A. Aiba, C. Chen, K. Herrup, C. Rosenmund, C. Stevens et al., Reduced hippocampal long-term potentiation and context-specific deficit in associative learning in mGluR1 mutant mice, Cell, vol.79, issue.2, pp.365-75, 1994.
DOI : 10.1016/0092-8674(94)90204-6

B. Alstermark and C. Ekerot, The lateral reticular nucleus: a precerebellar centre providing the cerebellum with overview and integration of motor functions at systems level. A new hypothesis, The Journal of Physiology, vol.411, issue.Suppl. 608, pp.5453-5461, 2013.
DOI : 10.1113/jphysiol.2013.256669

L. Ambrose, S. Gallagher, E. Unterwald, and E. Van-bockstaele, Dopamine-D1 and ??-opioid receptors co-exist in rat striatal neurons, Neuroscience Letters, vol.399, issue.3, pp.191-197, 2006.
DOI : 10.1016/j.neulet.2006.02.027

P. Arlotta, B. Molyneaux, D. Jabaudon, Y. Yoshida, and J. Macklis, Ctip2 Controls the Differentiation of Medium Spiny Neurons and the Establishment of the Cellular Architecture of the Striatum, Journal of Neuroscience, vol.28, issue.3, pp.622-654, 2008.
DOI : 10.1523/JNEUROSCI.2986-07.2008

R. Aupperle and M. Paulus, Neural systems underlying approach and avoidance in anxiety disorders, Dialogues Clin Neurosci, vol.12, pp.517-548, 2010.

B. Baldo and A. Kelley, Discrete neurochemical coding of distinguishable motivational processes: insights from nucleus accumbens control of feeding, Psychopharmacology, vol.43, issue.3, pp.439-59, 2007.
DOI : 10.1007/s00213-007-0741-z

D. Bannerman, B. Niewoehner, L. Lyon, C. Romberg, W. Schmitt et al., NMDA Receptor Subunit NR2A Is Required for Rapidly Acquired Spatial Working Memory But Not Incremental Spatial Reference Memory, Journal of Neuroscience, vol.28, issue.14, pp.3623-3653, 2008.
DOI : 10.1523/JNEUROSCI.3639-07.2008

URL : http://hdl.handle.net/11858/00-001M-0000-0019-98B9-B

D. Bannerman, B. Yee, M. Good, M. Heupel, S. Iversen et al., Double dissociation of function within the hippocampus: A comparison of dorsal, ventral, and complete hippocampal cytotoxic lesions., Behavioral Neuroscience, vol.113, issue.6, pp.1170-88, 1999.
DOI : 10.1037/0735-7044.113.6.1170

J. Becker, D. Clesse, C. Spiegelhalter, Y. Schwab, L. Merrer et al., Autistic-Like Syndrome in Mu Opioid Receptor Null Mice is Relieved by Facilitated mGluR4 Activity, Neuropsychopharmacology, vol.14, issue.9, pp.2049-60, 2014.
DOI : 10.1038/nature11208

F. Berrendero, A. Plaza-zabala, L. Galeote, A. Flores, S. Bura et al., Influence of ??-Opioid Receptors in the Behavioral Effects of Nicotine, Neuropsychopharmacology, vol.292, issue.10, pp.2332-2376, 2012.
DOI : 10.1038/npp.2012.88

J. Bertran-gonzalez, V. Laurent, B. Chieng, M. Christie, and B. Balleine, Learning-Related Translocation of ??-Opioid Receptors on Ventral Striatal Cholinergic Interneurons Mediates Choice between Goal-Directed Actions, Journal of Neuroscience, vol.33, issue.41, pp.16060-71, 2013.
DOI : 10.1523/JNEUROSCI.1927-13.2013

R. Bodnar, Endogenous opioids and feeding behavior: a 30-year historical perspective, Peptides, vol.25, issue.4, pp.697-725, 2004.
DOI : 10.1016/j.peptides.2004.01.006

R. Bodnar, N. Lamonte, Y. Israel, Y. Kandov, T. Ackerman et al., Reciprocal opioid???opioid interactions between the ventral tegmental area and nucleus accumbens regions in mediating ?? agonist-induced feeding in rats, Peptides, vol.26, issue.4, pp.621-630, 2005.
DOI : 10.1016/j.peptides.2004.11.007

C. Bramham, N. Milgram, and B. Srebro, ?? Opioid receptor activation is required to induce LTP of synaptic transmission in the lateral perforant path in vivo, Brain Research, vol.567, issue.1, pp.42-50, 1991.
DOI : 10.1016/0006-8993(91)91433-2

B. Buzas, J. Rosenberger, and B. Cox, Ca2+/Calmodulin-Dependent Transcriptional Activation of ??-Opioid Receptor Gene Expression Induced by Membrane Depolarization in NG108-15 Cells, Journal of Neurochemistry, vol.70, issue.1, pp.105-117, 1998.
DOI : 10.1046/j.1471-4159.1998.70010105.x

P. Calabresi, B. Picconi, A. Tozzi, V. Ghiglieri, D. Filippo et al., Direct and indirect pathways of basal ganglia: a critical reappraisal, Nature Neuroscience, vol.86, issue.8, pp.1022-1052, 2014.
DOI : 10.1038/466449a

A. Castane, D. Theobald, and T. Robbins, Selective lesions of the dorsomedial striatum impair serial spatial reversal learning in rats, Behavioural Brain Research, vol.210, issue.1, pp.74-83, 2010.
DOI : 10.1016/j.bbr.2010.02.017

B. Cen, Q. Yu, J. Guo, Y. Wu, K. Ling et al., Direct binding of ??-arrestins to two distinct intracellular domains of the ?? opioid receptor, Journal of Neurochemistry, vol.190, issue.6, pp.1887-94, 2001.
DOI : 10.1046/j.1471-4159.2001.00204.x

C. Chavkin, W. Shoemaker, J. Mcginty, A. Bayon, and F. Bloom, Characterization of the prodynorphin and proenkephalin neuropeptide systems in rat hippocampus, The Journal of neuroscience : the official journal of the Society for Neuroscience, vol.5, pp.808-824, 1985.

V. Chefer and T. Shippenberg, Augmentation of Morphine-Induced Sensitization but Reduction in Morphine Tolerance and Reward in Delta-Opioid Receptor Knockout Mice, Neuropsychopharmacology, vol.24, issue.4, pp.887-98, 2009.
DOI : 10.1038/npp.2008.128

S. Childers, Opioid receptor-coupled second messenger systems, Life Sciences, vol.48, issue.21, pp.1991-2003, 1991.
DOI : 10.1016/0024-3205(91)90154-4

C. Chung, P. Keyworth, H. Martin-garcia, E. Charbogne, P. Darcq et al., A Novel Anxiogenic Role for the Delta Opioid Receptor Expressed in GABAergic Forebrain Neurons, Biological Psychiatry, vol.77, issue.4, pp.404-419, 2015.
DOI : 10.1016/j.biopsych.2014.07.033

C. Chung, P. Kieffer, and B. , Delta opioid receptors in brain function and diseases, Pharmacology & Therapeutics, vol.140, issue.1, pp.112-132, 2013.
DOI : 10.1016/j.pharmthera.2013.06.003

A. Ciamei and A. Morton, Progressive imbalance in the interaction between spatial and procedural memory systems in the R6/2 mouse model of Huntington???s disease, Neurobiology of Learning and Memory, vol.92, issue.3, pp.417-445, 2009.
DOI : 10.1016/j.nlm.2009.06.002

R. Ciccocioppo, R. Martin-fardon, and F. Weiss, Effect of Selective Blockade of ??1 or ?? Opioid Receptors on Reinstatement of Alcohol-Seeking Behavior by Drug-Associated Stimuli in Rats,, Neuropsychopharmacology, vol.27, issue.3, pp.391-400, 2002.
DOI : 10.1016/S0893-133X(02)00302-0

L. Corbit and B. Balleine, Learning and Motivational Processes Contributing to Pavlovian???Instrumental Transfer and Their Neural Bases: Dopamine and Beyond, Curr Top Behav Neurosci, 2015.
DOI : 10.1007/7854_2015_388

B. Crain, K. Chang, and J. Mcnamara, Quantitative autoradiographic analysis of Mu and delta opioid binding sites in the rat hippocampal formation, The Journal of Comparative Neurology, vol.205, issue.2, pp.170-80, 1986.
DOI : 10.1002/cne.902460203

C. Cunningham, P. Groblewski, and C. Voorhees, Place conditioning Animal models of addiction (Neuromethods), pp.167-190, 2011.

M. Dang, F. Yokoi, H. Yin, D. Lovinger, Y. Wang et al., Disrupted motor learning and long-term synaptic plasticity in mice lacking NMDAR1 in the striatum, Proceedings of the National Academy of Sciences, vol.103, issue.41, pp.15254-15263, 2006.
DOI : 10.1073/pnas.0601758103

V. David, A. Matifas, S. Gavello-baudy, L. Decorte, B. Kieffer et al., Brain Regional Fos Expression Elicited by the Activation of ??- but not ??-Opioid Receptors of the Ventral Tegmental Area: Evidence for an Implication of the Ventral Thalamus in Opiate Reward, Neuropsychopharmacology, vol.92, issue.7, pp.1746-59, 2008.
DOI : 10.1038/sj.npp.1301529

URL : https://hal.archives-ouvertes.fr/hal-00282970

D. Leonibus, E. Pascucci, T. Lopez, S. Oliverio, A. Amalric et al., Spatial deficits in a mouse model of Parkinson disease, Psychopharmacology, vol.142, issue.2, pp.517-542, 2007.
DOI : 10.1007/s00213-007-0862-4

URL : https://hal.archives-ouvertes.fr/hal-01384868

A. Deipolyi, S. Fang, J. Palop, G. Yu, X. Wang et al., Altered navigational strategy use and visuospatial deficits in hAPP transgenic mice, Neurobiology of Aging, vol.29, issue.2, pp.253-66, 2008.
DOI : 10.1016/j.neurobiolaging.2006.10.021

B. Dickerson and H. Eichenbaum, The Episodic Memory System: Neurocircuitry and Disorders, Neuropsychopharmacology, vol.6, issue.1, pp.86-104, 2010.
DOI : 10.1126/science.1077775

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2882963

Y. Dikshtein, R. Barnea, N. Kronfeld, E. Lax, I. Roth-deri et al., ??-Endorphin via the Delta Opioid Receptor is a Major Factor in the Incubation of Cocaine Craving, Neuropsychopharmacology, vol.286, issue.12, pp.2508-2522, 2013.
DOI : 10.1016/j.neulet.2007.11.041

J. Ding, J. Guzman, J. Peterson, J. Goldberg, and D. Surmeier, Thalamic Gating of Corticostriatal Signaling by Cholinergic Interneurons, Neuron, vol.67, issue.2, pp.294-307, 2010.
DOI : 10.1016/j.neuron.2010.06.017

J. Do, J. Kim, J. Bakes, K. Lee, and B. Kaang, Functional roles of neurotransmitters and neuromodulators in the dorsal striatum, Learning & Memory, vol.20, issue.1, pp.21-29, 2012.
DOI : 10.1101/lm.025015.111

P. Durieux, B. Bearzatto, S. Guiducci, T. Buch, A. Waisman et al., D2R striatopallidal neurons inhibit both locomotor and drug reward processes, Nature Neuroscience, vol.47, issue.4, pp.393-398, 2009.
DOI : 10.1038/nn.2286

URL : https://dipot.ulb.ac.be/dspace/bitstream/2013/50721/1/NatureNeuro2009DTR.pdf

P. Durieux, S. Schiffmann, and . De-kerchove, Differential regulation of motor control and response to dopaminergic drugs by D1R and D2R neurons in distinct dorsal striatum subregions, The EMBO Journal, vol.12, issue.3, pp.640-53
DOI : 10.1038/emboj.2011.400

A. Ennaceur, One-trial object recognition in rats and mice: Methodological and theoretical issues, Behavioural Brain Research, vol.215, issue.2, pp.244-54, 2010.
DOI : 10.1016/j.bbr.2009.12.036

A. Ennaceur, N. Neave, and J. Aggleton, Spontaneous object recognition and object location memory in rats: the effects of lesions in the cingulate cortices, the medial prefrontal cortex, the cingulum bundle and the fornix, Experimental Brain Research, vol.113, issue.3, pp.509-528, 1997.
DOI : 10.1007/PL00005603

E. Erbs, L. Faget, G. Scherrer, A. Matifas, D. Filliol et al., A mu???delta opioid receptor brain atlas reveals neuronal co-occurrence in subcortical networks, Brain Structure and Function, vol.72, issue.4, pp.677-702
DOI : 10.1007/s00429-014-0717-9

L. Faget, E. Erbs, L. Merrer, J. Scherrer, G. Matifas et al., In Vivo Visualization of Delta Opioid Receptors upon Physiological Activation Uncovers a Distinct Internalization Profile, Journal of Neuroscience, vol.32, issue.21, pp.7301-7311, 2012.
DOI : 10.1523/JNEUROSCI.0185-12.2012

G. Fenalti, P. Giguere, V. Katritch, X. Huang, A. Thompson et al., Molecular control of ??-opioid receptor signalling, Nature, vol.492, issue.7487, pp.191-197, 2014.
DOI : 10.1038/nature12944

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3931418

D. Filliol, S. Ghozland, J. Chluba, M. Martin, H. Matthes et al., Mice deficient for delta-and mu-opioid receptors exhibit opposing alterations of emotional responses, Nat Genet, vol.25, pp.195-200, 2000.

E. Gazyakan, M. Hennegriff, A. Haaf, G. Landwehrmeyer, T. Feuerstein et al., Characterization of opioid receptor types modulating acetylcholine release in septal regions of the rat brain, Naunyn-Schmiedeberg's Archives of Pharmacology, vol.362, issue.1, pp.32-40, 2000.
DOI : 10.1007/s002100000253

Z. Georgoussi, L. Leontiadis, G. Mazarakou, M. Merkouris, K. Hyde et al., Selective interactions between G protein subunits and RGS4 with the C-terminal domains of the ??- and ??-opioid receptors regulate opioid receptor signaling, Cellular Signalling, vol.18, issue.6, pp.771-82, 2006.
DOI : 10.1016/j.cellsig.2005.07.003

A. Ghate, K. Befort, J. Becker, D. Filliol, C. Bole-feysot et al., Identification of novel striatal genes by expression profiling in adult mouse brain, Neuroscience, vol.146, issue.3, pp.1182-92, 2007.
DOI : 10.1016/j.neuroscience.2007.02.040

URL : https://hal.archives-ouvertes.fr/hal-00189988

V. Ghiglieri, C. Sgobio, C. Costa, B. Picconi, and P. Calabresi, Striatum???hippocampus balance: From physiological behavior to interneuronal pathology, Progress in Neurobiology, vol.94, issue.2, pp.102-116, 2011.
DOI : 10.1016/j.pneurobio.2011.04.005

S. Ghozland, H. Matthes, F. Simonin, D. Filliol, B. Kieffer et al., Motivational effects of cannabinoids are mediated by mu-opioid and kappa-opioid receptors, The Journal of neuroscience : the official journal of the Society for Neuroscience, vol.22, pp.1146-54, 2002.

A. Giralt, A. Saavedra, O. Carreton, H. Arumi, S. Tyebji et al., PDE10 inhibition increases GluA1 and CREB phosphorylation and improves spatial and recognition memories in a Huntington's disease mouse model, Hippocampus, vol.164, issue.8, pp.684-95, 2013.
DOI : 10.1002/hipo.22128

J. Gomeza, L. Zhang, E. Kostenis, C. Felder, F. Bymaster et al., Enhancement of D1 dopamine receptor-mediated locomotor stimulation in M4 muscarinic acetylcholine receptor knockout mice, Proceedings of the National Academy of Sciences, vol.96, issue.18, pp.10483-10491, 1999.
DOI : 10.1073/pnas.96.18.10483

G. Govindaiah, Y. Wang, and C. Cox, Substance P selectively modulates GABAA receptor-mediated synaptic transmission in striatal cholinergic interneurons, Neuropharmacology, vol.58, issue.2, pp.413-435, 2010.
DOI : 10.1016/j.neuropharm.2009.09.011

M. Guo, L. Mao, and J. Wang, ???????????????????????????M4 ???????????????????????????, Neuroscience Bulletin, vol.325, issue.6, pp.469-73, 2010.
DOI : 10.1007/s12264-010-0933-0

J. Gutierrez-cuesta, A. Burokas, S. Mancino, S. Kummer, E. Martin-garcia et al., Effects of Genetic Deletion of Endogenous Opioid System Components on the Reinstatement of Cocaine-Seeking Behavior in Mice, Neuropsychopharmacology, vol.285, issue.1, pp.2974-88, 2014.
DOI : 10.1038/npp.2014.149

G. Hart, B. Leung, and B. Balleine, Dorsal and ventral streams: The distinct role of striatal subregions in the acquisition and performance of goal-directed actions, Neurobiology of Learning and Memory, vol.108, pp.104-122, 2014.
DOI : 10.1016/j.nlm.2013.11.003

M. Hasselmo and M. Sarter, Modes and Models of Forebrain Cholinergic Neuromodulation of Cognition, Neuropsychopharmacology, vol.18, issue.1, pp.52-73, 2011.
DOI : 10.1002/syn.20354

M. Heiman, A. Schaefer, S. Gong, J. Peterson, M. Day et al., A Translational Profiling Approach for the Molecular Characterization of CNS Cell Types, Cell, vol.135, issue.4, pp.738-786, 2008.
DOI : 10.1016/j.cell.2008.10.028

B. Henry, S. Fox, A. Crossman, and J. Brotchie, ??- and ??-Opioid Receptor Antagonists Reduce Levodopa-Induced Dyskinesia in the MPTP-Lesioned Primate Model of Parkinson's Disease, Experimental Neurology, vol.171, issue.1, pp.139-185, 2001.
DOI : 10.1006/exnr.2001.7727

C. Hopkins, C. Lindsley, and C. Niswender, mGluR4-positive allosteric modulation as potential treatment for Parkinson???s disease, Future Medicinal Chemistry, vol.1, issue.3, pp.501-514, 2009.
DOI : 10.4155/fmc.09.38

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2790174

D. Hutcheson, H. Matthes, E. Valjent, P. Sanchez-blazquez, M. Rodriguez-diaz et al., Lack of dependence and rewarding effects of deltorphin II in mu-opioid receptor-deficient mice, European Journal of Neuroscience, vol.24, issue.1, pp.153-61, 2001.
DOI : 10.1016/0196-9781(96)00073-3

R. Ilyutchenok and N. Dubrovina, Memory retrieval enhancement by kappa opioid agonist and mu, delta antagonists, Pharmacology Biochemistry and Behavior, vol.52, issue.4, pp.683-690, 1995.
DOI : 10.1016/0091-3057(95)00099-I

T. Inui and T. Shimura, Delta-opioid receptor blockade in the ventral pallidum increases perceived palatability and consumption of saccharin solution in rats, Behavioural Brain Research, vol.269, pp.20-27, 2014.
DOI : 10.1016/j.bbr.2014.04.005

T. Jacobson, B. Gruenbaum, and E. Markus, Extensive training and hippocampus or striatum lesions: Effect on place and response strategies, Physiology & Behavior, vol.105, issue.3, pp.645-52, 2012.
DOI : 10.1016/j.physbeh.2011.09.027

Z. Jiang and R. North, Pre-and postsynaptic inhibition by opioids in rat striatum, The Journal of neuroscience : the official journal of the Society for Neuroscience, vol.12, pp.356-61, 1992.

J. Jo, G. Son, B. Winters, M. Kim, D. Whitcomb et al., Muscarinic receptors induce LTD of NMDAR EPSCs via a mechanism involving hippocalcin, AP2 and PSD-95, Nature Neuroscience, vol.296, issue.10, pp.2-95, 2010.
DOI : 10.1038/nn.2636

URL : https://hal.archives-ouvertes.fr/hal-00578277

E. Jocoy, V. Andre, D. Cummings, S. Rao, N. Wu et al., Dissecting the contribution of individual receptor subunits to the enhancement of N-methyl-daspartate currents by dopamine D1 receptor activation in striatum, Front Syst Neurosci, vol.5, p.28, 2011.

E. Jutkiewicz, S. Kaminsky, K. Rice, J. Traynor, and J. Woods, Differential Behavioral Tolerance to the ??-Opioid Agonist SNC80 ([(+)-4-[(??R)-??-[(2S,5R)-2,5-Dimethyl-4-(2-propenyl)-1-piperazinyl]-(3-methoxyphenyl)methyl]-N,N-diethylbenzamide) in Sprague-Dawley Rats, Journal of Pharmacology and Experimental Therapeutics, vol.315, issue.1, pp.414-436, 2005.
DOI : 10.1124/jpet.105.088831

E. Jutkiewicz, K. Rice, J. Woods, and P. Winsauer, Effects of the delta-opioid receptor agonist SNC80 on learning relative to its antidepressant-like effects in rats, Behavioural Pharmacology, vol.14, issue.7, pp.509-525, 2003.
DOI : 10.1097/00008877-200311000-00003

Y. Katsuura and S. Taha, Mu opioid receptor antagonism in the nucleus accumbens shell blocks consumption of a preferred sucrose solution in an anticipatory contrast paradigm, Neuroscience, vol.261, pp.144-52, 2014.
DOI : 10.1016/j.neuroscience.2013.12.004

T. Kenakin, Functional Selectivity and Biased Receptor Signaling, Journal of Pharmacology and Experimental Therapeutics, vol.336, issue.2, pp.296-302, 2011.
DOI : 10.1124/jpet.110.173948

E. Khaimova, Y. Kandov, Y. Israel, G. Cataldo, M. Hadjimarkou et al., Opioid receptor subtype antagonists differentially alter GABA agonist-induced feeding elicited from either the nucleus accumbens shell or ventral tegmental area regions in rats, Brain Research, vol.1026, issue.2, pp.284-94, 2004.
DOI : 10.1016/j.brainres.2004.08.032

T. Kitamura, M. Pignatelli, J. Suh, K. Kohara, A. Yoshiki et al., Island Cells Control Temporal Association Memory, Science, vol.343, issue.6173, pp.896-901, 2014.
DOI : 10.1126/science.1244634

I. Kitchen, S. Slowe, H. Matthes, and B. Kieffer, Quantitative autoradiographic mapping of ??-, ??- and ??-opioid receptors in knockout mice lacking the ??-opioid receptor gene, Brain Research, vol.778, issue.1, pp.73-88, 1997.
DOI : 10.1016/S0006-8993(97)00988-8

P. Klenowski, M. Morgan, and S. Bartlett, The role of ??-opioid receptors in learning and memory underlying the development of addiction, British Journal of Pharmacology, vol.117, issue.1 Pt 2, pp.297-310, 2015.
DOI : 10.1111/bph.12618

J. Ko, U. Arvidsson, F. Williams, P. Law, R. Elde et al., Visualization of time-dependent redistribution of ??-opioid receptors in neuronal cells during prolonged agonist exposure, Molecular Brain Research, vol.69, issue.2, pp.171-85, 1999.
DOI : 10.1016/S0169-328X(99)00094-7

T. Korotkova, E. Fuchs, A. Ponomarenko, J. Von-engelhardt, and H. Monyer, NMDA Receptor Ablation on Parvalbumin-Positive Interneurons Impairs Hippocampal Synchrony, Spatial??Representations, and Working Memory, Neuron, vol.68, issue.3, pp.557-69, 2010.
DOI : 10.1016/j.neuron.2010.09.017

URL : http://doi.org/10.1016/j.neuron.2010.09.017

A. Kovoor, V. Nappey, B. Kieffer, and C. Chavkin, ?? and ?? Opioid Receptors Are Differentially Desensitized by the Coexpression of ??-Adrenergic Receptor Kinase 2 and ??-Arrestin 2 in Xenopus Oocytes, Journal of Biological Chemistry, vol.272, issue.44, pp.27605-27616, 1997.
DOI : 10.1074/jbc.272.44.27605

V. Laurent, J. Bertran-gonzalez, B. Chieng, and B. Balleine, ??-Opioid and Dopaminergic Processes in Accumbens Shell Modulate the Cholinergic Control of Predictive Learning and Choice, Journal of Neuroscience, vol.34, issue.4, pp.1358-69, 2014.
DOI : 10.1523/JNEUROSCI.4592-13.2014

V. Laurent, B. Leung, N. Maidment, and B. Balleine, ??- and ??-Opioid-Related Processes in the Accumbens Core and Shell Differentially Mediate the Influence of Reward-Guided and Stimulus-Guided Decisions on Choice, Journal of Neuroscience, vol.32, issue.5, pp.1875-83, 2012.
DOI : 10.1523/JNEUROSCI.4688-11.2012

V. Laurent, A. Morse, and B. Balleine, The role of opioid processes in reward and decision-making, British Journal of Pharmacology, vol.73, issue.Database Issue, pp.449-59, 2015.
DOI : 10.1111/bph.12818

L. Bourdonnec, B. Windh, R. Ajello, C. Leister, L. Gu et al., -Diethyl-4-(5-hydroxyspiro[chromene-2,4???-piperidine]-4-yl)benzamide (ADL5859), Journal of Medicinal Chemistry, vol.51, issue.19, pp.5893-5899, 2008.
DOI : 10.1021/jm8008986

URL : https://hal.archives-ouvertes.fr/hal-01408431

L. Bourdonnec, B. Windh, R. Leister, L. Zhou, Q. Ajello et al., -Diethyl-3-hydroxy-4-(spiro[chromene-2,4???-piperidine]-4-yl) Benzamide (ADL5747), Journal of Medicinal Chemistry, vol.52, issue.18, pp.5685-702, 2009.
DOI : 10.1021/jm900773n

URL : https://hal.archives-ouvertes.fr/hal-01408431

L. Merrer, J. Becker, J. Befort, K. Kieffer, and B. , Reward Processing by the Opioid System in the Brain, Physiological Reviews, vol.89, issue.4, pp.1379-412, 2009.
DOI : 10.1152/physrev.00005.2009

URL : https://hal.archives-ouvertes.fr/inserm-00438654

L. Merrer, J. Faget, L. Matifas, A. Kieffer, and B. , Cues predicting drug or food reward restore morphineinduced place conditioning in mice lacking delta opioid receptors, Psychopharmacology, 2012.

L. Merrer, J. Plaza-zabala, A. , D. Boca, C. Matifas et al., Deletion of the ?? Opioid Receptor Gene Impairs Place Conditioning But Preserves Morphine Reinforcement, Biological Psychiatry, vol.69, issue.7, pp.700-703, 2011.
DOI : 10.1016/j.biopsych.2010.10.021

L. Merrer, J. Rezai, X. Scherrer, G. Becker, J. Kieffer et al., Impaired Hippocampus-Dependent and Facilitated Striatum-Dependent Behaviors in Mice Lacking the Delta Opioid Receptor, Neuropsychopharmacology, vol.12, issue.6, 2013.
DOI : 10.1038/npp.2013.1

L. Moine, C. Kieffer, B. Gaveriaux-ruff, C. Befort, K. Bloch et al., Delta-opioid receptor gene expression in the mouse forebrain: Localization in cholinergic neurons of the striatum, Neuroscience, vol.62, issue.3, pp.635-675, 1994.
DOI : 10.1016/0306-4522(94)90464-2

A. Levine, M. Grace, P. Portoghese, and C. Billington, The effect of selective opioid antagonists on butorphanol-induced feeding, Brain Research, vol.637, issue.1-2, pp.242-250, 1994.
DOI : 10.1016/0006-8993(94)91239-4

M. Lobo, Y. Cui, S. Ostlund, B. Balleine, and X. Yang, Genetic control of instrumental conditioning by striatopallidal neuron???specific S1P receptor Gpr6, Nature Neuroscience, vol.29, issue.11, pp.1395-1402, 2007.
DOI : 10.1038/nn1987

S. Logue, S. Grauer, J. Paulsen, R. Graf, N. Taylor et al., The orphan GPCR, GPR88, modulates function of the striatal dopamine system: A possible therapeutic target for psychiatric disorders?, Molecular and Cellular Neuroscience, vol.42, issue.4, pp.438-485, 2009.
DOI : 10.1016/j.mcn.2009.09.007

R. Longoni, C. Cadoni, A. Mulas, D. Chiara, G. Spina et al., Dopamine-dependent behavioural stimulation by non-peptide delta opioids BW373U86 and SNC 80: 2. Place-preference and brain microdialysis studies in rats, Behav Pharmacol, vol.9, pp.9-14, 1998.

D. Lovinger, Neurotransmitter roles in synaptic modulation, plasticity and learning in the dorsal striatum, Neuropharmacology, vol.58, issue.7, pp.951-61, 2010.
DOI : 10.1016/j.neuropharm.2010.01.008

A. Luo, P. Tahsili-fahadan, R. Wise, C. Lupica, and G. Aston-jones, Linking Context with Reward: A Functional Circuit from Hippocampal CA3 to Ventral Tegmental Area, Science, vol.333, issue.6040, pp.353-360, 2011.
DOI : 10.1126/science.1204622

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3150711

C. Lupica, Delta and mu enkephalins inhibit spontaneous GABA-mediated IPSCs via a cyclic AMPindependent mechanism in the rat hippocampus, The Journal of neuroscience : the official journal of the Society for Neuroscience, vol.15, pp.737-786, 1995.

O. Mabrouk, M. Marti, S. Salvadori, and M. Morari, The novel delta opioid receptor agonist UFP-512 dually modulates motor activity in hemiparkinsonian rats via control of the nigro-thalamic pathway, Neuroscience, vol.164, issue.2, pp.360-369, 2009.
DOI : 10.1016/j.neuroscience.2009.08.058

O. Mabrouk, R. Viaro, M. Volta, A. Ledonne, N. Mercuri et al., Stimulation of ?? Opioid Receptor and Blockade of Nociceptin/Orphanin FQ Receptor Synergistically Attenuate Parkinsonism, Journal of Neuroscience, vol.34, issue.39, pp.12953-62, 2014.
DOI : 10.1523/JNEUROSCI.4677-13.2014

A. Mansour, C. Fox, H. Akil, and S. Watson, Opioid-receptor mRNA expression in the rat CNS: anatomical and functional implications, Trends in Neurosciences, vol.18, issue.1, pp.22-31, 1995.
DOI : 10.1016/0166-2236(95)93946-U

A. Mansour, C. Fox, S. Burke, F. Meng, R. Thompson et al., Mu, delta, and kappa opioid receptor mRNA expression in the rat CNS: An in situ hybridization study, The Journal of Comparative Neurology, vol.7, issue.3, pp.412-450, 1994.
DOI : 10.1002/cne.903500307

A. Mansour, R. Thompson, H. Akil, and S. Watson, Delta opioid receptor mRNA distribution in the brain: Comparison to delta receptor binding and proenkephalin mRNA, Journal of Chemical Neuroanatomy, vol.6, issue.6, pp.351-62, 1993.
DOI : 10.1016/0891-0618(93)90010-2

P. Marinelli, D. Funk, S. Harding, Z. Li, W. Juzytsch et al., Roles of opioid receptor subtypes in mediating alcohol-seeking induced by discrete cues and context, European Journal of Neuroscience, vol.167, issue.4, pp.671-679, 2009.
DOI : 10.1111/j.1460-9568.2009.06851.x

T. Martin, S. Kim, D. Cannon, G. Sizemore, D. Bian et al., Antagonism of delta(2)- opioid receptors by naltrindole-5'-isothiocyanate attenuates heroin self-administration but not antinociception in rats, J Pharmacol Exp Ther, vol.294, pp.975-82, 2000.

J. Martinez, J. Olson, K. Hilston, and C. , Opposite effects of met-enkephalin and leu-enkephalin on a discriminated shock-escape task., Behavioral Neuroscience, vol.98, issue.3, pp.487-95, 1984.
DOI : 10.1037/0735-7044.98.3.487

R. Massart, J. Guilloux, V. Mignon, P. Sokoloff, and J. Diaz, Striatal GPR88 expression is confined to the whole projection neuron population and is regulated by dopaminergic and glutamatergic afferents, European Journal of Neuroscience, vol.12, issue.Suppl. 1, pp.397-414, 2009.
DOI : 10.1111/j.1460-9568.2009.06842.x

S. Mccormick and A. Stoessl, Blockade of nigral and pallidal opioid receptors suppresses vacuous chewing movements in a rodent model of tardive dyskinesia, Neuroscience, vol.112, issue.4, pp.851-860, 2002.
DOI : 10.1016/S0306-4522(02)00127-6

C. Mcdermott and L. Schrader, Activation of ?? opioid receptors increases intrinsic excitability of dentate gyrus granule cells, The Journal of Physiology, vol.85, issue.14, pp.3517-3549, 2011.
DOI : 10.1113/jphysiol.2011.211623

A. Mcquiston, Mu opioid receptor activation normalizes temporo-ammonic pathway driven inhibition in hippocampal CA1, Neuropharmacology, vol.60, issue.2-3, pp.472-481, 2011.
DOI : 10.1016/j.neuropharm.2010.10.029

A. Meirsman, L. Merrer, J. Pellissier, L. Diaz, J. Clesse et al., Mice Lacking GPR88 Show Motor Deficit, Improved Spatial Learning, and Low Anxiety Reversed by Delta Opioid Antagonist, Biological Psychiatry, vol.79, issue.11, 2015.
DOI : 10.1016/j.biopsych.2015.05.020

A. Meirsman, L. Merrer, J. Pellissier, L. Diaz, J. Clesse et al., Mice Lacking GPR88 Show Motor Deficit, Improved Spatial Learning, and Low Anxiety Reversed by Delta Opioid Antagonist, Biological Psychiatry, vol.79, issue.11, pp.917-944, 2016.
DOI : 10.1016/j.biopsych.2015.05.020

K. Menkens, E. Bilsky, K. Wild, P. Portoghese, L. Reid et al., Cocaine place preference is blocked by the ??-opioid receptor antagonist, naltrindole, European Journal of Pharmacology, vol.219, issue.2, pp.345-351, 1992.
DOI : 10.1016/0014-2999(92)90319-Y

S. Middei, R. Geracitano, A. Caprioli, N. Mercuri, and M. Ammassari-teule, Preserved Fronto-Striatal Plasticity and Enhanced Procedural Learning in a Transgenic Mouse Model of Alzheimer's Disease Overexpressing Mutant hAPPswe, Learning & Memory, vol.11, issue.4, pp.447-52, 2004.
DOI : 10.1101/lm.80604

P. Miner, L. Shimonova, A. Khaimov, Y. Borukhova, E. Ilyayeva et al., General, kappa, delta and mu opioid receptor antagonists mediate feeding elicited by the GABA-B agonist baclofen in the ventral tegmental area and nucleus accumbens shell in rats: Reciprocal and regional interactions, Brain Research, vol.1443, pp.34-51, 2012.
DOI : 10.1016/j.brainres.2012.01.015

K. Montgomery, The relation between fear induced by novel stimulation and exploratory drive., Journal of Comparative and Physiological Psychology, vol.48, issue.4, pp.254-60, 1955.
DOI : 10.1037/h0043788

L. Morales, C. Perez-garcia, and L. Alguacil, Effects of yohimbine on the antinociceptive and place conditioning effects of opioid agonists in rodents, British Journal of Pharmacology, vol.80, issue.Suppl 1, pp.172-180, 2001.
DOI : 10.1038/sj.bjp.0704057

A. Mulder, G. Wardeh, F. Hogenboom, and A. Frankhuyzen, ?? and ??-opioid receptor agonists differentially inhibit striatal dopamine and acetylcholine release, Nature, vol.307, issue.5956, pp.278-80, 1984.
DOI : 10.1038/308278a0

D. Mumby, S. Gaskin, M. Glenn, T. Schramek, and H. Lehmann, Hippocampal Damage and Exploratory Preferences in Rats: Memory for Objects, Places, and Contexts, Learning & Memory, vol.9, issue.2, pp.49-57, 2002.
DOI : 10.1101/lm.41302

C. Nielsen, J. Simms, J. Bito-onon, R. Li, S. Ananthan et al., The delta opioid receptor antagonist, SoRI-9409, decreases yohimbine stress-induced reinstatement of ethanol-seeking, Addiction Biology, vol.167, issue.2, pp.224-258, 2012.
DOI : 10.1111/j.1369-1600.2010.00295.x

R. Nogueiras, A. Romero-pico, M. Vazquez, M. Novelle, M. Lopez et al., The Opioid System and Food Intake: Homeostatic and Hedonic Mechanisms, Obesity Facts, vol.5, issue.2, pp.196-207, 2012.
DOI : 10.1159/000338163

C. Nozaki, L. Bourdonnec, B. Reiss, D. Windh, R. Little et al., ??-Opioid Mechanisms for ADL5747 and ADL5859 Effects in Mice: Analgesia, Locomotion, and Receptor Internalization, Journal of Pharmacology and Experimental Therapeutics, vol.342, issue.3, pp.799-807, 2012.
DOI : 10.1124/jpet.111.188987

R. Ogier, L. Wrobel, and M. Raggenbass, Action of tachykinins in the hippocampus: Facilitation of inhibitory drive to GABAergic interneurons, Neuroscience, vol.156, issue.3, pp.527-563, 2008.
DOI : 10.1016/j.neuroscience.2008.08.001

A. Oliveira, J. Hawk, T. Abel, and R. Havekes, Post-training reversible inactivation of the hippocampus enhances novel object recognition memory, Learning & Memory, vol.17, issue.3, pp.155-60, 2010.
DOI : 10.1101/lm.1625310

M. Olmstead, A. Ouagazzal, and B. Kieffer, Mu and Delta Opioid Receptors Oppositely Regulate Motor Impulsivity in the Signaled Nose Poke Task, PLoS ONE, vol.68, issue.2, p.4410, 2009.
DOI : 10.1371/journal.pone.0004410.t002

URL : https://hal.archives-ouvertes.fr/inserm-00370156

D. Overton, Basic mechanisms of state-dependent learning, Psychopharmacol Bull, vol.14, pp.67-75, 1978.

M. Packard, Exhumed from thought: Basal ganglia and response learning in the plus-maze, Behavioural Brain Research, vol.199, issue.1, pp.24-31, 2009.
DOI : 10.1016/j.bbr.2008.12.013

M. Packard and J. Mcgaugh, Inactivation of Hippocampus or Caudate Nucleus with Lidocaine Differentially Affects Expression of Place and Response Learning, Neurobiology of Learning and Memory, vol.65, issue.1, pp.65-72, 1996.
DOI : 10.1006/nlme.1996.0007

F. Pavone, R. Populin, C. Castellano, G. Kreil, and P. Melchiorri, Deltorphin, a naturally occurring peptide with high selectivity for ?? opioid receptors, improves memory consolidation in two inbred strains of mice, Peptides, vol.11, issue.3, pp.591-595, 1990.
DOI : 10.1016/0196-9781(90)90063-B

G. Pei, B. Kieffer, R. Lefkowitz, and N. Freedman, Agonist-dependent phosphorylation of the mouse delta-opioid receptor: involvement of G protein-coupled receptor kinases but not protein kinase C, Molecular pharmacology, vol.48, pp.173-180, 1995.

M. Perreault, D. Graham, S. Scattolon, Y. Wang, H. Szechtman et al., Cotreatment with the kappa opioid agonist U69593 enhances locomotor sensitization to the D2/D3 dopamine agonist quinpirole and alters dopamine D2 receptor and prodynorphin mRNA expression in rats, Psychopharmacology, vol.30, issue.4, pp.485-96, 2007.
DOI : 10.1007/s00213-007-0855-3

S. Perrine, B. Hoshaw, and E. Unterwald, Delta opioid receptor ligands modulate anxiety-like behaviors in the rat, British Journal of Pharmacology, vol.21, issue.(2 Suppl), pp.864-72, 2006.
DOI : 10.1038/sj.bjp.0706686

R. Piskorowski and V. Chevaleyre, Delta-Opioid Receptors Mediate Unique Plasticity onto Parvalbumin-Expressing Interneurons in Area CA2 of the Hippocampus, Journal of Neuroscience, vol.33, issue.36, pp.14567-78, 2013.
DOI : 10.1523/JNEUROSCI.0649-13.2013

R. Place, C. Lykken, Z. Beer, J. Suh, T. Mchugh et al., NMDA signaling in CA1 mediates selectively the spatial component of episodic memory, Learning & Memory, vol.19, issue.4, pp.164-173, 2012.
DOI : 10.1101/lm.025254.111

S. Powell, M. Geyer, D. Gallagher, and M. Paulus, The balance between approach and avoidance behaviors in a novel object exploration paradigm in mice, Behavioural Brain Research, vol.152, issue.2, pp.341-350, 2004.
DOI : 10.1016/j.bbr.2003.10.020

A. Pradhan and P. Clarke, Comparison between ?-opioid receptor functional response and autoradiographic labeling in rat brain and spinal cord, The Journal of Comparative Neurology, vol.36, issue.4, pp.416-442, 2005.
DOI : 10.1002/cne.20378

A. Quintana, E. Sanz, W. Wang, G. Storey, A. Guler et al., Lack of GPR88 enhances medium spiny neuron activity and alters motor- and cue-dependent behaviors, Nature Neuroscience, vol.18, issue.11, pp.1547-55, 2012.
DOI : 10.1172/JCI62923

X. Rezai, L. Faget, E. Bednarek, Y. Schwab, B. Kieffer et al., Mouse Delta Opioid Receptors are Located on Presynaptic Afferents to Hippocampal Pyramidal Cells, Cellular and Molecular Neurobiology, vol.95, issue.2, 2012.
DOI : 10.1007/s10571-011-9791-1

J. Richard, D. Castro, A. Difeliceantonio, M. Robinson, and K. Berridge, Mapping brain circuits of reward and motivation: In the footsteps of Ann Kelley, Neuroscience & Biobehavioral Reviews, vol.37, issue.9, pp.1919-1950, 2013.
DOI : 10.1016/j.neubiorev.2012.12.008

A. Roberts, L. Gold, I. Polis, J. Mcdonald, D. Filliol et al., Increased ethanol selfadministration in delta-opioid receptor knockout mice, Alcohol Clin Exp Res, vol.25, pp.1249-56, 2001.
DOI : 10.1111/j.1530-0277.2001.tb02344.x

Y. Robles, P. Vivas-mejia, H. Ortiz-zuazaga, J. Felix, X. Ramos et al., Hippocampal gene expression profiling in spatial discrimination learning, Neurobiology of Learning and Memory, vol.80, issue.1, pp.80-95, 2003.
DOI : 10.1016/S1074-7427(03)00025-X

M. Rodriguez, P. Abdala, P. Barroso-chinea, and T. Gonzalez-hernandez, The deep mesencephalic nucleus as an output center of basal ganglia: Morphological and electrophysiological similarities with the substantia nigra, Journal of Comparative Neurology, vol.656, issue.suppl, pp.12-31, 2001.
DOI : 10.1002/cne.1299

A. Saitoh, A. Sugiyama, T. Nemoto, H. Fujii, K. Wada et al., The novel ?? opioid receptor agonist KNT-127 produces antidepressant-like and antinociceptive effects in mice without producing convulsions, Behavioural Brain Research, vol.223, issue.2, pp.271-280, 2011.
DOI : 10.1016/j.bbr.2011.04.041

G. Scherrer, K. Befort, C. Contet, J. Becker, A. Matifas et al., The delta agonists DPDPE and deltorphin II recruit predominantly mu receptors to produce thermal analgesia: a parallel study of mu, delta and combinatorial opioid receptor knockout mice, European Journal of Neuroscience, vol.24, issue.8, pp.2239-2287, 2004.
DOI : 10.1146/annurev.pharmtox.36.1.379

G. Scherrer, P. Tryoen-toth, D. Filliol, A. Matifas, D. Laustriat et al., Knockin mice expressing fluorescent ??-opioid receptors uncover G protein-coupled receptor dynamics in vivo, Proceedings of the National Academy of Sciences, vol.103, issue.25, pp.9691-9697, 2006.
DOI : 10.1073/pnas.0603359103

URL : https://hal.archives-ouvertes.fr/hal-00188131

M. Schmelzeis and G. Mittleman, The hippocampus and reward: Effects of hippocampal lesions on progressive-ratio responding., Behavioral Neuroscience, vol.110, issue.5, pp.1049-66, 1996.
DOI : 10.1037/0735-7044.110.5.1049

J. Schroeder, J. Wingard, and M. Packard, Post-training reversible inactivation of hippocampus reveals interference between memory systems, Hippocampus, vol.218, issue.2, pp.280-284, 2002.
DOI : 10.1002/hipo.10024

G. Schulteis, J. Martinez, and J. , ICI 174,864, a selective delta opioid antagonist, reverses the learning impairment produced by [leu]enkephalin, Psychopharmacology, vol.14, issue.1, pp.102-111, 1990.
DOI : 10.1007/BF02245798

N. Shahabi, Y. Daaka, K. Mcallen, and B. Sharp, Delta opioid receptors expressed by stably transfected jurkat cells signal through the map kinase pathway in a ras-independent manner, Journal of Neuroimmunology, vol.94, issue.1-2, pp.48-57, 1999.
DOI : 10.1016/S0165-5728(98)00211-2

M. Shiflett and B. Balleine, At the limbic-motor interface: disconnection of basolateral amygdala from nucleus accumbens core and shell reveals dissociable components of incentive motivation, European Journal of Neuroscience, vol.50, issue.10, pp.1735-1778, 2010.
DOI : 10.1111/j.1460-9568.2010.07439.x

T. Shippenberg, R. Bals-kubik, and A. Herz, Motivational properties of opioids: evidence that an activation of ??-receptors mediates reinforcement processes, Brain Research, vol.436, issue.2, pp.234-243, 1987.
DOI : 10.1016/0006-8993(87)91667-2

D. Simmons and D. Self, Role of Mu- and Delta-Opioid Receptors in the Nucleus Accumbens in Cocaine-Seeking Behavior, Neuropsychopharmacology, vol.17, issue.8, pp.1946-57, 2009.
DOI : 10.1038/nm1196-1225

R. Simon, H. Brylka, H. Schwegler, S. Venkataramanappa, J. Andratschke et al., A dual function of Bcl11b/Ctip2 in hippocampal neurogenesis, The EMBO Journal, vol.139, issue.13, pp.2922-2958, 2012.
DOI : 10.1038/emboj.2012.142

S. Slowe, F. Simonin, B. Kieffer, and I. Kitchen, Quantitative autoradiography of ??-,??- and ??1 opioid receptors in ??-opioid receptor knockout mice, Brain Research, vol.818, issue.2, pp.335-380, 1999.
DOI : 10.1016/S0006-8993(98)01201-3

D. Stephens, T. Duka, H. Crombag, C. Cunningham, M. Heilig et al., Reward sensitivity: issues of measurement, and achieving consilience between human and animal phenotypes, Addiction Biology, vol.202, issue.2, pp.145-68, 2010.
DOI : 10.1111/j.1369-1600.2009.00193.x

S. Sutt, S. Raud, U. Abramov, J. Innos, H. Luuk et al., Relation of exploratory behaviour to plasma corticosterone and Wfs1 gene expression in Wistar rats, Journal of Psychopharmacology, vol.17, issue.6, pp.905-918, 2010.
DOI : 10.1177/0269881109102738

T. Suzuki, M. Tsuji, T. Mori, H. Ikeda, M. Misawa et al., Involvement of dopamine-dependent and -independent mechanisms in the rewarding effects mediated by ?? opioid receptor subtypes in mice, Brain Research, vol.744, issue.2, pp.327-361, 1997.
DOI : 10.1016/S0006-8993(96)01119-5

T. Suzuki, M. Yoshiike, H. Mizoguchi, J. Kamei, M. Misawa et al., Blockade of .DELTA.-Opioid Receptors Prevents Morphine-Induced Place Preference in Mice., The Japanese Journal of Pharmacology, vol.66, issue.1, pp.131-138, 1994.
DOI : 10.1254/jjp.66.131

K. Svoboda, C. Adams, and C. Lupica, Opioid receptor subtype expression defines morphologically distinct classes of hippocampal interneurons, The Journal of neuroscience : the official journal of the Society for Neuroscience, vol.19, pp.85-95, 1999.

P. Tsao and M. Von-zastrow, Type-specific Sorting of G Protein-coupled Receptors after Endocytosis, Journal of Biological Chemistry, vol.275, issue.15, pp.11130-11170, 2000.
DOI : 10.1074/jbc.275.15.11130

E. Tzavara, F. Bymaster, R. Davis, M. Wade, K. Perry et al., M4 muscarinic receptors regulate the dynamics of cholinergic and dopaminergic neurotransmission: relevance to the pathophysiology and treatment of related CNS pathologies, Faseb J, vol.18, pp.1410-1412, 2004.

M. Ukai, A. Takada, Y. Sasaki, and T. Kameyama, Stimulation of ??1- and ??2-opioid receptors produces amnesia in mice, European Journal of Pharmacology, vol.338, issue.1, pp.1-6, 1997.
DOI : 10.1016/S0014-2999(97)01310-1

J. Wang, Y. Ma, and M. Van-den-buuse, Improved spatial recognition memory in mice lacking adenosine A2A receptors, Experimental Neurology, vol.199, issue.2, pp.438-483, 2006.
DOI : 10.1016/j.expneurol.2006.01.005

S. Ward and D. Roberts, Microinjection of the ??-opioid receptor selective antagonist naltrindole 5???-isothiocyanate site specifically affects cocaine self-administration in rats responding under a progressive ratio schedule of reinforcement, Behavioural Brain Research, vol.182, issue.1, pp.140-144, 2007.
DOI : 10.1016/j.bbr.2007.05.003

C. Wei, P. Singer, J. Coelho, D. Boison, J. Feldon et al., Selective inactivation of adenosine A2A receptors in striatal neurons enhances working memory and reversal learning, Learning & Memory, vol.18, issue.7, pp.459-74, 2011.
DOI : 10.1101/lm.2136011

N. White and R. Mcdonald, Multiple Parallel Memory Systems in the Brain of the Rat, Neurobiology of Learning and Memory, vol.77, issue.2, pp.125-84, 2002.
DOI : 10.1006/nlme.2001.4008

C. Xie and D. Lewis, Endogenous opioids regulate long-term potentiation of synaptic inhibition in the dentate gyrus of rat hippocampus, The Journal of neuroscience : the official journal of the Society for Neuroscience, vol.15, pp.3788-95, 1995.

S. Yang, Y. Kawamura, and M. Yoshikawa, Effect of rubiscolin, a ?? opioid peptide derived from Rubisco, on memory consolidation, Peptides, vol.24, issue.2, pp.325-333, 2003.
DOI : 10.1016/S0196-9781(03)00044-5

B. Yin and W. Meck, Comparison of interval timing behaviour in mice following dorsal or ventral hippocampal lesions with mice having ??-opioid receptor gene deletion, Philosophical Transactions of the Royal Society B: Biological Sciences, vol.14, issue.6, p.20120466, 2014.
DOI : 10.1111/j.1460-9568.2006.05064.x

B. Yin and A. Troger, Exploring the 4th Dimension: Hippocampus, Time, and Memory Revisited, Frontiers in Integrative Neuroscience, vol.5, p.36, 2011.
DOI : 10.3389/fnint.2011.00036

URL : http://doi.org/10.3389/fnint.2011.00036

H. Yin, S. Mulcare, M. Hilario, E. Clouse, T. Holloway et al., Dynamic reorganization of striatal circuits during the acquisition and consolidation of a skill, Nature Neuroscience, vol.5, issue.3, pp.333-374, 2009.
DOI : 10.1038/nn.2261

Z. Zhang, S. Xin, G. Wu, W. Zhang, L. Ma et al., Endogenous ??-Opioid and ORL1 Receptors Couple to Phosphorylation and Activation of p38 MAPK in NG108-15 Cells and This Is Regulated by Protein Kinase A and Protein Kinase C, Journal of Neurochemistry, vol.272, issue.4, pp.1502-1511, 1999.
DOI : 10.1046/j.1471-4159.1999.0731502.x

S. Zhou, M. Zhu, D. Shu, X. Du, X. Song et al., Preferential enhancement of working memory in mice lacking adenosine A2A receptors, Brain Research, vol.1303, pp.74-83, 2009.
DOI : 10.1016/j.brainres.2009.09.082