A. Basbaum, D. Bautista, G. Scherrer, and J. D. , Cellular and Molecular Mechanisms of Pain, Cell, vol.139, issue.2, pp.267-284, 2009.
DOI : 10.1016/j.cell.2009.09.028

H. Zeilhofer, D. Benke, and G. Yévenes, Chronic Pain States: Pharmacological Strategies to Restore Diminished Inhibitory Spinal Pain Control, Annual Review of Pharmacology and Toxicology, vol.52, issue.1, pp.111-133, 2012.
DOI : 10.1146/annurev-pharmtox-010611-134636

C. Luo, T. Kuner, and R. Kuner, Synaptic plasticity in pathological pain, Trends in Neurosciences, vol.37, issue.6, pp.343-355, 2014.
DOI : 10.1016/j.tins.2014.04.002

J. Sandkühler, Models and Mechanisms of Hyperalgesia and Allodynia, Physiological Reviews, vol.89, issue.2, pp.707-758, 2009.
DOI : 10.1097/00000542-200003000-00037

J. Coull, Trans-synaptic shift in anion gradient in spinal lamina I neurons as a mechanism of neuropathic pain, Nature, vol.424, issue.6951, pp.938-942, 2003.
DOI : 10.1038/nature01868

R. Harvey, GlyR ??3: An Essential Target for Spinal PGE2-Mediated Inflammatory Pain Sensitization, Science, vol.304, issue.5672, pp.884-887, 2004.
DOI : 10.1126/science.1094925

URL : https://hal.archives-ouvertes.fr/hal-00142904

J. Lynch, Native glycine receptor subtypes and their physiological roles, Neuropharmacology, vol.56, issue.1, pp.303-309, 2009.
DOI : 10.1016/j.neuropharm.2008.07.034

G. Yévenes and H. Zeilhofer, Allosteric modulation of glycine receptors, British Journal of Pharmacology, vol.89, issue.Pt 2, pp.224-236, 2011.
DOI : 10.1152/jn.00614.2002

B. Laube, G. Maksay, R. Schemm, and H. Betz, Modulation of glycine receptor function: a novel approach for therapeutic intervention at inhibitory synapses?, Trends in Pharmacological Sciences, vol.23, issue.11, pp.519-527, 2002.
DOI : 10.1016/S0165-6147(02)02138-7

J. De-la-roche, 4-Chloropropofol enhances chloride currents in human hyperekplexic and artificial mutated glycine receptors, BMC Neurology, vol.15, issue.6, p.104, 2012.
DOI : 10.1097/00001756-199504190-00018

V. Eckle, 4-bromopropofol decreases action potential generation in spinal neurons by inducing a glycine receptor-mediated tonic conductance, British Journal of Pharmacology, vol.92, issue.Database Issue, pp.5790-5801, 2014.
DOI : 10.1152/physrev.00043.2010

W. Xiong, Cannabinoid potentiation of glycine receptors contributes to cannabis-induced analgesia, Nature Chemical Biology, vol.23, issue.5, pp.296-303, 2011.
DOI : 10.1016/S0006-3495(02)75166-7

W. Xiong, Cannabinoids suppress inflammatory and neuropathic pain by targeting ??3 glycine receptors, The Journal of Experimental Medicine, vol.31, issue.6, pp.1121-1134, 2012.
DOI : 10.1073/pnas.96.10.5780

URL : http://jem.rupress.org/content/jem/209/6/1121.full.pdf

Z. Yang, K. Aubrey, I. Alroy, R. Harvey, R. Vandenberg et al., Subunit-specific modulation of glycine receptors by cannabinoids and N-arachidonyl-glycine, Biochemical Pharmacology, vol.76, issue.8, pp.1014-1023, 2008.
DOI : 10.1016/j.bcp.2008.07.037

J. Zhang, N. Gong, J. Huang, L. Guo, and Y. Wang, Gelsemine, a principal alkaloid from Gelsemium sempervirens Ait., exhibits potent and specific antinociception in chronic pain by acting at spinal ??3 glycine receptors, Pain, vol.154, issue.11, pp.2452-2462, 2013.
DOI : 10.1016/j.pain.2013.07.027

J. Ahrens, 2,6 Di-tert-butylphenol, a Nonanesthetic Propofol Analog, Modulates ??1?? Glycine Receptor Function in a Manner Distinct from Propofol, Anesthesia & Analgesia, vol.99, issue.1, pp.91-96, 2004.
DOI : 10.1213/01.ANE.0000120083.10269.54

J. Ahrens, M. Leuwer, J. De-la-roche, N. Foadi, K. Krampfl et al., The Non-Anaesthetic Propofol Analogue 2,6-Di-tert-Butylphenol Fails to Modulate GABA<sub>A</sub> Receptor Function, Pharmacology, vol.83, issue.2, pp.95-98, 2009.
DOI : 10.1159/000180125

M. Krasowski, A. Jenkins, P. Flood, A. Kung, A. Hopfinger et al., General anesthetic potencies of a series of propofol analogs correlate with potency for potentiation of ?-aminobutyric acid (GABA) current at the GABA A receptor but not with lipid solubility, J Pharmacol Exp Ther, vol.297, issue.1, pp.338-351, 2001.

A. Marabelli, M. Moroni, R. Lape, and L. Sivilotti, The kinetic properties of the ??3 rat glycine receptor make it suitable for mediating fast synaptic inhibition, The Journal of Physiology, vol.VII, issue.13, pp.3289-3308, 2013.
DOI : 10.1002/pol.1951.120070506

J. Ahrens, A Transmembrane Residue Influences the Interaction of Propofol with the Strychnine-Sensitive Glycine ??1 and ??1?? Receptor, Anesthesia & Analgesia, vol.107, issue.6, pp.1875-1883, 2008.
DOI : 10.1213/ane.0b013e3181875a31

G. Moraga-cid, G. Yévenes, G. Schmalzing, R. Peoples, and L. Aguayo, A Single Phenylalanine Residue in the Main Intracellular Loop of ??1 ??-Aminobutyric Acid Type A and Glycine Receptors Influences Their Sensitivity to Propofol, Anesthesiology, vol.115, issue.3, pp.464-473, 2011.
DOI : 10.1097/ALN.0b013e31822550f7

T. Lynagh and B. Laube, Opposing Effects of the Anesthetic Propofol at Pentameric Ligand-Gated Ion Channels Mediated by a Common Site, Journal of Neuroscience, vol.34, issue.6, pp.2155-2159, 2014.
DOI : 10.1523/JNEUROSCI.4307-13.2014

S. Mihic, Sites of alcohol and volatile anaesthetic action on GABAA and glycine receptors, Nature, vol.268, issue.6649, pp.385-389, 1997.
DOI : 10.1016/0922-4106(94)90190-2

G. Findlay, Glycine receptor knock-in mice and hyperekplexia-like phenotypes: comparisons with the null mutant, J Neurosci, vol.23, issue.22, pp.8051-8059, 2003.

J. Carland, Characterization of the Effects of Charged Residues in the Intracellular Loop on Ion Permeation in ??1 Glycine Receptor Channels, Journal of Biological Chemistry, vol.34, issue.4, pp.2023-2030, 2009.
DOI : 10.1016/0079-6107(72)90005-3

K. Leuchowius, High Content Screening for Inhibitors of Protein Interactions and Post-translational Modifications in Primary Cells by Proximity Ligation, Molecular & Cellular Proteomics, vol.75, issue.1, pp.178-183, 2010.
DOI : 10.1038/nchembio0407-187

R. Hibbs and E. Gouaux, Principles of activation and permeation in an anion-selective Cys-loop receptor, Nature, vol.33, issue.7349, pp.54-60, 2011.
DOI : 10.1016/j.pep.2003.08.014

G. Moraga-cid, glycine receptor transmembrane structure, Proceedings of the National Academy of Sciences, vol.1838, issue.5, pp.2865-2870, 2015.
DOI : 10.1038/38738

URL : https://hal.archives-ouvertes.fr/hal-01230762

S. Zhao, Cell type???specific channelrhodopsin-2 transgenic mice for optogenetic dissection of neural circuitry function, Nature Methods, vol.305, issue.9, pp.745-752, 2011.
DOI : 10.1038/nature04053

E. Foster, Targeted Ablation, Silencing, and Activation Establish Glycinergic Dorsal Horn Neurons as Key Components of a Spinal Gate for Pain and Itch, Neuron, vol.85, issue.6, pp.1289-1304, 2015.
DOI : 10.1016/j.neuron.2015.02.028

E. Mitchell, L. Gentet, J. Dempster, and D. Belelli, GABA A and glycine receptor-mediated transmission in rat lamina II neurones: relevance to the analgesic actions of neuroactive steroids

A. Todd and R. Spike, The localization of classical transmitters and neuropeptides within neurons in laminae I???III of the mammalian spinal dorsal horn, Progress in Neurobiology, vol.41, issue.5, pp.609-645, 1993.
DOI : 10.1016/0301-0082(93)90045-T

S. Meller and G. Gebhart, Intraplantar zymosan as a reliable, quantifiable model of thermal and mechanical hyperalgesia in the rat, European Journal of Pain, vol.649, issue.1, pp.43-52, 1997.
DOI : 10.1016/0006-8993(94)91044-8

H. Reinold, Spinal inflammatory hyperalgesia is mediated by prostaglandin E receptors of the EP2 subtype, Journal of Clinical Investigation, vol.115, issue.3, pp.673-679, 2005.
DOI : 10.1172/JCI23618DS1

H. Schaible, S. Mcmahon, M. Koltzenburg, I. Tracey, and D. Turk, Joint pain: basic mechanisms Wall and Melzack's Textbook of Pain, pp.609-619

K. Hösl, H. Reinold, R. Harvey, U. Müller, S. Narumiya et al., Spinal prostaglandin E receptors of the EP2 subtype and the glycine receptor ?3 subunit, which mediate central inflammatory hyperalgesia, do not contribute to pain after peripheral nerve injury or formalin injection, Pain, vol.126, pp.1-346, 2006.

G. Tibbs, HCN1 Channels as Targets for Anesthetic and Nonanesthetic Propofol Analogs in the Amelioration of Mechanical and Thermal Hyperalgesia in a Mouse Model of Neuropathic Pain, Journal of Pharmacology and Experimental Therapeutics, vol.345, issue.3, pp.363-373, 2013.
DOI : 10.1124/jpet.113.203620

G. Bennett and Y. Xie, A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man, Pain, vol.33, issue.1, pp.87-107, 1988.
DOI : 10.1016/0304-3959(88)90209-6

A. Momin, H. Cadiou, A. Mason, and P. Mcnaughton, in somatosensory neurons, The Journal of Physiology, vol.42, issue.24, pp.5911-5929, 2008.
DOI : 10.1038/nn809

G. Ku, N. Doherty, L. Schmidt, R. Jackson, and R. Dinerstein, Ex vivo lipopolysaccharide-induced interleukin-1 secretion from murine peritoneal macrophages inhibited by probucol, a hypocholesterolemic agent with antioxidant properties., The FASEB Journal, vol.4, issue.6, pp.1645-1653, 1990.
DOI : 10.1096/fasebj.4.6.2318380

R. Shakir, A. Ariffin, and M. Abdulla, Synthesis of New 2,5-Di-substituted 1,3,4-Oxadiazoles Bearing 2,6-Di-tert-butylphenol Moieties and Evaluation of Their Antioxidant Activity, Molecules, vol.28, issue.3, pp.3436-3449, 2014.
DOI : 10.1016/S0023-6438(95)80008-5

M. Nelson, Molecular Mechanisms of Subtype-Specific Inhibition of Neuronal T-Type Calcium Channels by Ascorbate, Journal of Neuroscience, vol.27, issue.46, pp.12577-12583, 2007.
DOI : 10.1523/JNEUROSCI.2206-07.2007

W. Ralvenius, D. Benke, M. Acuña, U. Rudolph, and H. Zeilhofer, Analgesia and unwanted benzodiazepine effects in point-mutated mice expressing only one benzodiazepine-sensitive GABAA receptor subtype, Nature Communications, vol.16, issue.1, p.6803, 2015.
DOI : 10.1016/j.neuropharm.2006.03.020

J. Simon, H. Wakimoto, N. Fujita, M. Lalande, and E. Barnard, Analysis of the Set of GABAA Receptor Genes in the Human Genome, Journal of Biological Chemistry, vol.279, issue.40, pp.41422-41435, 2004.
DOI : 10.1074/jbc.M401354200

S. Haverkamp, U. Müller, K. Harvey, R. Harvey, H. Betz et al., Diversity of glycine receptors in the mouse retina: Localization of the ??3 subunit, Journal of Comparative Neurology, vol.38, issue.4, pp.524-539, 2003.
DOI : 10.1016/S0042-6989(97)00300-3

M. Malosio, B. Marqueze-pouey, J. Kuhse, and H. Betz, Widespread expression of glycine receptor subunit mRNAs in the adult and developing rat brain, EMBO J, vol.10, issue.9, pp.2401-2409, 1991.

W. Xiong, Presynaptic glycine receptors as a potential therapeutic target for hyperekplexia disease, Nature Neuroscience, vol.11, issue.2, pp.232-239, 2014.
DOI : 10.1523/JNEUROSCI.6347-11.2012

B. Hruskova, Differential Distribution of Glycine Receptor Subtypes at the Rat Calyx of Held Synapse, Journal of Neuroscience, vol.32, issue.47, pp.17012-17024, 2012.
DOI : 10.1523/JNEUROSCI.1547-12.2012

G. Yévenes and H. Zeilhofer, Molecular Sites for the Positive Allosteric Modulation of Glycine Receptors by Endocannabinoids, PLoS ONE, vol.131, issue.8, p.23886, 2011.
DOI : 10.1371/journal.pone.0023886.s003

C. Burgos, P. Castro, T. Mariqueo, M. Bunster, L. Guzman et al., Evidence for ??-Helices in the Large Intracellular Domain Mediating Modulation of the ??1-Glycine Receptor by Ethanol and G????, Journal of Pharmacology and Experimental Therapeutics, vol.352, issue.1, pp.148-155, 2015.
DOI : 10.1124/jpet.114.217976

D. Xu and Y. Zhang, Ab initio protein structure assembly using continuous structure fragments and optimized knowledge-based force field, Proteins: Structure, Function, and Bioinformatics, vol.19, issue.7, pp.1715-1735, 2012.
DOI : 10.1016/j.str.2011.05.004

URL : http://europepmc.org/articles/pmc3370074?pdf=render

N. Eswar, Comparative protein structure modeling using Modeller, Curr Protoc Bioinformatics, vol.5, 2006.

G. Hassaine, X-ray structure of the mouse serotonin 5-HT3 receptor, Nature, vol.57, issue.7514, pp.276-281, 2014.
DOI : 10.1085/jgp.200709857

URL : https://hal.archives-ouvertes.fr/hal-01102530

J. Irwin, T. Sterling, M. Mysinger, E. Bolstad, and R. Coleman, ZINC: A Free Tool to Discover Chemistry for Biology, Journal of Chemical Information and Modeling, vol.52, issue.7, pp.1757-1768, 2012.
DOI : 10.1021/ci3001277

R. Friesner, Extra Precision Glide:?? Docking and Scoring Incorporating a Model of Hydrophobic Enclosure for Protein???Ligand Complexes, Journal of Medicinal Chemistry, vol.49, issue.21, pp.6177-6196, 2006.
DOI : 10.1021/jm051256o

D. Mowrey, Open-Channel Structures of the Human Glycine Receptor ??1 Full-Length Transmembrane Domain, Structure, vol.21, issue.10, pp.1897-1904, 2013.
DOI : 10.1016/j.str.2013.07.014

K. Zemoura and D. Benke, C Terminus with the Proteasomal ATPase Rtp6 and Regulated by Neuronal Activity, Journal of Biological Chemistry, vol.21, issue.11, pp.7738-7746, 2014.
DOI : 10.1016/j.cell.2010.01.024

R. James and J. Glen, Synthesis, biological evaluation, and preliminary structure-activity considerations of a series of alkylphenols as intravenous anesthetic agents, Journal of Medicinal Chemistry, vol.23, issue.12, pp.1350-1357, 1980.
DOI : 10.1021/jm00186a013

E. Bonetti, Benzodiazepine antagonist Ro 15-1788: Neurological and behavioral effects, Psychopharmacology, vol.78, issue.1, pp.8-18, 1982.
DOI : 10.1007/BF00470579