I, Bruin G, Antoni C, Drealos Z, Gold MH, Durez P, Tal PP, Gomez-Reino JJ, Foster CS, Kim RY, Samson CM, Falk NS, Chu DS, Callanan D, Nguyen QD, Rose K, Haider A, DiPadova F: Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis. Sci Transl Med. 2010, 2: 52ra72-10.1126/scitranslmed.3001107.PubMed Hueber W, Patel DD, Dryja T, Wright AM, Koroleva I, Bruin G, Antoni C, Drealos Z, Gold MH, Durez P, Tal PP, Gomez-Reino JJ, Foster CS, Kim RY, Samson CM, Falk NS, Chu DS, Callanan D, Nguyen QD, Rose K, Haider A, DiPadova F: Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis. Sci Transl Med. 2010, 2: 52ra72-10.1126/scitranslmed.3001107.PubMed
51.
go back to reference Segond von Banchet G, Boettger MK, König C, Iwakura Y, Bräuer R, Schaible H-G: Neuronal IL-17 receptor upregulates TRPV4 but not TRPV1 receptors in DRG neurons and mediates mechanical but not thermal hyperalgesia. Mol Cell Neurosci. 2013, 52: 152-160. 10.1016/j.mcn.2012.11.006.CrossRefPubMed Segond von Banchet G, Boettger MK, König C, Iwakura Y, Bräuer R, Schaible H-G: Neuronal IL-17 receptor upregulates TRPV4 but not TRPV1 receptors in DRG neurons and mediates mechanical but not thermal hyperalgesia. Mol Cell Neurosci. 2013, 52: 152-160. 10.1016/j.mcn.2012.11.006.CrossRefPubMed
52.
go back to reference Pinto LG, Cunha TM, Vieira SM, Lemos HP, Verri WA, Cunha FQ, Ferreira SH: IL-17 mediates articular hypernociception in antigen-induced arthritis in mice. Pain. 2010, 148: 247-256. 10.1016/j.pain.2009.11.006.CrossRefPubMed Pinto LG, Cunha TM, Vieira SM, Lemos HP, Verri WA, Cunha FQ, Ferreira SH: IL-17 mediates articular hypernociception in antigen-induced arthritis in mice. Pain. 2010, 148: 247-256. 10.1016/j.pain.2009.11.006.CrossRefPubMed
53.
go back to reference McNamee KE, Alzabin S, Hughes JP, Anand P, Feldmann M, Williams RO, Inglis JJ: IL-17 induces hyperalgesia via TNF-dependent neutrophil infiltration. Pain. 2011, 152: 1838-1845. 10.1016/j.pain.2011.03.035.CrossRefPubMed McNamee KE, Alzabin S, Hughes JP, Anand P, Feldmann M, Williams RO, Inglis JJ: IL-17 induces hyperalgesia via TNF-dependent neutrophil infiltration. Pain. 2011, 152: 1838-1845. 10.1016/j.pain.2011.03.035.CrossRefPubMed
54.
go back to reference Richter F, Natura G, Ebbinghaus M, Segond von Banchet G, Hensellek S, König C, Bräuer R, Schaible H-G: Interleukin-17 sensitizes joint nociceptors for mechanical stimuli and contributes to arthritic pain through neuronal IL-17 receptors in rodents. Arthritis Rheum. 2012, 64: 4125-4134. 10.1002/art.37695.CrossRefPubMed Richter F, Natura G, Ebbinghaus M, Segond von Banchet G, Hensellek S, König C, Bräuer R, Schaible H-G: Interleukin-17 sensitizes joint nociceptors for mechanical stimuli and contributes to arthritic pain through neuronal IL-17 receptors in rodents. Arthritis Rheum. 2012, 64: 4125-4134. 10.1002/art.37695.CrossRefPubMed
55.
go back to reference Liedtke W: Molecular mechanisms of TRPV4-mediated neural signaling. Ann NY Acad Sci. 2008, 1144: 42-52. 10.1196/annals.1418.012.CrossRefPubMed Liedtke W: Molecular mechanisms of TRPV4-mediated neural signaling. Ann NY Acad Sci. 2008, 1144: 42-52. 10.1196/annals.1418.012.CrossRefPubMed
56.
go back to reference Kim CF, Malem-Taylor G: Interleukin-17 contributes to neuroinflammation and neuropathic pain following peripheral nerve injury in mice. J Pain. 2011, 12: 370-383. 10.1016/j.jpain.2010.08.003.CrossRefPubMed Kim CF, Malem-Taylor G: Interleukin-17 contributes to neuroinflammation and neuropathic pain following peripheral nerve injury in mice. J Pain. 2011, 12: 370-383. 10.1016/j.jpain.2010.08.003.CrossRefPubMed
57.
go back to reference Santello M, Volterra A: TNFα in synaptic function: switching gears. TINS. 2012, 35: 638-647.PubMed Santello M, Volterra A: TNFα in synaptic function: switching gears. TINS. 2012, 35: 638-647.PubMed
Metadata
Title
Nociceptive neurons detect cytokines in arthritis
Author
Hans-Georg Schaible
Publication date
01-10-2014
Publisher
BioMed Central
Published in
Arthritis Research & Therapy / Issue 5/2014
Electronic ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-014-0470-8

Other articles of this Issue 5/2014

Arthritis Research & Therapy 5/2014 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine