E. Denarier, M. Aguezzoul, C. Jolly, C. Vourc-'h, A. Roure et al., Genomic Structure and Chromosomal Mapping of the Mouse STOP Gene (Mtap6), Biochemical and Biophysical Research Communications, vol.243, issue.3, pp.791-796, 1998.
DOI : 10.1006/bbrc.1998.8179

E. Denarier, A. Fourest-lieuvin, C. Bosc, F. Pirollet, A. Chapel et al., Nonneuronal isoforms of STOP protein are responsible for microtubule cold stability in mammalian fibroblasts, Proceedings of the National Academy of Sciences, vol.95, issue.11, pp.6055-6060, 1998.
DOI : 10.1073/pnas.95.11.6055

M. Aguezzoul, A. Andrieux, and E. Denarier, Overlap of promoter and coding sequences in the mouse STOP gene (Mtap6)???, Genomics, vol.81, issue.6, pp.623-627, 2003.
DOI : 10.1016/S0888-7543(03)00053-3

P. Baas and S. Heidemann, Microtubule reassembly from nucleating fragments during the regrowth of amputated neurites, The Journal of Cell Biology, vol.103, issue.3, pp.917-927, 1986.
DOI : 10.1083/jcb.103.3.917

L. Guillaud, C. Bosc, A. Fourest-lieuvin, E. Denarier, F. Pirollet et al., STOP Proteins are Responsible for the High Degree of Microtubule Stabilization Observed in Neuronal Cells, The Journal of Cell Biology, vol.15, issue.1, pp.167-179, 1998.
DOI : 10.1083/jcb.133.1.151

B. Schaar and S. Mcconnell, Cytoskeletal coordination during neuronal migration, Proceedings of the National Academy of Sciences, vol.102, issue.38, pp.13652-13657, 2005.
DOI : 10.1073/pnas.0506008102

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1199551

L. Tsai and J. Gleeson, Nucleokinesis in Neuronal Migration, Neuron, vol.46, issue.3, pp.383-388, 2005.
DOI : 10.1016/j.neuron.2005.04.013

J. Baratier, L. Peris, J. Brocard, S. Gory-faure, F. Dufour et al., Phosphorylation of Microtubule-associated Protein STOP by Calmodulin Kinase II, Journal of Biological Chemistry, vol.281, issue.28, pp.19561-19569, 2006.
DOI : 10.1074/jbc.M509602200

URL : https://hal.archives-ouvertes.fr/hal-00100341

S. Gory-faure, V. Windscheid, C. Bosc, L. Peris, D. Proietto et al., STOP-like Protein 21 Is a Novel Member of the STOP Family, Revealing a Golgi Localization of STOP Proteins, Journal of Biological Chemistry, vol.281, issue.38, pp.28387-28396, 2006.
DOI : 10.1074/jbc.M603380200

URL : https://hal.archives-ouvertes.fr/inserm-00380095

A. Andrieux, P. Salin, M. Vernet, P. Kujala, J. Baratier et al., The suppression of brain cold-stable microtubules in mice induces synaptic defects associated with neuroleptic-sensitive behavioral disorders, Genes & Development, vol.16, issue.18, pp.2350-2364, 2002.
DOI : 10.1101/gad.223302

P. Brun, M. Begou, A. Andrieux, L. Mouly-badina, M. Clerget et al., Dopaminergic transmission in STOP null mice, Journal of Neurochemistry, vol.65, issue.1, pp.63-73, 2005.
DOI : 10.1016/0165-0173(90)90015-G

URL : https://hal.archives-ouvertes.fr/inserm-00380189

M. Begou, J. Volle, J. Bertrand, P. Brun, J. D. Schweitzer et al., The stop null mice model for schizophrenia displays cognitive and social deficits partly alleviated by neuroleptics, Neuroscience, vol.157, issue.1, pp.29-39, 2008.
DOI : 10.1016/j.neuroscience.2008.07.080

URL : https://hal.archives-ouvertes.fr/inserm-00375621

R. Fradley, O. Meara, G. Newman, R. Andrieux, A. Job et al., STOP knockout and NMDA NR1 hypomorphic mice exhibit deficits in sensorimotor gating, Behavioural Brain Research, vol.163, issue.2, pp.257-264, 2005.
DOI : 10.1016/j.bbr.2005.05.012

URL : https://hal.archives-ouvertes.fr/inserm-00380197

M. Zervas, T. Opitz, W. Edelmann, B. Wainer, R. Kucherlapati et al., Impaired hippocampal long-term potentiation in microtubule-associated protein 1B-deficient mice, Journal of Neuroscience Research, vol.34, issue.1, pp.83-92, 2005.
DOI : 10.1002/jnr.20624

S. Eastwood, L. Lyon, L. George, A. Andrieux, D. Job et al., Altered expression of synaptic protein mRNAs in STOP (MAP6) mutant mice, Journal of Psychopharmacology, vol.105, issue.6, pp.635-644, 2007.
DOI : 10.1177/0269881106068825

URL : https://hal.archives-ouvertes.fr/inserm-00380032

J. Charlton, P. Allen, K. Psifogeorgou, S. Chakravarty, I. Gomes et al., Multiple Actions of Spinophilin Regulate Mu Opioid Receptor Function, Neuron, vol.58, issue.2, pp.238-247, 2008.
DOI : 10.1016/j.neuron.2008.02.006

G. Stefano, Y. Goumon, F. Casares, P. Cadet, G. Fricchione et al., Endogenous morphine, Trends in Neurosciences, vol.23, issue.9, pp.436-442, 2000.
DOI : 10.1016/S0166-2236(00)01611-8

R. Herbert, H. Venter, and S. Pos, Do mammals make their own morphine? Nat Prod Rep, pp.317-322, 2000.
DOI : 10.1039/a809409h

Y. Goumon, A. Laux, A. Muller, and D. Aunis, Central and Peripheral Endogenous Morphine. An R Acad Nac Farm, pp.389-418, 2009.

E. Glattard, I. Welters, T. Lavaux, A. Muller, A. Laux et al., Endogenous Morphine Levels Are Increased in Sepsis: A Partial Implication of Neutrophils, PLoS ONE, vol.5, issue.1, p.8791, 2010.
DOI : 10.1371/journal.pone.0008791.t001

Y. Goumon, A. Muller, E. Glattard, C. Marban, C. Gasnier et al., Identification of Morphine-6-glucuronide in Chromaffin Cell Secretory Granules, Journal of Biological Chemistry, vol.281, issue.12, pp.8082-8089, 2006.
DOI : 10.1074/jbc.M502298200

URL : https://hal.archives-ouvertes.fr/hal-00092673

A. Muller, E. Glattard, O. Taleb, V. Kemmel, A. Laux et al., Endogenous Morphine in SH-SY5Y Cells and the Mouse Cerebellum, PLoS ONE, vol.391, issue.2, 2008.
DOI : 10.1371/journal.pone.0001641.g007

E. Hazum, J. Sabatka, K. Chang, D. Brent, J. Findlay et al., Morphine in cow and human milk: could dietary morphine constitute a ligand for specific morphine (mu) receptors?, Science, vol.213, issue.4511, pp.1010-1012, 1981.
DOI : 10.1126/science.6267691

A. Goldstein, R. Barrett, I. James, L. Lowney, C. Weitz et al., Morphine and other opiates from beef brain and adrenal., Proceedings of the National Academy of Sciences, vol.82, issue.15, pp.5203-5207, 1985.
DOI : 10.1073/pnas.82.15.5203

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC390528

Y. Goumon, S. Bouret, F. Casares, W. Zhu, J. Beauvillain et al., Lipopolysaccharide increases endogenous morphine levels in rat brain, Neuroscience Letters, vol.293, issue.2, pp.135-138, 2000.
DOI : 10.1016/S0304-3940(00)01507-X

A. Gintzler, M. Gershon, and S. Spector, A nonpeptide morphine-like compound: immunocytochemical localization in the mouse brain, Science, vol.199, issue.4327, pp.447-448, 1978.
DOI : 10.1126/science.339350

M. Guarna, E. Bianchi, A. Bartolini, C. Ghelardini, N. Galeotti et al., Endogenous morphine modulates acute thermonociception in mice, Journal of Neurochemistry, vol.80, issue.2, pp.271-277, 2002.
DOI : 10.1046/j.0022-3042.2001.00708.x

E. Bianchi, M. Guarna, and A. Tagliamonte, Immunocytochemical localization of endogenous codeine and morphine, Advances in Neuroimmunology, vol.4, issue.2, pp.83-92, 1994.
DOI : 10.1016/S0960-5428(05)80003-8

W. Zhu, Y. Ma, A. Bell, T. Esch, M. Guarna et al., Presence of morphine in rat amygdala: evidence for the mu3 opiate receptor subtype via nitric oxide release in limbic structures, Med Sci Monit, vol.10, pp.433-439, 2004.

E. Bianchi, C. Alessandrini, M. Guarna, and A. Tagliamonte, Endogenous codeine and morphine are stored in specific brain neurons, Brain Research, vol.627, issue.2, pp.210-215, 1993.
DOI : 10.1016/0006-8993(93)90323-F

C. Boettcher, M. Fellermeier, C. Boettcher, B. Drager, and M. Zenk, How human neuroblastoma cells make morphine, Proceedings of the National Academy of Sciences, vol.102, issue.24, pp.8495-8500, 2005.
DOI : 10.1073/pnas.0503244102

R. Kream and G. Stefano, De novo biosynthesis of morphine in animal cells: An evidence-based model, Med Sci Monit, vol.12, pp.207-219, 2006.

C. Poeaknapo, J. Schmidt, M. Brandsch, B. Drager, and M. Zenk, Endogenous formation of morphine in human cells, Proceedings of the National Academy of Sciences, vol.101, issue.39, pp.14091-14096, 2004.
DOI : 10.1073/pnas.0405430101

J. Lotsch and G. Geisslinger, Morphine-6-Glucuronide, Clinical Pharmacokinetics, vol.95, issue.16, pp.485-499, 2001.
DOI : 10.2165/00003088-200140070-00001

P. Cadet, K. Mantione, and G. Stefano, Molecular Identification and Functional Expression of ??3, a Novel Alternatively Spliced Variant of the Human ?? Opiate Receptor Gene, The Journal of Immunology, vol.170, issue.10, pp.5118-5123, 2003.
DOI : 10.4049/jimmunol.170.10.5118

P. Cadet, K. Mantione, W. Zhu, R. Kream, M. Sheehan et al., A Functionally Coupled ??3-Like Opiate Receptor/Nitric Oxide Regulatory Pathway in Human Multi-Lineage Progenitor Cells, The Journal of Immunology, vol.179, issue.9, pp.5839-5844, 2007.
DOI : 10.4049/jimmunol.179.9.5839

A. Aloisi, C. Aurilio, V. Bachiocco, G. Biasi, P. Fiorenzani et al., Endocrine consequences of opioid therapy, Psychoneuroendocrinology, vol.34, issue.1, pp.34-162, 2009.
DOI : 10.1016/j.psyneuen.2009.05.013

J. Loeser and R. Treede, The Kyoto protocol of IASP Basic Pain Terminology???, Pain, vol.137, issue.3, pp.473-477, 2008.
DOI : 10.1016/j.pain.2008.04.025

G. Doyle, X. Sheng, S. Lin, D. Press, D. Grice et al., Identification of five mouse ??-opioid receptor (MOR) gene (Oprm1) splice variants containing a newly identified alternatively spliced exon, Gene, vol.395, issue.1-2, pp.98-107, 2007.
DOI : 10.1016/j.gene.2007.02.004

P. Skyers, S. Einheber, J. Pierce, and T. Milner, Increased ??-opioid receptor labeling is found on inner molecular layer terminals of the dentate gyrus following seizures??????Supported by NIH Grants DA08259 (T.M.), NS41490 (J.P.), and HL18974 and a grant from the Epilepsy Foundation (S.E.). P.S. Skyers was supported, in part, from the Avon Summer Research Program., Experimental Neurology, vol.179, issue.2, pp.200-209, 2003.
DOI : 10.1016/S0014-4886(02)00018-3

T. Sargeant, D. Day, E. Mrkusich, D. Foo, and J. Miller, Mu opioid receptors are expressed on radial glia but not migrating neuroblasts in the late embryonic mouse brain, Brain Research, vol.1175, pp.28-38, 2007.
DOI : 10.1016/j.brainres.2007.07.091

Y. Zhang, Y. Pan, Y. Kolesnikov, and G. Pasternak, Immunohistochemical labeling of the mu opioid receptor carboxy terminal splice variant mMOR-1B4 in the mouse central nervous system, Brain Research, vol.1099, issue.1, pp.33-43, 2006.
DOI : 10.1016/j.brainres.2006.04.133

M. Pawar, P. Kumar, S. Sunkaraneni, S. Sirohi, E. Walker et al., Opioid agonist efficacy predicts the magnitude of tolerance and the regulation of ??-opioid receptors and dynamin-2, European Journal of Pharmacology, vol.563, issue.1-3, pp.92-101, 2007.
DOI : 10.1016/j.ejphar.2007.01.059

L. Brady, M. Herkenham, J. Long, and R. Rothman, Chronic morphine increases ??-opiate receptor binding in rat brain: a quantitative autoradiographic study, Brain Research, vol.477, issue.1-2, pp.382-386, 1989.
DOI : 10.1016/0006-8993(89)91432-7

N. Bergasa, R. Rothman, E. Mukerjee, J. Vergalla, and E. Jones, Up-regulation of central mu???opioid receptors in a model of hepatic encephalopathy: A potential mechanism for increased sensitivity to morphine in liver failure, Life Sciences, vol.70, issue.14, pp.1701-1708, 2002.
DOI : 10.1016/S0024-3205(02)01487-X

M. Guarna, C. Ghelardini, N. Galeotti, A. Bartolini, L. Noli et al., Effects of endogenous morphine deprivation on memory retention of passive avoidance learning in mice, The International Journal of Neuropsychopharmacology, vol.7, issue.3, pp.311-319, 2004.
DOI : 10.1017/S1461145704004341

E. Van-dorp, B. Kest, W. Kowalczyk, A. Morariu, A. Waxman et al., Morphine-6??-glucuronide Rapidly Increases Pain Sensitivity Independently of Opioid Receptor Activity in Mice and Humans, Anesthesiology, vol.110, issue.6, pp.1356-1363, 2009.
DOI : 10.1097/ALN.0b013e3181a105de

D. Liang, X. Shi, Y. Qiao, M. Angst, D. Yeomans et al., Chronic Morphine Administration Enhances Nociceptive Sensitivity and Local Cytokine Production after Incision, Molecular Pain, vol.87, issue.4, p.7, 2008.
DOI : 10.1186/1744-8069-4-7

X. Li, M. Angst, and J. Clark, Opioid-Induced Hyperalgesia and Incisional Pain, Anesthesia and Analgesia, vol.93, pp.204-209, 2001.
DOI : 10.1097/00000539-200107000-00040

S. Crain and K. Shen, Opioids can evoke direct receptor-mediated excitatory effects on sensory neurons, Trends in Pharmacological Sciences, vol.11, issue.2, pp.77-81, 1990.
DOI : 10.1016/0165-6147(90)90322-Y

S. Crain and K. Shen, Ultra-low concentrations of naloxone selectively antagonize excitatory effects of morphine on sensory neurons, thereby increasing its antinociceptive potency and attenuating tolerance/dependence during chronic cotreatment., Proceedings of the National Academy of Sciences, vol.92, issue.23, pp.10540-10544, 1995.
DOI : 10.1073/pnas.92.23.10540

K. Powell, N. Abul-husn, A. Jhamandas, M. Olmstead, R. Beninger et al., Paradoxical Effects of the Opioid Antagonist Naltrexone on Morphine Analgesia, Tolerance, and Reward in Rats, Journal of Pharmacology and Experimental Therapeutics, vol.300, issue.2, pp.588-596, 2002.
DOI : 10.1124/jpet.300.2.588

S. Chaplan, F. Bach, J. Pogrel, J. Chung, and T. Yaksh, Quantitative assessment of tactile allodynia in the rat paw, Journal of Neuroscience Methods, vol.53, issue.1, pp.55-63, 1994.
DOI : 10.1016/0165-0270(94)90144-9

I. Yalcin, A. Charlet, M. Freund-mercier, M. Barrot, and P. Poisbeau, Differentiating Thermal Allodynia and Hyperalgesia Using Dynamic Hot and Cold Plate in Rodents, The Journal of Pain, vol.10, issue.7, pp.767-773, 2009.
DOI : 10.1016/j.jpain.2009.01.325

URL : https://hal.archives-ouvertes.fr/hal-00399549

G. Hedou, S. Chasserot-golaz, V. Kemmel, S. Gobaille, G. Roussel et al., Immunohistochemical studies of the localization of neurons containing the enzyme that synthesizes dopamine, GABA, or ?-hydroxybutyrate in the rat substantia nigra and striatum, The Journal of Comparative Neurology, vol.105, issue.4, pp.549-560, 2000.
DOI : 10.1002/1096-9861(20001030)426:4<549::AID-CNE4>3.0.CO;2-Y

Y. Goumon, T. Angelone, F. Schoentgen, S. Chasserot-golaz, B. Almas et al., The Hippocampal Cholinergic Neurostimulating Peptide, the N-terminal Fragment of the Secreted Phosphatidylethanolamine-binding Protein, Possesses a New Biological Activity on Cardiac Physiology, Journal of Biological Chemistry, vol.279, issue.13, pp.13054-13064, 2004.
DOI : 10.1074/jbc.M308533200

URL : https://hal.archives-ouvertes.fr/hal-00113048

R. Coles, M. Kushnir, G. Nelson, G. Mcmillin, and F. Urry, Simultaneous Determination of Codeine, Morphine, Hydrocodone, Hydromorphone, Oxycodone, and 6-Acetylmorphine in Urine, Serum, Plasma, Whole Blood, and Meconium by LC-MS-MS, Journal of Analytical Toxicology, vol.31, issue.1, pp.1-14, 2007.
DOI : 10.1093/jat/31.1.1