J. Boue, C. Blanpied, P. Brousset, N. Vergnolle, and G. Dietrich, Endogenous Opioid-Mediated Analgesia Is Dependent on Adaptive T Cell Response in Mice, The Journal of Immunology, vol.186, issue.9, pp.5078-84, 2011.
DOI : 10.4049/jimmunol.1003335

P. Cabot, L. Carter, C. Gaiddon, Q. Zhang, M. Schäfer et al., Immune cell-derived beta-endorphin. Production, release, and control of inflammatory pain in rats., Journal of Clinical Investigation, vol.100, issue.1, pp.142-150, 1997.
DOI : 10.1172/JCI119506

H. Machelska, P. Cabot, S. Mousa, Q. Zhang, and C. Stein, Pain control in inflammation governed by selectins, Nature Medicine, vol.15, issue.12, pp.1425-1433, 1998.
DOI : 10.1016/0014-4886(88)90133-1

H. Machelska, J. Schopohl, S. Mousa, D. Labuz, M. Schafer et al., Different mechanisms of intrinsic pain inhibition in early and late inflammation, Journal of Neuroimmunology, vol.141, issue.1-2, pp.30-39, 2003.
DOI : 10.1016/S0165-5728(03)00213-3

H. Rittner, A. Brack, H. Machelska, S. Mousa, M. Bauer et al., Opioid Peptide???expressing Leukocytes, Anesthesiology, vol.95, issue.2, pp.500-508, 2001.
DOI : 10.1097/00000542-200108000-00036

J. Boue, C. Blanpied, M. Djata-cabral, L. Pelletier, N. Vergnolle et al., Immune conditions associated with CD4+ T effector-induced opioid release and analgesia, Pain, vol.153, issue.2, pp.485-93, 2012.
DOI : 10.1016/j.pain.2011.11.013

C. Gaveriaux-ruff, L. Karchewski, X. Hever, A. Matifas, and B. Kieffer, Inflammatory pain is enhanced in delta opioid receptor-knockout mice, European Journal of Neuroscience, vol.177, issue.10, pp.2558-67, 2008.
DOI : 10.1111/j.1460-9568.2008.06223.x

URL : https://hal.archives-ouvertes.fr/inserm-00311101

C. Gaveriaux-ruff, C. Nozaki, X. Nadal, X. Hever, R. Weibel et al., Genetic ablation of delta opioid receptors in nociceptive sensory neurons increases chronic pain and abolishes opioid analgesia, Pain, vol.152, issue.6, pp.1238-1286, 2011.
DOI : 10.1016/j.pain.2010.12.031

R. Weibel, D. Reiss, L. Karchewski, O. Gardon, A. Matifas et al., Mu Opioid Receptors on Primary Afferent Nav1.8 Neurons Contribute to Opiate-Induced Analgesia: Insight from Conditional Knockout Mice, PLoS ONE, vol.122, issue.9, p.74706, 2013.
DOI : 10.1371/journal.pone.0074706.g010

J. Boue, L. Basso, N. Cenac, C. Blanpied, M. Rolli-derkinderen et al., Endogenous Regulation of Visceral Pain via Production of Opioids by Colitogenic CD4+ T Cells in Mice, Gastroenterology, vol.146, issue.1, pp.166-75, 2014.
DOI : 10.1053/j.gastro.2013.09.020

D. Labuz, Y. Schmidt, A. Schreiter, H. Rittner, S. Mousa et al., Immune cell-derived opioids protect against neuropathic pain in mice, J Clin Invest, vol.119, pp.278-86, 2009.

D. Labuz, A. Schreiter, Y. Schmidt, A. Brack, and H. Machelska, T lymphocytes containing ??-endorphin ameliorate mechanical hypersensitivity following nerve injury, Brain, Behavior, and Immunity, vol.24, issue.7, pp.1045-53, 2010.
DOI : 10.1016/j.bbi.2010.04.001

E. Valdez-morales, R. Guerrero-alba, F. Ochoa-cortes, J. Benson, I. Spreadbury et al., Release of endogenous opioids during a chronic IBD model suppresses the excitability of colonic DRG neurons, Neurogastroenterology & Motility, vol.17, issue.1, pp.39-46, 2013.
DOI : 10.1111/nmo.12008

M. Verma-gandhu, P. Bercik, Y. Motomura, E. Verdu, W. Khan et al., CD4+ T-Cell Modulation of Visceral Nociception in Mice, Gastroenterology, vol.130, issue.6, pp.1721-1729, 2006.
DOI : 10.1053/j.gastro.2006.01.045

M. Verma-gandhu, E. Verdu, P. Bercik, P. Blennerhassett, N. Mutawaly et al., Visceral pain perception is determined by the duration of colitis and associated neuropeptide expression in the mouse, Gut, vol.56, issue.3, pp.358-64, 2007.
DOI : 10.1136/gut.2006.100016

M. Verma-gandhu, E. Verdu, D. Cohen-lyons, and S. Collins, Lymphocyte-mediated regulation of beta-endorphin in the myenteric plexus, AJP: Gastrointestinal and Liver Physiology, vol.292, issue.1, pp.344-352, 2007.
DOI : 10.1152/ajpgi.00318.2006

F. Talmont, L. Mouledous, J. Boue, C. Mollereau, and G. Dietrich, Denatured G-Protein Coupled Receptors as Immunogens to Generate Highly Specific Antibodies, PLoS ONE, vol.7, issue.9, p.46348, 2012.
DOI : 10.1371/journal.pone.0046348.t001

G. Mace, M. Jaume, C. Blanpied, L. Stephan, J. Coudert et al., Anti-micro-opioid-receptor IgG antibodies are commonly present in serum from healthy blood donors: evidence for a role in apoptotic immune cell death, Blood, vol.100, issue.9, pp.3261-3269, 2002.
DOI : 10.1182/blood-2002-01-0055

L. Martin, C. Auge, J. Boue, M. Buresi, K. Chapman et al., Thrombin receptor: An endogenous inhibitor of inflammatory pain, activating opioid pathways, Pain, vol.146, issue.1, pp.121-130, 2009.
DOI : 10.1016/j.pain.2009.07.016

A. Benard, P. Cavailles, J. Boue, E. Chapey, J. Bayry et al., ??-Opioid Receptor Is Induced by IL-13 within Lymph Nodes from Patients with S??zary Syndrome, Journal of Investigative Dermatology, vol.130, issue.5, pp.1337-1381, 2010.
DOI : 10.1038/jid.2009.433

E. Brunner, S. Domhof, and F. Langer, Nonparametric analysis of longitudinal data in factorial experiments, 2002.

E. Brunner and M. Puri, Nonparametric methods in factorial designs, Statistical Papers, vol.85, issue.1, pp.1-52, 2001.
DOI : 10.1007/s003620000039

K. Noguchi, Y. Gel, E. Brunner, and F. Konietschke, Software Package for the Nonparametric Analysis of Longitudinal Data in Factorial Experiments, Journal of Statistical Software, vol.50, issue.12, 2012.
DOI : 10.18637/jss.v050.i12

L. Basso, A. Bourreille, and G. Dietrich, Intestinal inflammation and pain management, Current Opinion in Pharmacology, vol.25, pp.50-55, 2015.
DOI : 10.1016/j.coph.2015.11.004

H. Rittner, D. Labuz, J. Richter, A. Brack, M. Schafer et al., CXCR1/2 ligands induce p38 MAPK-dependent translocation and release of opioid peptides from primary granules in vitro and in vivo, Brain, Behavior, and Immunity, vol.21, issue.8, pp.1021-1053, 2007.
DOI : 10.1016/j.bbi.2007.05.002

L. Basso, J. Boue, A. Bourreille, and G. Dietrich, Endogenous Regulation of Inflammatory Pain by T-cell-derived Opioids, Inflammatory Bowel Diseases, vol.20, issue.10, pp.1870-1877, 2014.
DOI : 10.1097/MIB.0000000000000073

E. Kearney, K. Pape, D. Loh, and M. Jenkins, Visualization of peptide-specific T cell immunity and peripheral tolerance induction in vivo, Immunity, vol.1, issue.4, pp.327-366, 1994.
DOI : 10.1016/1074-7613(94)90084-1

A. Benard, J. Boue, E. Chapey, M. Jaume, B. Gomes et al., Delta opioid receptors mediate chemotaxis in bone marrow-derived dendritic cells, Journal of Neuroimmunology, vol.197, issue.1, pp.21-29, 2008.
DOI : 10.1016/j.jneuroim.2008.03.020

M. Jaume, S. Laffont, E. Chapey, C. Blanpied, and G. Dietrich, Opioid receptor blockade increases the number of lymphocytes without altering T cell response in draining lymph nodes in vivo, Journal of Neuroimmunology, vol.188, issue.1-2, pp.95-102, 2007.
DOI : 10.1016/j.jneuroim.2007.06.013

D. Philippe, L. Dubuquoy, H. Groux, V. Brun, M. Chuoi-mariot et al., Anti-inflammatory properties of the ?? opioid receptor support its use in the treatment of colon inflammation, Journal of Clinical Investigation, vol.111, issue.9, pp.1329-1367, 2003.
DOI : 10.1172/JCI200316750

P. Sacerdote, B. Manfredi, L. Gaspani, and A. Panerai, The opioid antagonist naloxone induces a shift from type 2 to type 1 cytokine pattern in BALB/cJ mice, Blood, vol.95, pp.2031-2037, 2000.

P. Hughes, A. Harrington, J. Castro, T. Liebregts, B. Adam et al., Sensory neuro-immune interactions differ between Irritable Bowel Syndrome subtypes, Gut, vol.18, issue.Suppl 1, pp.1456-65, 2013.
DOI : 10.1136/gutjnl-2011-301856

S. Mousa, R. Straub, M. Schafer, and C. Stein, ??-Endorphin, Met-enkephalin and corresponding opioid receptors within synovium of patients with joint trauma, osteoarthritis and rheumatoid arthritis, Annals of the Rheumatic Diseases, vol.66, issue.7, pp.871-880, 2007.
DOI : 10.1136/ard.2006.067066

M. Busch-dienstfertig, D. Labuz, T. Wolfram, N. Vogel, and C. Stein, JAK-STAT1/3-Induced Expression of Signal Sequence-Encoding Proopiomelanocortin mRNA in Lymphocytes Reduces Inflammatory Pain in Rats, Molecular Pain, vol.87, p.83, 2012.
DOI : 10.1038/227680a0

R. Buzzetti, L. Mcloughlin, P. Lavender, A. Clark, and L. Rees, Expression of pro-opiomelanocortin gene and quantification of adrenocorticotropic hormone-like immunoreactivity in human normal peripheral mononuclear cells and lymphoid and myeloid malignancies., Journal of Clinical Investigation, vol.83, issue.2, pp.733-740, 1989.
DOI : 10.1172/JCI113940

N. Sitte, M. Busch, S. Mousa, D. Labuz, H. Rittner et al., Lymphocytes upregulate signal sequence-encoding proopiomelanocortin mRNA and beta-endorphin during painful inflammation in vivo, Journal of Neuroimmunology, vol.183, issue.1-2, pp.133-178, 2007.
DOI : 10.1016/j.jneuroim.2006.11.033

M. Sobczak, M. Salaga, M. Storr, and J. Fichna, Physiology, signaling, and pharmacology of opioid receptors and their ligands in the gastrointestinal tract: current concepts and future perspectives, Journal of Gastroenterology, vol.70, issue.2, pp.24-45, 2014.
DOI : 10.1007/s00535-013-0753-x

C. Stein, M. Millan, T. Shippenberg, K. Peter, and A. Herz, Peripheral opioid receptors mediating antinociception in inflammation. Evidence for involvement of mu, delta and kappa receptors, J Pharmacol Exp Ther, vol.248, pp.1269-75, 1989.

D. Labuz, S. Mousa, M. Schafer, C. Stein, and H. Machelska, Relative contribution of peripheral versus central opioid receptors to antinociception, Brain Research, vol.1160, pp.30-38, 2007.
DOI : 10.1016/j.brainres.2007.05.049

C. Stein, Targeting pain and inflammation by peripherally acting opioids, Frontiers in Pharmacology, vol.4, p.123, 2013.
DOI : 10.3389/fphar.2013.00123

C. Stein, Opioids, sensory systems and chronic pain, European Journal of Pharmacology, vol.716, issue.1-3, pp.179-87, 2013.
DOI : 10.1016/j.ejphar.2013.01.076

G. Thompson, M. Canals, and D. Poole, Biological redundancy of endogenous GPCR ligands in the gut and the potential for endogenous functional selectivity, Frontiers in Pharmacology, vol.35, issue.2, p.262, 2014.
DOI : 10.1016/j.it.2014.02.004