High levels of cerebrospinal fluid chemokines point to the presence of neuroinflammation in peripheral neuropathic pain: a cross-sectional study of 2 cohorts of patients compared with healthy controls

Emmanuel Bäckryd, Anne-Li Lind, Måns Thulin, Anders Larsson, Björn Gerdle, Torsten Gordh, Emmanuel Bäckryd, Anne-Li Lind, Måns Thulin, Anders Larsson, Björn Gerdle, Torsten Gordh

Abstract

Animal models suggest that chemokines are important mediators in the pathophysiology of neuropathic pain. Indeed, these substances have been called "gliotransmitters," a term that illustrates the close interplay between glial cells and neurons in the context of neuroinflammation and pain. However, evidence in humans is scarce. The aim of the study was to determine a comprehensive cerebrospinal fluid (CSF) inflammatory profile of patients with neuropathic pain. Our hypothesis was that we would thereby find indications of a postulated on-going process of central neuroinflammation. Samples of CSF were collected from 2 cohorts of patients with neuropathic pain (n = 11 and n = 16, respectively) and healthy control subjects (n = 11). The samples were analyzed with a multiplex proximity extension assay in which 92 inflammation-related proteins were measured simultaneously (Proseek Multiplex Inflammation I; Olink Bioscience, Uppsala, Sweden). Univariate testing with control of false discovery rate, as well as orthogonal partial least squares discriminant analysis, were used for statistical analyses. Levels of chemokines CXCL6, CXCL10, CCL8, CCL11, CCL23 in CSF, as well as protein LAPTGF-beta-1, were significantly higher in both neuropathic pain cohorts compared with healthy controls, pointing to neuroinflammation in patients. These 6 proteins were also major results in a recent similar study in patients with fibromyalgia. The findings need to be confirmed in larger cohorts, and the question of causality remains to be settled. Because it has been suggested that prevalent comorbidities to chronic pain (eg, depression, anxiety, poor sleep, and tiredness) also are associated with neuroinflammation, it will be important to determine whether neuroinflammation is a common mediator.

Conflict of interest statement

Sponsorships or competing interests that may be relevant to content are disclosed at the end of this article.

Figures

Figure 1.
Figure 1.
Expression of the 11 most group-discriminating inflammation-related proteins in the cerebrospinal fluid of neuropathic pain patients (cohort 1a) versus healthy control subjects (cohort 1b). The protein levels (y-axis) are expressed as normalized protein expression, as described in the proximity extension assay subsection. Median values are represented by horizontal lines and the interquartile ranges by boxes. The ends of the whiskers depict the lowest and highest datum within 1.5 interquartile range of the lower or upper quartile, respectively. Points represent outliers.
Figure 2.
Figure 2.
Two-dimensional score plot of orthogonal partial least squares discriminant analysis comparing inflammation-related proteins in the cerebrospinal fluid of patients with neuropathic pain (cohort 1a) with healthy control subjects (cohort 1b). Class separation between neuropathic pain patients (n = 11, green dots marked “1”) and healthy controls (n = 11, blue dots marked “2”) occurs along the t[1] axis (interclass variation). The to[1] axis represents intraclass variation. The ellipse represents the Hotelling T2 95% confidence interval used when identifying strong outliers.

References

    1. Almay BG, Johansson F, Von Knorring L, Le Greves P, Terenius L. Substance P in CSF of patients with chronic pain syndromes. PAIN 1988;33:3–9.
    1. Antunes-Martins A, Perkins JR, Lees J, Hildebrandt T, Orengo C, Bennett DL. Systems biology approaches to finding novel pain mediators. Wiley Interdiscip Rev Syst Biol Med 2013;5:11–35.
    1. Assarsson E, Lundberg M, Holmquist G, Bjorkesten J, Thorsen SB, Ekman D, Eriksson A, Rennel Dickens E, Ohlsson S, Edfeldt G, Andersson AC, Lindstedt P, Stenvang J, Gullberg M, Fredriksson S. Homogenous 96-plex PEA immunoassay exhibiting high sensitivity, specificity, and excellent scalability. PLoS One 2014;9:e95192.
    1. Baraniuk JN, Whalen G, Cunningham J, Clauw DJ. Cerebrospinal fluid levels of opioid peptides in fibromyalgia and chronic low back pain. BMC Musculoskelet Disord 2004;5:48.
    1. Baron R, Binder A, Wasner G. Neuropathic pain: diagnosis, pathophysiological mechanisms, and treatment. Lancet Neurol 2010;9:807–19.
    1. Beggs S, Trang T, Salter MW. P2X4R+ microglia drive neuropathic pain. Nat Neurosci 2012;15:1068–73.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B Stat Methodol 1995;57:289–300.
    1. Bouhassira D, Lanteri-Minet M, Attal N, Laurent B, Touboul C. Prevalence of chronic pain with neuropathic characteristics in the general population. PAIN 2008;136:380–7.
    1. Brisby H, Olmarker K, Larsson K, Nutu M, Rydevik B. Proinflammatory cytokines in cerebrospinal fluid and serum in patients with disc herniation and sciatica. Eur Spine J 2002;11:62–6.
    1. Bäckryd E, Ghafouri B, Carlsson AK, Olausson P, Gerdle B. Multivariate proteomic analysis of the cerebrospinal fluid of patients with peripheral neuropathic pain and healthy controls—a hypothesis-generating pilot study. J Pain Res 2015;8:321–33.
    1. Bäckryd E, Ghafouri B, Larsson B, Gerdle B. Do low levels of beta-endorphin in the cerebrospinal fluid indicate defective top-down inhibition in patients with chronic neuropathic pain? A cross-sectional, comparative study. Pain Med 2014;15:111–19.
    1. Bäckryd E, Sorensen J, Gerdle B. Ziconotide trialing by intrathecal bolus injections: an open-label non-randomized clinical trial in postoperative/posttraumatic neuropathic pain patients refractory to conventional treatment. Neuromodulation 2015;18:404–13.
    1. Bäckryd E, Tanum L, Lind AL, Larsson A, Gordh T. Evidence of both systemic inflammation and neuroinflammation in fibromyalgia patients, as assessed by a multiplex protein panel applied to the cerebrospinal fluid and to plasma. J Pain Res 2017;10:515–25.
    1. Calvo M, Dawes JM, Bennett DL. The role of the immune system in the generation of neuropathic pain. Lancet Neurol 2012;11:629–42.
    1. Capelle HH, Weigel R, Schmelz M, Krauss JK. Neurotrophins in the cerebrospinal fluid of patient cohorts with neuropathic pain, nociceptive pain, or normal pressure hydrocephalus. Clin J Pain 2009;25:729–33.
    1. Carr FB, Geranton SM, Hunt SP. Descending controls modulate inflammatory joint pain and regulate CXC chemokine and iNOS expression in the dorsal horn. Mol Pain 2014;10:39.
    1. Chen G, Park CK, Xie RG, Berta T, Nedergaard M, Ji RR. Connexin-43 induces chemokine release from spinal cord astrocytes to maintain late-phase neuropathic pain in mice. Brain 2014;137:2193–209.
    1. Clark AK, Malcangio M. Fractalkine/CX3CR1 signaling during neuropathic pain. Front Cell Neurosci 2014;8:121.
    1. Clark AK, Yip PK, Grist J, Gentry C, Staniland AA, Marchand F, Dehvari M, Wotherspoon G, Winter J, Ullah J, Bevan S, Malcangio M. Inhibition of spinal microglial cathepsin S for the reversal of neuropathic pain. Proc Natl Acad Sci U S A 2007;104:10655–60.
    1. Clark AK, Yip PK, Malcangio M. The liberation of fractalkine in the dorsal horn requires microglial cathepsin S. J Neurosci 2009;29:6945–54.
    1. Colburn RW, Rickman AJ, DeLeo JA. The effect of site and type of nerve injury on spinal glial activation and neuropathic pain behavior. Exp Neurol 1999;157:289–304.
    1. 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.
    1. Dominguez CA, Kalliomaki M, Gunnarsson U, Moen A, Sandblom G, Kockum I, Lavant E, Olsson T, Nyberg F, Rygh LJ, Roe C, Gjerstad J, Gordh T, Piehl F. The DQB1 *03:02 HLA haplotype is associated with increased risk of chronic pain after inguinal hernia surgery and lumbar disc herniation. PAIN 2013;154:427–33.
    1. Ellis A, Bennett DL. Neuroinflammation and the generation of neuropathic pain. Br J Anaesth 2013;111:26–37.
    1. Eriksson L, Byrne T, Johansson E, Trygg J, Vikström C. Multi- and megavariate data analysis: basic principles and applications. Malmö: MKS Umetrics AB 2013:1–254.
    1. Estes ML, McAllister AK. Alterations in immune cells and mediators in the brain: it's not always neuroinflammation! Brain Pathol 2014;24:623–30.
    1. Finnerup NB, Attal N, Haroutounian S, McNicol E, Baron R, Dworkin RH, Gilron I, Haanpaa M, Hansson P, Jensen TS, Kamerman PR, Lund K, Moore A, Raja SN, Rice AS, Rowbotham M, Sena E, Siddall P, Smith BH, Wallace M. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol 2015;14:162–73.
    1. Gao YJ, Ji RR. Chemokines, neuronal-glial interactions, and central processing of neuropathic pain. Pharmacol Ther 2010;126:56–68.
    1. 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;29:4096–108.
    1. Gerdle B, Ghafouri B, Ghafouri N, Backryd E, Gordh T. Signs of ongoing inflammation in female patients with chronic widespread pain: a multivariate, explorative, cross-sectional study of blood samples. Medicine (Baltimore) 2017;96:e6130.
    1. Gosselin RD, Suter MR, Ji RR, Decosterd I. Glial cells and chronic pain. Neuroscientist 2010;16:519–31.
    1. Grace PM, Hutchinson MR, Maier SF, Watkins LR. Pathological pain and the neuroimmune interface. Nat Rev Immunol 2014;14:217–31.
    1. Jensen TS, Baron R, Haanpaa M, Kalso E, Loeser JD, Rice AS, Treede RD. A new definition of neuropathic pain. PAIN 2011;152:2204–5.
    1. Ji RR, Berta T, Nedergaard M. Glia and pain: is chronic pain a gliopathy? PAIN 2013;154(suppl 1):S10–28.
    1. Ji RR, Chamessian A, Zhang YQ. Pain regulation by non-neuronal cells and inflammation. Science 2016;354:572–7.
    1. Ji RR, Xu ZZ, Gao YJ. Emerging targets in neuroinflammation-driven chronic pain. Nat Rev Drug Discov 2014;13:533–48.
    1. Jiang BC, He LN, Wu XB, Shi H, Zhang WW, Zhang ZJ, Cao DL, Li CH, Gu J, Gao YJ. Promoted interaction of C/EBPalpha with demethylated Cxcr3 gene promoter contributes to neuropathic pain in mice. J Neurosci 2017;37:685–700.
    1. Kadetoff D, Lampa J, Westman M, Andersson M, Kosek E. Evidence of central inflammation in fibromyalgia-increased cerebrospinal fluid interleukin-8 levels. J Neuroimmunol 2012;242:33–8.
    1. Karshikoff B, Jensen KB, Kosek E, Kalpouzos G, Soop A, Ingvar M, Olgart Hoglund C, Lekander M, Axelsson J. Why sickness hurts: a central mechanism for pain induced by peripheral inflammation. Brain Behav Immun 2016;57:38–46.
    1. Katsura H, Obata K, Mizushima T, Sakurai J, Kobayashi K, Yamanaka H, Dai Y, Fukuoka T, Sakagami M, Noguchi K. Activation of Src-family kinases in spinal microglia contributes to mechanical hypersensitivity after nerve injury. J Neurosci 2006;26:8680–90.
    1. Kawasaki Y, Zhang L, Cheng JK, Ji RR. Cytokine mechanisms of central sensitization: distinct and overlapping role of interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in regulating synaptic and neuronal activity in the superficial spinal cord. J Neurosci 2008;28:5189–94.
    1. Kiguchi N, Kobayashi Y, Kishioka S. Chemokines and cytokines in neuroinflammation leading to neuropathic pain. Curr Opin Pharmacol 2012;12:55–61.
    1. Kwiatkowski K, Piotrowska A, Rojewska E, Makuch W, Jurga A, Slusarczyk J, Trojan E, Basta-Kaim A, Mika J. Beneficial properties of maraviroc on neuropathic pain development and opioid effectiveness in rats. Prog Neuropsychopharmacol Biol Psychiatry 2016;64:68–78.
    1. Larsson A, Carlsson L, Gordh T, Lind AL, Thulin M, Kamali-Moghaddam M. The effects of age and gender on plasma levels of 63 cytokines. J Immunol Methods 2015;425:58–61.
    1. Lin CP, Kang KH, Lin TH, Wu MY, Liou HC, Chuang WJ, Sun WZ, Fu WM. Role of spinal CXCL1 (GROalpha) in opioid tolerance: a human-to-rodent translational study. Anesthesiology 2015;122:666–76.
    1. Lind AL, Emami Khoonsari P, Sjodin M, Katila L, Wetterhall M, Gordh T, Kultima K. Spinal cord stimulation alters protein levels in the cerebrospinal fluid of neuropathic pain patients: a proteomic mass spectrometric analysis. Neuromodulation 2016;19:549–62.
    1. Lundberg M, Eriksson A, Tran B, Assarsson E, Fredriksson S. Homogeneous antibody-based proximity extension assays provide sensitive and specific detection of low-abundant proteins in human blood. Nucleic Acids Res 2011;39:e102.
    1. Lundborg C, Hahn-Zoric M, Biber B, Hansson E. Glial cell line-derived neurotrophic factor is increased in cerebrospinal fluid but decreased in blood during long-term pain. J Neuroimmunol 2010;220:108–13.
    1. Makker PG, Duffy SS, Lees JG, Perera CJ, Tonkin RS, Butovsky O, Park SB, Goldstein D, Moalem-Taylor G. Characterisation of immune and neuroinflammatory changes associated with chemotherapy-induced peripheral neuropathy. PLoS One 2017;12:e0170814.
    1. Mao J. Translational pain research: achievements and challenges. J Pain 2009;10:1001–11.
    1. Mika J, Zychowska M, Popiolek-Barczyk K, Rojewska E, Przewlocka B. Importance of glial activation in neuropathic pain. Eur J Pharmacol 2013;716:106–19.
    1. Miller RJ, Jung H, Bhangoo SK, White FA. Cytokine and chemokine regulation of sensory neuron function. Handb Exp Pharmacol 2009:417–49.
    1. Milligan E, Zapata V, Schoeniger D, Chacur M, Green P, Poole S, Martin D, Maier SF, Watkins LR. An initial investigation of spinal mechanisms underlying pain enhancement induced by fractalkine, a neuronally released chemokine. Eur J Neurosci 2005;22:2775–82.
    1. Milligan ED, Zapata V, Chacur M, Schoeniger D, Biedenkapp J, O'Connor KA, Verge GM, Chapman G, Green P, Foster AC, Naeve GS, Maier SF, Watkins LR. Evidence that exogenous and endogenous fractalkine can induce spinal nociceptive facilitation in rats. Eur J Neurosci 2004;20:2294–302.
    1. Moen A, Lind AL, Thulin M, Kamali-Moghaddam M, Roe C, Gjerstad J, Gordh T. Inflammatory serum protein profiling of patients with lumbar radicular pain one year after disc herniation. Int J Inflam 2016;2016:3874964.
    1. Mullington JM, Simpson NS, Meier-Ewert HK, Haack M. Sleep loss and inflammation. Best Pract Res Clin Endocrinol Metab 2010;24:775–84.
    1. O'Callaghan JP, Sriram K, Miller DB. Defining “neuroinflammation.” Ann N Y Acad Sci 2008;1139:318–30.
    1. O'Hayre M, Salanga CL, Handel TM, Hamel DJ. Emerging concepts and approaches for chemokine-receptor drug discovery. Expert Opin Drug Discov 2010;5:1109–22.
    1. Olausson P, Gerdle B, Ghafouri N, Sjostrom D, Blixt E, Ghafouri B. Protein alterations in women with chronic widespread pain—an explorative proteomic study of the trapezius muscle. Sci Rep 2015;5:11894.
    1. Old EA, Clark AK, Malcangio M. The role of glia in the spinal cord in neuropathic and inflammatory pain. Handb Exp Pharmacol 2015;227:145–70.
    1. Parisien M, Khoury S, Chabot-Dore AJ, Sotocinal SG, Slade GD, Smith SB, Fillingim RB, Ohrbach R, Greenspan JD, Maixner W, Mogil JS, Belfer I, Diatchenko L. Effect of human genetic variability on gene expression in dorsal root ganglia and association with pain phenotypes. Cell Rep 2017;19:1940–52.
    1. Pedersen BK. Muscles and their myokines. J Exp Biol 2011;214:337–46.
    1. Raghavendra V, Tanga F, DeLeo JA. Inhibition of microglial activation attenuates the development but not existing hypersensitivity in a rat model of neuropathy. J Pharmacol Exp Ther 2003;306:624–30.
    1. Roche S, Gabelle A, Lehmann S. Clinical proteomics of the cerebrospinal fluid: towards the discovery of new biomarkers. Proteomics Clin Appl 2008;2:428–36.
    1. Russell IJ, Orr MD, Littman B, Vipraio GA, Alboukrek D, Michalek JE, Lopez Y, MacKillip F. Elevated cerebrospinal fluid levels of substance P in patients with the fibromyalgia syndrome. Arthritis Rheum 1994;37:1593–601.
    1. Sato-Takeda M, Ihn H, Ohashi J, Tsuchiya N, Satake M, Arita H, Tamaki K, Hanaoka K, Tokunaga K, Yabe T. The human histocompatibility leukocyte antigen (HLA) haplotype is associated with the onset of postherpetic neuralgia after herpes zoster. PAIN 2004;110:329–36.
    1. Schmitz K, Pickert G, Wijnvoord N, Haussler A, Tegeder I. Dichotomy of CCL21 and CXCR3 in nerve injury-evoked and autoimmunity-evoked hyperalgesia. Brain Behav Immun 2013;32:186–200.
    1. Strong JA, Xie W, Coyle DE, Zhang JM. Microarray analysis of rat sensory ganglia after local inflammation implicates novel cytokines in pain. PLoS One 2012;7:e40779.
    1. 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;13:263–72.
    1. Themistocleous AC, Ramirez JD, Shillo PR, Lees JG, Selvarajah D, Orengo C, Tesfaye S, Rice AS, Bennett DL. The Pain in Neuropathy Study (PiNS): a cross-sectional observational study determining the somatosensory phenotype of painful and painless diabetic neuropathy. PAIN 2016;157:1132–45.
    1. Tiwari V, Guan Y, Raja SN. Modulating the delicate glial-neuronal interactions in neuropathic pain: promises and potential caveats. Neurosci Biobehav Rev 2014;45C:19–27.
    1. Treede RD, Jensen TS, Campbell JN, Cruccu G, Dostrovsky JO, Griffin JW, Hansson P, Hughes R, Nurmikko T, Serra J. Neuropathic pain: redefinition and a grading system for clinical and research purposes. Neurology 2008;70:1630–5.
    1. Vaeroy H, Helle R, Forre O, Kass E, Terenius L. Cerebrospinal fluid levels of beta-endorphin in patients with fibromyalgia (fibrositis syndrome). J Rheumatol 1988;15:1804–6.
    1. Vaeroy H, Helle R, Forre O, Kass E, Terenius L. Elevated CSF levels of substance P and high incidence of Raynaud phenomenon in patients with fibromyalgia: new features for diagnosis. PAIN 1988;32:21–6.
    1. Walker AK, Kavelaars A, Heijnen CJ, Dantzer R. Neuroinflammation and comorbidity of pain and depression. Pharmacol Rev 2014;66:80–101.
    1. Vallejo R, Tilley DM, Vogel L, Benyamin R. The role of glia and the immune system in the development and maintenance of neuropathic pain. Pain Pract 2010;10:167–84.
    1. van Rooijen DE, Roelen DL, Verduijn W, Haasnoot GW, Huygen FJ, Perez RS, Claas FH, Marinus J, van Hilten JJ, van den Maagdenberg AM. Genetic HLA associations in complex regional pain syndrome with and without dystonia. J Pain 2012;13:784–9.
    1. Watkins LR, Milligan ED, Maier SF. Mechanisms of glial activation after nerve injury. In: Basbaum AI, Bushnell MC, editors. Science of pain. Oxford, UK: Elsevier, 2009. pp. 429–33.
    1. Wheelock AM, Wheelock CE. Trials and tribulations of 'omics data analysis: assessing quality of SIMCA-based multivariate models using examples from pulmonary medicine. Mol Biosyst 2013;9:2589–96.
    1. Zhang J, De Koninck Y. Spatial and temporal relationship between monocyte chemoattractant protein-1 expression and spinal glial activation following peripheral nerve injury. J Neurochem 2006;97:772–83.
    1. Zhang ZJ, Cao DL, Zhang X, Ji RR, Gao YJ. Chemokine contribution to neuropathic pain: respective induction of CXCL1 and CXCR2 in spinal cord astrocytes and neurons. PAIN 2013;154:2185–97.
    1. Zhuang ZY, Kawasaki Y, Tan PH, Wen YR, Huang J, Ji RR. Role of the CX3CR1/p38 MAPK pathway in spinal microglia for the development of neuropathic pain following nerve injury-induced cleavage of fractalkine. Brain Behav Immun 2007;21:642–651.

Source: PubMed

3
Iratkozz fel