Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2016

Open Access 01-12-2016 | Research

Endogenous analgesia mediated by CD4+ T lymphocytes is dependent on enkephalins in mice

Authors: Lilian Basso, Jérôme Boué, Karim Mahiddine, Catherine Blanpied, Sébastien Robiou-du-Pont, Nathalie Vergnolle, Céline Deraison, Gilles Dietrich

Published in: Journal of Neuroinflammation | Issue 1/2016

Login to get access

Abstract

Background

T cell-derived opioids play a key role in the control of inflammatory pain. However, the nature of opioids produced by T cells is still matter of debate in mice. Whereas β-endorphin has been found in T lymphocytes by using antibody-based methods, messenger RNA (mRNA) quantification shows mainly mRNA encoding for enkephalins. The objective of the study is to elucidate the nature of T cell-derived opioids responsible for analgesia and clarify discrepancy of the results at the protein and genetic levels.

Methods

CD4+ T lymphocytes were isolated from wild-type and enkephalin-deficient mice. mRNA encoding for β-endorphin and enkephalin was quantified by RT-qPCR. The binding of commercially available polyclonal anti-endorphin antibodies to lymphocytes from wild-type or enkephalin knockout mice was assessed by cytofluorometry. Opioid-mediated analgesic properties of T lymphocytes from wild-type and enkephalin-deficient mice were compared in a model of inflammation-induced somatic pain by measuring sensitivity to mechanical stimuli using calibrated von Frey filaments.

Results

CD4+ T lymphocytes expressed high level of mRNA encoding for enkephalins but not for β-endorphin in mice. Anti-β-endorphin polyclonal IgG antibodies are specific for β-endorphin but cross-react with enkephalins. Anti-β-endorphin polyclonal antibodies bound to wild-type but not enkephalin-deficient CD4+ T lymphocytes. Endogenous regulation of inflammatory pain by wild-type T lymphocytes was completely abolished when T lymphocytes were deficient in enkephalins. Pain behavior of immune-deficient (i.e., without B and T lymphocytes) mice was superimposable to that of mice transferred with enkephalin-deficient lymphocytes.

Conclusions

Rabbit polyclonal anti-β-endorphin serum IgG bind to CD4+ T lymphocytes because of their cross-reactivity towards enkephalins. Thus, staining of T lymphocytes by anti-β-endorphin polyclonal IgG reported in most of studies in mice is because of their binding to enkephalins. In mice, CD4+ T lymphocytes completely lose their analgesic opioid-mediated activity when lacking enkephalins.
Literature
1.
go back to reference Boue J, Blanpied C, Brousset P, Vergnolle N, Dietrich G. Endogenous opioid-mediated analgesia is dependent on adaptive T cell response in mice. J Immunol. 2011;186:5078–84.CrossRefPubMed Boue J, Blanpied C, Brousset P, Vergnolle N, Dietrich G. Endogenous opioid-mediated analgesia is dependent on adaptive T cell response in mice. J Immunol. 2011;186:5078–84.CrossRefPubMed
2.
go back to reference Cabot PJ, Carter L, Gaiddon C, Zhang Q, Schäfer M, Loeffler J, Stein C. Immune cell-derived ß-endorphin. Production, release, and control of inflammatory pain in rats. J Clin Invest. 1997;100:142–8.CrossRefPubMedPubMedCentral Cabot PJ, Carter L, Gaiddon C, Zhang Q, Schäfer M, Loeffler J, Stein C. Immune cell-derived ß-endorphin. Production, release, and control of inflammatory pain in rats. J Clin Invest. 1997;100:142–8.CrossRefPubMedPubMedCentral
3.
go back to reference Machelska H, Cabot PJ, Mousa SA, Zhang Q, Stein C. Pain control in inflammation governed by selectins. Nat Med. 1998;4:1425–8.CrossRefPubMed Machelska H, Cabot PJ, Mousa SA, Zhang Q, Stein C. Pain control in inflammation governed by selectins. Nat Med. 1998;4:1425–8.CrossRefPubMed
4.
go back to reference Machelska H, Schopohl JK, Mousa SA, Labuz D, Schafer M, Stein C. Different mechanisms of intrinsic pain inhibition in early and late inflammation. J Neuroimmunol. 2003;141:30–9.CrossRefPubMed Machelska H, Schopohl JK, Mousa SA, Labuz D, Schafer M, Stein C. Different mechanisms of intrinsic pain inhibition in early and late inflammation. J Neuroimmunol. 2003;141:30–9.CrossRefPubMed
5.
go back to reference Rittner HL, Brack A, Machelska H, Mousa SA, Bauer M, Schafer M, Stein C. Opioid peptide-expressing leukocytes: identification, recruitment, and simultaneously increasing inhibition of inflammatory pain. Anesthesiology. 2001;95:500–8.CrossRefPubMed Rittner HL, Brack A, Machelska H, Mousa SA, Bauer M, Schafer M, Stein C. Opioid peptide-expressing leukocytes: identification, recruitment, and simultaneously increasing inhibition of inflammatory pain. Anesthesiology. 2001;95:500–8.CrossRefPubMed
6.
go back to reference Boue J, Blanpied C, Djata-Cabral M, Pelletier L, Vergnolle N, Dietrich G. Immune conditions associated with CD4+ T effector-induced opioid release and analgesia. Pain. 2012;153:485–93.CrossRefPubMed Boue J, Blanpied C, Djata-Cabral M, Pelletier L, Vergnolle N, Dietrich G. Immune conditions associated with CD4+ T effector-induced opioid release and analgesia. Pain. 2012;153:485–93.CrossRefPubMed
7.
go back to reference Gaveriaux-Ruff C, Karchewski LA, Hever X, Matifas A, Kieffer BL. Inflammatory pain is enhanced in delta opioid receptor-knockout mice. Eur J Neurosci. 2008;27:2558–67.CrossRefPubMedPubMedCentral Gaveriaux-Ruff C, Karchewski LA, Hever X, Matifas A, Kieffer BL. Inflammatory pain is enhanced in delta opioid receptor-knockout mice. Eur J Neurosci. 2008;27:2558–67.CrossRefPubMedPubMedCentral
8.
go back to reference Gaveriaux-Ruff C, Nozaki C, Nadal X, Hever XC, Weibel R, Matifas A, Reiss D, Filliol D, Nassar MA, Wood JN, Maldonado R, Kieffer BL. Genetic ablation of delta opioid receptors in nociceptive sensory neurons increases chronic pain and abolishes opioid analgesia. Pain. 2011;152:1238–48.CrossRefPubMed Gaveriaux-Ruff C, Nozaki C, Nadal X, Hever XC, Weibel R, Matifas A, Reiss D, Filliol D, Nassar MA, Wood JN, Maldonado R, Kieffer BL. Genetic ablation of delta opioid receptors in nociceptive sensory neurons increases chronic pain and abolishes opioid analgesia. Pain. 2011;152:1238–48.CrossRefPubMed
9.
go back to reference Weibel R, Reiss D, Karchewski L, Gardon O, Matifas A, Filliol D, Becker JA, Wood JN, Kieffer BL, Gaveriaux-Ruff C. Mu opioid receptors on primary afferent nav1.8 neurons contribute to opiate-induced analgesia: insight from conditional knockout mice. PLoS One. 2013;8:e74706.CrossRefPubMedPubMedCentral Weibel R, Reiss D, Karchewski L, Gardon O, Matifas A, Filliol D, Becker JA, Wood JN, Kieffer BL, Gaveriaux-Ruff C. Mu opioid receptors on primary afferent nav1.8 neurons contribute to opiate-induced analgesia: insight from conditional knockout mice. PLoS One. 2013;8:e74706.CrossRefPubMedPubMedCentral
10.
go back to reference Boue J, Basso L, Cenac N, Blanpied C, Rolli-Derkinderen M, Neunlist M, Vergnolle N, Dietrich G. Endogenous regulation of visceral pain via production of opioids by colitogenic CD4(+) T cells in mice. Gastroenterology. 2014;146:166–75.CrossRefPubMed Boue J, Basso L, Cenac N, Blanpied C, Rolli-Derkinderen M, Neunlist M, Vergnolle N, Dietrich G. Endogenous regulation of visceral pain via production of opioids by colitogenic CD4(+) T cells in mice. Gastroenterology. 2014;146:166–75.CrossRefPubMed
11.
go back to reference Labuz D, Schmidt Y, Schreiter A, Rittner HL, Mousa SA, Machelska H. Immune cell-derived opioids protect against neuropathic pain in mice. J Clin Invest. 2009;119:278–86.CrossRefPubMedPubMedCentral Labuz D, Schmidt Y, Schreiter A, Rittner HL, Mousa SA, Machelska H. Immune cell-derived opioids protect against neuropathic pain in mice. J Clin Invest. 2009;119:278–86.CrossRefPubMedPubMedCentral
12.
go back to reference Labuz D, Schreiter A, Schmidt Y, Brack A, Machelska H. T lymphocytes containing beta-endorphin ameliorate mechanical hypersensitivity following nerve injury. Brain Behav Immun. 2010;24:1045–53.CrossRefPubMed Labuz D, Schreiter A, Schmidt Y, Brack A, Machelska H. T lymphocytes containing beta-endorphin ameliorate mechanical hypersensitivity following nerve injury. Brain Behav Immun. 2010;24:1045–53.CrossRefPubMed
13.
go back to reference Valdez-Morales E, Guerrero-Alba R, Ochoa-Cortes F, Benson J, Spreadbury I, Hurlbut D, Miranda-Morales M, Lomax AE, Vanner S. Release of endogenous opioids during a chronic IBD model suppresses the excitability of colonic DRG neurons. Neurogastroenterol Motil. 2013;25:39–46.CrossRefPubMed Valdez-Morales E, Guerrero-Alba R, Ochoa-Cortes F, Benson J, Spreadbury I, Hurlbut D, Miranda-Morales M, Lomax AE, Vanner S. Release of endogenous opioids during a chronic IBD model suppresses the excitability of colonic DRG neurons. Neurogastroenterol Motil. 2013;25:39–46.CrossRefPubMed
14.
go back to reference Verma-Gandhu M, Bercik P, Motomura Y, Verdu EF, Khan WI, Blennerhassett PA, Wang L, El-Sharkawy RT, Collins SM. CD4+ T-cell modulation of visceral nociception in mice. Gastroenterology. 2006;130:1721–8.CrossRefPubMed Verma-Gandhu M, Bercik P, Motomura Y, Verdu EF, Khan WI, Blennerhassett PA, Wang L, El-Sharkawy RT, Collins SM. CD4+ T-cell modulation of visceral nociception in mice. Gastroenterology. 2006;130:1721–8.CrossRefPubMed
15.
go back to reference Verma-Gandhu M, Verdu EF, Bercik P, Blennerhassett PA, Al-Mutawaly N, Ghia JE, Collins SM. Visceral pain perception is determined by the duration of colitis and associated neuropeptide expression in the mouse. Gut. 2007;56:358–64.CrossRefPubMedPubMedCentral Verma-Gandhu M, Verdu EF, Bercik P, Blennerhassett PA, Al-Mutawaly N, Ghia JE, Collins SM. Visceral pain perception is determined by the duration of colitis and associated neuropeptide expression in the mouse. Gut. 2007;56:358–64.CrossRefPubMedPubMedCentral
16.
go back to reference Verma-Gandhu M, Verdu EF, Cohen-Lyons D, Collins SM. Lymphocyte-mediated regulation of beta-endorphin in the myenteric plexus. Am J Physiol Gastrointest Liver Physiol. 2007;292:G344–8.CrossRefPubMed Verma-Gandhu M, Verdu EF, Cohen-Lyons D, Collins SM. Lymphocyte-mediated regulation of beta-endorphin in the myenteric plexus. Am J Physiol Gastrointest Liver Physiol. 2007;292:G344–8.CrossRefPubMed
17.
go back to reference Talmont F, Mouledous L, Boue J, Mollereau C, Dietrich G. Denatured G-protein coupled receptors as immunogens to generate highly specific antibodies. PLoS One. 2012;7:e46348.CrossRefPubMedPubMedCentral Talmont F, Mouledous L, Boue J, Mollereau C, Dietrich G. Denatured G-protein coupled receptors as immunogens to generate highly specific antibodies. PLoS One. 2012;7:e46348.CrossRefPubMedPubMedCentral
18.
go back to reference Mace G, Jaume M, Blanpied C, Stephan L, Coudert JD, Druet P,Dietrich G. Anti-mu-opioid-receptor IgG antibodies are commonly present in serum from healthy blood donors: evidence for a role in apoptotic immune cell death. Blood. 2002;100:3261–8.CrossRefPubMed Mace G, Jaume M, Blanpied C, Stephan L, Coudert JD, Druet P,Dietrich G. Anti-mu-opioid-receptor IgG antibodies are commonly present in serum from healthy blood donors: evidence for a role in apoptotic immune cell death. Blood. 2002;100:3261–8.CrossRefPubMed
19.
go back to reference Martin L, Auge C, Boue J, Buresi MC, Chapman K, Asfaha S, Andrade-Gordon P, Steinhoff M, Cenac N, Dietrich G, Vergnolle N. Thrombin receptor: an endogenous inhibitor of inflammatory pain, activating opioid pathways. Pain. 2009;146:121–9.CrossRefPubMed Martin L, Auge C, Boue J, Buresi MC, Chapman K, Asfaha S, Andrade-Gordon P, Steinhoff M, Cenac N, Dietrich G, Vergnolle N. Thrombin receptor: an endogenous inhibitor of inflammatory pain, activating opioid pathways. Pain. 2009;146:121–9.CrossRefPubMed
20.
go back to reference Benard A, Cavailles P, Boue J, Chapey E, Bayry J, Blanpied C, Meyer N, Lamant L, Kaveri SV, Brousset P, Dietrich G. mu-Opioid receptor is induced by IL-13 within lymph nodes from patients with Sezary syndrome. J Invest Dermatol. 2010;130:1337–44.CrossRefPubMed Benard A, Cavailles P, Boue J, Chapey E, Bayry J, Blanpied C, Meyer N, Lamant L, Kaveri SV, Brousset P, Dietrich G. mu-Opioid receptor is induced by IL-13 within lymph nodes from patients with Sezary syndrome. J Invest Dermatol. 2010;130:1337–44.CrossRefPubMed
21.
go back to reference Brunner E, Domhof S, Langer F. Nonparametric analysis of longitudinal data in factorial experiments: J. Wiley; 2002. Brunner E, Domhof S, Langer F. Nonparametric analysis of longitudinal data in factorial experiments: J. Wiley; 2002.
22.
go back to reference Brunner E, Puri M. Nonparametric methods in factorial designs. Stat Pap. 2001;42:1–52.CrossRef Brunner E, Puri M. Nonparametric methods in factorial designs. Stat Pap. 2001;42:1–52.CrossRef
23.
go back to reference Noguchi K, Gel YR, Brunner E, Konietschke F. nparLD: an R software package for the nonparametric analysis of longitudinal data in factorial experiments. J Stat Soft. 2012;50. Noguchi K, Gel YR, Brunner E, Konietschke F. nparLD: an R software package for the nonparametric analysis of longitudinal data in factorial experiments. J Stat Soft. 2012;50.
24.
go back to reference Basso L, Bourreille A, Dietrich G. Intestinal inflammation and pain management. Curr Opin Pharmacol. 2015;25:50–5.CrossRefPubMed Basso L, Bourreille A, Dietrich G. Intestinal inflammation and pain management. Curr Opin Pharmacol. 2015;25:50–5.CrossRefPubMed
25.
go back to reference Rittner HL, Labuz D, Richter JF, Brack A, Schafer M, Stein C, Mousa SA. CXCR1/2 ligands induce p38 MAPK-dependent translocation and release of opioid peptides from primary granules in vitro and in vivo. Brain Behav Immun. 2007;21:1021–32.CrossRefPubMed Rittner HL, Labuz D, Richter JF, Brack A, Schafer M, Stein C, Mousa SA. CXCR1/2 ligands induce p38 MAPK-dependent translocation and release of opioid peptides from primary granules in vitro and in vivo. Brain Behav Immun. 2007;21:1021–32.CrossRefPubMed
26.
go back to reference Basso L, Boue J, Bourreille A, Dietrich G. Endogenous regulation of inflammatory pain by T-cell-derived opioids: when friend turns to foe. Inflamm Bowel Dis. 2014;20:1870–7.CrossRefPubMed Basso L, Boue J, Bourreille A, Dietrich G. Endogenous regulation of inflammatory pain by T-cell-derived opioids: when friend turns to foe. Inflamm Bowel Dis. 2014;20:1870–7.CrossRefPubMed
27.
go back to reference Kearney ER, Pape KA, Loh DY, Jenkins MK. Visualization of peptide-specific T cell immunity and peripheral tolerance induction in vivo. Immunity. 1994;1:327–39.CrossRefPubMed Kearney ER, Pape KA, Loh DY, Jenkins MK. Visualization of peptide-specific T cell immunity and peripheral tolerance induction in vivo. Immunity. 1994;1:327–39.CrossRefPubMed
28.
go back to reference Benard A, Boue J, Chapey E, Jaume M, Gomes B, Dietrich G. Delta opioid receptors mediate chemotaxis in bone marrow-derived dendritic cells. J Neuroimmunol. 2008;197:21–8.CrossRefPubMed Benard A, Boue J, Chapey E, Jaume M, Gomes B, Dietrich G. Delta opioid receptors mediate chemotaxis in bone marrow-derived dendritic cells. J Neuroimmunol. 2008;197:21–8.CrossRefPubMed
29.
go back to reference Jaume M, Laffont S, Chapey E, Blanpied C, Dietrich G. Opioid receptor blockade increases the number of lymphocytes without altering T cell response in draining lymph nodes in vivo. J Neuroimmunol. 2007;188:95–102.CrossRefPubMed Jaume M, Laffont S, Chapey E, Blanpied C, Dietrich G. Opioid receptor blockade increases the number of lymphocytes without altering T cell response in draining lymph nodes in vivo. J Neuroimmunol. 2007;188:95–102.CrossRefPubMed
30.
go back to reference Philippe D, Dubuquoy L, Groux H, Brun V, Chuoi-Mariot MT, Gaveriaux-Ruff C, Colombel JF, Kieffer BL, Desreumaux P. Anti-inflammatory properties of the mu-opioid receptor support its use in the treatment of colon inflammation. J Clin Invest. 2003;111:1329–38.CrossRefPubMedPubMedCentral Philippe D, Dubuquoy L, Groux H, Brun V, Chuoi-Mariot MT, Gaveriaux-Ruff C, Colombel JF, Kieffer BL, Desreumaux P. Anti-inflammatory properties of the mu-opioid receptor support its use in the treatment of colon inflammation. J Clin Invest. 2003;111:1329–38.CrossRefPubMedPubMedCentral
31.
go back to reference Sacerdote P, Manfredi B, Gaspani L, Panerai AE. The opioid antagonist naloxone induces a shift from type 2 to type 1 cytokine pattern in BALB/cJ mice. Blood. 2000;95:2031–6.PubMed Sacerdote P, Manfredi B, Gaspani L, Panerai AE. The opioid antagonist naloxone induces a shift from type 2 to type 1 cytokine pattern in BALB/cJ mice. Blood. 2000;95:2031–6.PubMed
32.
go back to reference Hughes PA, Harrington AM, Castro J, Liebregts T, Adam B, Grasby DJ, Isaacs NJ, Maldeniya L, Martin CM, Persson J, Andrews JM, Holtmann G, Blackshaw LA, Brierley SM. Sensory neuro-immune interactions differ between irritable bowel syndrome subtypes. Gut. 2013;62:1456–65.CrossRefPubMed Hughes PA, Harrington AM, Castro J, Liebregts T, Adam B, Grasby DJ, Isaacs NJ, Maldeniya L, Martin CM, Persson J, Andrews JM, Holtmann G, Blackshaw LA, Brierley SM. Sensory neuro-immune interactions differ between irritable bowel syndrome subtypes. Gut. 2013;62:1456–65.CrossRefPubMed
33.
go back to reference Mousa SA, Straub RH, Schafer M, Stein C. Beta-endorphin, Met-enkephalin and corresponding opioid receptors within synovium of patients with joint trauma, osteoarthritis and rheumatoid arthritis. Ann Rheum Dis. 2007;66:871–9.CrossRefPubMedPubMedCentral Mousa SA, Straub RH, Schafer M, Stein C. Beta-endorphin, Met-enkephalin and corresponding opioid receptors within synovium of patients with joint trauma, osteoarthritis and rheumatoid arthritis. Ann Rheum Dis. 2007;66:871–9.CrossRefPubMedPubMedCentral
34.
go back to reference Busch-Dienstfertig M, Labuz D, Wolfram T, Vogel NN, Stein C. JAK-STAT1/3-induced expression of signal sequence-encoding proopiomelanocortin mRNA in lymphocytes reduces inflammatory pain in rats. Mol Pain. 2012;8:83.CrossRefPubMedPubMedCentral Busch-Dienstfertig M, Labuz D, Wolfram T, Vogel NN, Stein C. JAK-STAT1/3-induced expression of signal sequence-encoding proopiomelanocortin mRNA in lymphocytes reduces inflammatory pain in rats. Mol Pain. 2012;8:83.CrossRefPubMedPubMedCentral
35.
go back to reference Buzzetti R, McLoughlin L, Lavender PM, Clark AJL, Rees LH. Expression of pro-opiomelanocortin gene and quantification of adrenocorticotropic hormone-like immunoreactivity in human normal peripheral mononuclear cells and lymphoid and myeloid malignancies. J Clin Invest. 1989;83:733–7.CrossRefPubMedPubMedCentral Buzzetti R, McLoughlin L, Lavender PM, Clark AJL, Rees LH. Expression of pro-opiomelanocortin gene and quantification of adrenocorticotropic hormone-like immunoreactivity in human normal peripheral mononuclear cells and lymphoid and myeloid malignancies. J Clin Invest. 1989;83:733–7.CrossRefPubMedPubMedCentral
36.
go back to reference Sitte N, Busch M, Mousa SA, Labuz D, Rittner H, Gore C, Krause H, Stein C, Schafer M. Lymphocytes upregulate signal sequence-encoding proopiomelanocortin mRNA and beta-endorphin during painful inflammation in vivo. J Neuroimmunol. 2007;183:133–45.CrossRefPubMed Sitte N, Busch M, Mousa SA, Labuz D, Rittner H, Gore C, Krause H, Stein C, Schafer M. Lymphocytes upregulate signal sequence-encoding proopiomelanocortin mRNA and beta-endorphin during painful inflammation in vivo. J Neuroimmunol. 2007;183:133–45.CrossRefPubMed
37.
go back to reference Sobczak M, Salaga M, Storr MA, Fichna J. Physiology, signaling, and pharmacology of opioid receptors and their ligands in the gastrointestinal tract: current concepts and future perspectives. J Gastroenterol. 2014;49:24–45.CrossRefPubMedPubMedCentral Sobczak M, Salaga M, Storr MA, Fichna J. Physiology, signaling, and pharmacology of opioid receptors and their ligands in the gastrointestinal tract: current concepts and future perspectives. J Gastroenterol. 2014;49:24–45.CrossRefPubMedPubMedCentral
38.
go back to reference Stein C, Millan MJ, Shippenberg TS, Peter K, Herz A. Peripheral opioid receptors mediating antinociception in inflammation. Evidence for involvement of mu, delta and kappa receptors. J Pharmacol Exp Ther. 1989;248:1269–75.PubMed Stein C, Millan MJ, Shippenberg TS, Peter K, Herz A. Peripheral opioid receptors mediating antinociception in inflammation. Evidence for involvement of mu, delta and kappa receptors. J Pharmacol Exp Ther. 1989;248:1269–75.PubMed
39.
go back to reference Labuz D, Mousa SA, Schafer M, Stein C, Machelska H. Relative contribution of peripheral versus central opioid receptors to antinociception. Brain Res. 2007;1160:30–8.CrossRefPubMed Labuz D, Mousa SA, Schafer M, Stein C, Machelska H. Relative contribution of peripheral versus central opioid receptors to antinociception. Brain Res. 2007;1160:30–8.CrossRefPubMed
41.
42.
go back to reference Thompson GL, Canals M, Poole DP. Biological redundancy of endogenous GPCR ligands in the gut and the potential for endogenous functional selectivity. Front Pharmacol. 2014;5:262.CrossRefPubMedPubMedCentral Thompson GL, Canals M, Poole DP. Biological redundancy of endogenous GPCR ligands in the gut and the potential for endogenous functional selectivity. Front Pharmacol. 2014;5:262.CrossRefPubMedPubMedCentral
Metadata
Title
Endogenous analgesia mediated by CD4+ T lymphocytes is dependent on enkephalins in mice
Authors
Lilian Basso
Jérôme Boué
Karim Mahiddine
Catherine Blanpied
Sébastien Robiou-du-Pont
Nathalie Vergnolle
Céline Deraison
Gilles Dietrich
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2016
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-016-0591-x

Other articles of this Issue 1/2016

Journal of Neuroinflammation 1/2016 Go to the issue