S. Abdalla, H. Lother, A. M. Abdel-tawab, and U. Quitterer, The Angiotensin II AT2Receptor Is an AT1Receptor Antagonist, Journal of Biological Chemistry, vol.276, issue.43, pp.39721-39726, 2001.

F. Alkufri, A. Shaag, B. Abu-libdeh, and O. Elpeleg, Deleterious mutation inGPR88is associated with chorea, speech delay, and learning disabilities, Neurology Genetics, vol.2, issue.3, p.e64, 2016.

T. M. Arefin, A. E. Mechling, A. C. Meirsman, T. Bienert, N. S. Hübner et al., Remodeling of Sensorimotor Brain Connectivity in Gpr88-Deficient Mice, Brain Connectivity, vol.7, issue.8, pp.526-540, 2017.

M. A. Ayoub, H. B. See, R. M. Seeber, S. P. Armstrong, and K. D. Pfleger, Profiling Epidermal Growth Factor Receptor and Heregulin Receptor 3 Heteromerization Using Receptor Tyrosine Kinase Heteromer Investigation Technology, PLoS ONE, vol.8, issue.5, p.e64672, 2013.

A. Becker, G. Grecksch, J. Kraus, B. Peters, H. Schroeder et al., Loss of locomotor sensitisation in response to morphine in D1 receptor deficient mice, Naunyn-Schmiedeberg's Archives of Pharmacology, vol.363, issue.5, pp.562-568, 2001.

J. A. Becker, K. Befort, C. Blad, D. Filliol, A. Ghate et al., Transcriptome analysis identifies genes with enriched expression in the mouse central extended amygdala, Neuroscience, vol.156, issue.4, pp.950-965, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00350728

J. A. Becker, B. L. Kieffer, and J. Le-merrer, Differential behavioral and molecular alterations upon protracted abstinence from cocaine versus morphine, nicotine, THC and alcohol, Addiction Biology, vol.22, issue.5, pp.1205-1217, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01595696

B. Hamida, S. Mendonç-a-netto, S. Arefin, T. M. Nasseef, M. T. Boulos et al., Increased Alcohol Seeking in Mice Lacking Gpr88 Involves Dysfunctional Mesocorticolimbic Networks, Biological Psychiatry, vol.84, pp.202-212, 2018.

F. Berrendero, B. L. Kieffer, and R. Maldonado, Attenuation of Nicotine-Induced Antinociception, Rewarding Effects, and Dependence in ?-Opioid Receptor Knock-Out Mice, The Journal of Neuroscience, vol.22, issue.24, pp.10935-10940, 2002.

Y. Bi, C. D. Dzierba, C. Fink, Y. Garcia, M. Green et al., The discovery of potent agonists for GPR88, an orphan GPCR, for the potential treatment of CNS disorders, Bioorganic & Medicinal Chemistry Letters, vol.25, issue.7, pp.1443-1447, 2015.

A. Borgkvist, E. Valjent, E. Santini, D. Hervé, J. A. Girault et al., Delayed, context- and dopamine D1 receptor-dependent activation of ERK in morphine-sensitized mice, Neuropharmacology, vol.55, issue.2, pp.230-237, 2008.

D. O. Borroto-escuela, M. Flajolet, L. F. Agnati, P. Greengard, and K. Fuxe, Bioluminescence Resonance Energy Transfer Methods to Study G Protein-Coupled Receptor?Receptor Tyrosine Kinase Heteroreceptor Complexes, Methods in Cell Biology, vol.117, pp.141-164, 2013.

P. E. Brandish, M. Su, D. J. Holder, P. Hodor, J. Szumiloski et al., Regulation of Gene Expression by Lithium and Depletion of Inositol in Slices of Adult Rat Cortex, Neuron, vol.45, issue.6, pp.861-872, 2005.

P. Charbogne, O. Gardon, E. Martín-garcía, H. L. Keyworth, A. Matsui et al., Mu Opioid Receptors in Gamma-Aminobutyric Acidergic Forebrain Neurons Moderate Motivation for Heroin and Palatable Food, Biological Psychiatry, vol.81, issue.9, pp.778-788, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02349326

S. L. Cole, M. J. Robinson, and K. C. Berridge, Optogenetic self-stimulation in the nucleus accumbens: D1 reward versus D2 ambivalence, PLOS ONE, vol.13, issue.11, p.e0207694, 2018.

B. Conti, R. Maier, A. M. Barr, M. C. Morale, X. Lu et al., Region-specific transcriptional changes following the three antidepressant treatments electro convulsive therapy, sleep deprivation and fluoxetine, Molecular Psychiatry, vol.12, issue.2, pp.167-189, 2006.

A. M. Decker, E. A. Gay, K. M. Mathews, T. C. Rosa, T. L. Langston et al., Development and validation of a high-throughput calcium mobilization assay for the orphan receptor GPR88, Journal of Biomedical Science, vol.24, issue.1, p.23, 2017.

D. Zompo, M. Deleuze, J. F. Chillotti, C. Cousin, E. Niehaus et al., Association study in three different populations between the GPR88 gene and major psychoses, Molecular Genetics & Genomic Medicine, vol.2, pp.152-159, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02989910

C. D. Dzierba, Y. Bi, B. Dasgupta, R. A. Hartz, V. Ahuja et al., Design, synthesis, and evaluation of phenylglycinols and phenyl amines as agonists of GPR88, Bioorganic & Medicinal Chemistry Letters, vol.25, issue.7, pp.1448-1452, 2015.

A. T. Ehrlich, M. Semache, J. Bailly, S. Wojcik, T. M. Arefin et al., Mapping GPR88-Venus illuminates a novel role for GPR88 in sensory processing, Brain Structure and Function, vol.223, issue.3, pp.1275-1296, 2017.

A. T. Ehrlich, G. Maroteaux, A. Robe, L. Venteo, M. T. Nasseef et al., Expression map of 78 brain-expressed mouse orphan GPCRs provides a translational resource for neuropsychiatric research, Communications Biology, vol.1, issue.1, p.102, 2018.

S. Ferré, L. F. Agnati, F. Ciruela, C. Lluis, A. S. Woods et al., Neurotransmitter receptor heteromers and their integrative role in ?local modules?: The striatal spine module, Brain Research Reviews, vol.55, issue.1, pp.55-67, 2007.

W. Fujita, I. Gomes, and L. A. Devi, Revolution in GPCR signalling: opioid receptor heteromers as novel therapeutic targets: IUPHAR Review 10, British Journal of Pharmacology, vol.171, issue.18, pp.4155-4176, 2014.

A. Ghate, K. Befort, J. A. 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.1182-1192, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00189988

J. A. Girault, E. Valjent, J. Caboche, and D. Herve, ERK2: a logical AND gate critical for drug-induced plasticity?, Current Opinion in Pharmacology, vol.7, issue.1, pp.77-85, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00128883

C. Goudet, J. Kniazeff, V. Hlavackova, F. Malhaire, D. Maurel et al., Asymmetric Functioning of Dimeric Metabotropic Glutamate Receptors Disclosed by Positive Allosteric Modulators, Journal of Biological Chemistry, vol.280, issue.26, pp.24380-24385, 2005.
URL : https://hal.archives-ouvertes.fr/inserm-00318925

T. J. Haley and W. G. Mccormick, PHARMACOLOGICAL EFFECTS PRODUCED BY INTRACEREBRAL INJECTION OF DRUGS IN THE CONSCIOUS MOUSE, British Journal of Pharmacology and Chemotherapy, vol.12, issue.1, pp.12-15, 1957.

M. Heiman, A. Schaefer, S. Gong, J. D. Peterson, M. Day et al., A Translational Profiling Approach for the Molecular Characterization of CNS Cell Types, Cell, vol.135, issue.4, pp.738-748, 2008.

H. Ho, M. D. Both, A. Siniard, S. Sharma, J. H. Notwell et al., A Guide to Single-Cell Transcriptomics in Adult Rodent Brain: The Medium Spiny Neuron Transcriptome Revisited, Frontiers in Cellular Neuroscience, vol.12, p.159, 2018.

A. Ignatov, J. Lintzel, I. Hermans-borgmeyer, H. J. Kreienkamp, P. Joost et al., Role of the G-Protein-Coupled Receptor GPR12 as High-Affinity Receptor for Sphingosylphosphorylcholine and Its Expression and Function in Brain Development, The Journal of Neuroscience, vol.23, issue.3, pp.907-914, 2003.

C. Jin, A. M. Decker, X. P. Huang, B. P. Gilmour, B. E. Blough et al., Synthesis, Pharmacological Characterization, and Structure?Activity Relationship Studies of Small Molecular Agonists for the Orphan GPR88 Receptor, ACS Chemical Neuroscience, vol.5, issue.7, pp.576-587, 2014.

C. Jin, A. M. Decker, V. H. Makhijani, J. Besheer, E. Darcq et al., Discovery of a Potent, Selective, and Brain-Penetrant Small Molecule that Activates the Orphan Receptor GPR88 and Reduces Alcohol Intake, Journal of Medicinal Chemistry, vol.61, issue.15, pp.6748-6758, 2018.

P. Joost and A. Methner, Phylogenetic analysis of 277 human G-protein-coupled receptors as a tool for the prediction of orphan receptor ligands, Genome Biology, vol.3, issue.11, p.research0063.1, 2002.

K. K. Kakarala and K. Jamil, Sequence-structure based phylogeny of GPCR Class A Rhodopsin receptors, Molecular Phylogenetics and Evolution, vol.74, pp.66-96, 2014.

U. Klein, C. Müller, P. Chu, M. Birnbaumer, and M. Von-zastrow, Heterologous Inhibition of G Protein-coupled Receptor Endocytosis Mediated by Receptor-specific Trafficking of ?-Arrestins, Journal of Biological Chemistry, vol.276, issue.20, pp.17442-17447, 2001.

J. Le-merrer, J. A. Becker, K. A. Befort, and B. L. Kieffer, Reward Processing by the Opioid System in the Brain, Physiological Reviews, vol.89, issue.4, pp.1379-1412, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00438654

J. Le-merrer, K. Befort, O. Gardon, D. Filliol, E. Darcq et al., Protracted abstinence from distinct drugs of abuse shows regulation of a common gene network, Addiction Biology, vol.17, issue.1, pp.1-12, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02650740

J. Le-merrer, X. Rezai, G. Scherrer, J. A. Becker, and B. L. Kieffer, Impaired Hippocampus-Dependent and Facilitated Striatum-Dependent Behaviors in Mice Lacking the Delta Opioid Receptor, Neuropsychopharmacology, vol.38, issue.6, pp.1050-1059, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02266437

A. Levoye, J. Dam, M. A. Ayoub, J. Guillaume, C. Couturier et al., The orphan GPR50 receptor specifically inhibits MT1 melatonin receptor function through heterodimerization, The EMBO Journal, vol.25, issue.13, pp.3012-3023, 2006.

A. Levoye, J. Dam, M. A. Ayoub, J. L. Guillaume, and R. Jockers, Do orphan G?protein?coupled receptors have ligand?independent functions?, EMBO reports, vol.7, issue.11, pp.1094-1098, 2006.

S. F. Logue, S. M. 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.438-447, 2009.

R. Massart, J. P. 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.30, issue.3, pp.397-414, 2009.

R. Massart, V. Mignon, J. Stanic, P. Munoz-tello, J. A. Becker et al., Developmental and adult expression patterns of the G-protein-coupled receptor GPR88 in the rat: Establishment of a dual nuclear-cytoplasmic localization, Journal of Comparative Neurology, vol.524, issue.14, pp.2776-2802, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01594963

H. W. Matthes, R. Maldonado, F. Simonin, O. Valverde, S. Slowe et al., Loss of morphine-induced analgesia, reward effect and withdrawal symptoms in mice lacking the µ-opioid-receptor gene, Nature, vol.383, issue.6603, pp.819-823, 1996.

A. C. Meirsman, J. Le-merrer, L. P. Pellissier, J. P. Diaz, D. 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.917-927, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01594891

A. C. Meirsman, A. Robé, A. De-kerchove-d?exaerde, and B. L. Kieffer, GPR88 in A2AR Neurons Enhances Anxiety-Like Behaviors, eneuro, vol.3, issue.4, pp.ENEURO.0202-16.2016, 2016.

C. A. Ogden, M. E. Rich, N. J. Schork, M. P. Paulus, M. A. Geyer et al., Candidate genes, pathways and mechanisms for bipolar (manic?depressive) and related disorders: an expanded convergent functional genomics approach, Molecular Psychiatry, vol.9, issue.11, pp.1007-1029, 2004.

Z. W. Pavlovic, M. L. Cooper, and R. J. Bodnar, Opioid antagonists in the periaqueductal gray inhibit morphine and ?-endorphin analgesia elicited from the amygdala of rats, Brain Research, vol.741, issue.1-2, pp.13-26, 1996.

Z. W. Pavlovic and R. J. Bodnar, Opioid supraspinal analgesic synergy between the amygdala and periaqueductal gray in rats, Brain Research, vol.779, issue.1-2, pp.158-169, 1998.

L. P. Pellissier, C. N. Pujol, J. A. Becker, and J. Le-merrer, Delta Opioid Receptors: Learning and Motivation, Delta Opioid Receptor Pharmacology and Therapeutic Applications, vol.247, pp.227-260, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01437672

J. P. Pin, J. Kniazeff, L. Prézeau, J. F. Liu, and P. Rondard, GPCR interaction as a possible way for allosteric control between receptors, Molecular and Cellular Endocrinology, vol.486, pp.89-95, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02396289

A. Quintana, E. Sanz, W. Wang, G. P. Storey, A. D. Güler et al., Lack of GPR88 enhances medium spiny neuron activity and alters motor- and cue-dependent behaviors, Nature Neuroscience, vol.15, issue.11, pp.1547-1555, 2012.

A. Rainwater, E. Sanz, R. D. Palmiter, and A. Quintana, Striatal GPR88 Modulates Foraging Efficiency, The Journal of Neuroscience, vol.37, issue.33, pp.7939-7947, 2017.

K. Ramabadran, M. Bansinath, H. Turndorf, and M. M. Puig, Tail immersion test for the evaluation of a nociceptive reaction in mice, Journal of Pharmacological Methods, vol.21, issue.1, pp.21-31, 1989.

H. Ruan, J. Sun, X. Liu, L. Liu, R. Cui et al., Cholinergic M4 receptors are involved in morphine-induced expression of behavioral sensitization by regulating dopamine function in the nucleus accumbens of rats, Behavioural Brain Research, vol.360, pp.128-133, 2019.

F. Schmidlin, O. Dery, N. W. Bunnett, and E. F. Grady, Heterologous regulation of trafficking and signaling of G protein-coupled receptors: -Arrestin-dependent interactions between neurokinin receptors, Proceedings of the National Academy of Sciences, vol.99, issue.5, pp.3324-3329, 2002.

J. S. Surgand, J. Rodrigo, E. Kellenberger, and D. Rognan, A chemogenomic analysis of the transmembrane binding cavity of human G-protein-coupled receptors, Proteins: Structure, Function, and Bioinformatics, vol.62, issue.2, pp.509-538, 2005.

D. J. Surmeier, J. Ding, M. Day, Z. Wang, and W. Z. Shen, D1 and D2 dopamine-receptor modulation of striatal glutamatergic signaling in striatal medium spiny neurons, Trends in Neurosciences, vol.30, issue.5, pp.228-235, 2007.

Y. Tao, C. Yu, W. C. Wang, Y. Hou, X. Xu et al., Heteromers of ? opioid and dopamine D1receptors modulate opioid-induced locomotor sensitization in a dopamine-independent manner, British Journal of Pharmacology, vol.174, issue.17, pp.2842-2861, 2017.

N. M. Urs, T. L. Daigle, and M. G. Caron, A Dopamine D1 Receptor-Dependent ?-Arrestin Signaling Complex Potentially Regulates Morphine-Induced Psychomotor Activation but not Reward in Mice, Neuropsychopharmacology, vol.36, issue.3, pp.551-558, 2010.

E. Valjent, V. Pascoli, P. Svenningsson, S. Paul, H. Enslen et al., From The Cover: Regulation of a protein phosphatase cascade allows convergent dopamine and glutamate signals to activate ERK in the striatum, Proceedings of the National Academy of Sciences, vol.102, issue.2, pp.491-496, 2004.

E. Valjent, J. Bertran-gonzalez, B. Aubier, P. Greengard, D. Hervé et al., Mechanisms of Locomotor Sensitization to Drugs of Abuse in a Two-Injection Protocol, Neuropsychopharmacology, vol.35, issue.2, pp.401-415, 2009.

V. Van-waes, K. Y. Tseng, and H. Steiner, GPR88 -a putative signaling molecule predominantly expressed in the striatum: cellular localization and developmental regulation, Basal Ganglia, vol.1, pp.83-89, 2011.

D. K. Vassilatis, J. G. Hohmann, H. Zeng, F. Li, J. E. Ranchalis et al., The G protein-coupled receptor repertoires of human and mouse, Proceedings of the National Academy of Sciences, vol.100, issue.8, pp.4903-4908, 2003.

J. P. Vilardaga, V. O. Nikolaev, K. Lorenz, S. Ferrandon, Z. Zhuang et al., Conformational cross-talk between ?2A-adrenergic and ?-opioid receptors controls cell signaling, Nature Chemical Biology, vol.4, issue.2, pp.126-131, 2008.

A. O. Watts, M. M. Van-lipzig, W. C. Jaeger, R. M. Seeber, M. Van-zwam et al., Identification and profiling of CXCR3-CXCR4 chemokine receptor heteromer complexes, British Journal of Pharmacology, vol.168, issue.7, pp.1662-1674, 2013.

J. T. Williams, M. J. Christie, and O. Manzoni, Cellular and Synaptic Adaptations Mediating Opioid Dependence, Physiological Reviews, vol.81, issue.1, pp.299-343, 2001.

C. H. Yang, H. W. Huang, K. H. Chen, Y. H. Chen, S. M. Sheen-chen et al., Antinociceptive potentiation and attenuation of tolerance by intrathecal ?-arrestin 2 small interfering RNA in rats, British Journal of Anaesthesia, vol.107, issue.5, pp.774-781, 2011.