M. Rodbell, The role of hormone receptors and GTP-regulatory proteins in membrane transduction, Nature, vol.23, issue.5751, pp.17-22, 1980.
DOI : 10.1038/284017a0

W. Mcintire, Structural Determinants Involved in the Formation and Activation of G Protein βγ Dimers, Neurosignals, vol.17, issue.1, pp.82-99, 2009.
DOI : 10.1159/000186692

T. Hynes, S. Mervine, E. Yost, J. Sabo, and C. Berlot, -Adrenergic Receptor, Journal of Biological Chemistry, vol.279, issue.42, pp.44101-44113, 2004.
DOI : 10.1074/jbc.M405151200

URL : https://hal.archives-ouvertes.fr/hal-00159205

D. Dupre, M. Robitaille, R. Rebois, and T. Hebert, The Role of G???? Subunits in the Organization, Assembly, and Function of GPCR Signaling Complexes, Annual Review of Pharmacology and Toxicology, vol.49, issue.1, pp.31-56, 2009.
DOI : 10.1146/annurev-pharmtox-061008-103038

W. Oldham and H. Hamm, Heterotrimeric G protein activation by G-protein-coupled receptors, Nature Reviews Molecular Cell Biology, vol.409, issue.1, pp.60-71, 2008.
DOI : 10.1038/nrm2299

T. Iiri, Z. Farfel, and H. Bourne, G-protein diseases furnish a model for the turn-on switch, Nature, vol.394, pp.35-43, 1998.

H. Hamm, The Many Faces of G Protein Signaling, Journal of Biological Chemistry, vol.273, issue.2, pp.669-72, 1998.
DOI : 10.1074/jbc.273.2.669

M. Wall, D. Coleman, E. Lee, J. Iniguez-lluhi, B. Posner et al., The structure of the G protein heterotrimer Gi??1??1??2, Cell, vol.83, issue.6, pp.1047-58, 1995.
DOI : 10.1016/0092-8674(95)90220-1

D. Lambright, J. Sondek, A. Bohm, N. Skiba, H. Hamm et al., The 2.0 ?? crystal structure of a heterotrimeric G protein, Nature, vol.379, issue.6563, pp.311-320, 1996.
DOI : 10.1038/379311a0

D. Michaelson, I. Ahearn, M. Bergo, S. Young, and M. Philips, Membrane Trafficking of Heterotrimeric G Proteins via the Endoplasmic Reticulum and Golgi, Molecular Biology of the Cell, vol.13, issue.9, pp.3294-302, 2002.
DOI : 10.1091/mbc.E02-02-0095

H. Bourne, How receptors talk to trimeric G proteins, Current Opinion in Cell Biology, vol.9, issue.2, pp.134-176, 1997.
DOI : 10.1016/S0955-0674(97)80054-3

H. Bourne, D. Sanders, and F. Mccormick, The GTPase superfamily: conserved structure and molecular mechanism, Nature, vol.349, issue.6305, pp.117-144, 1991.
DOI : 10.1038/349117a0

C. Gales, V. Durm, J. Schaak, S. Pontier, S. Percherancier et al., Probing the activation-promoted structural rearrangements in preassembled receptor???G protein complexes, Nature Structural & Molecular Biology, vol.21, issue.9, pp.778-86, 2006.
DOI : 10.1006/smns.1998.0125

M. Bunemann, M. Frank, and M. Lohse, Gi protein activation in intact cells involves subunit rearrangement rather than dissociation, Proceedings of the National Academy of Sciences, vol.100, issue.26, pp.16077-82, 2003.
DOI : 10.1073/pnas.2536719100

M. Sato, J. Blumer, V. Simon, and S. Lanier, ACCESSORY PROTEINS FOR G PROTEINS: Partners in Signaling, Annual Review of Pharmacology and Toxicology, vol.46, issue.1, pp.151-87, 2006.
DOI : 10.1146/annurev.pharmtox.46.120604.141115

D. Siderovski and F. Willard, The GAPs, GEFs, and GDIs of heterotrimeric G-protein alpha subunits, International Journal of Biological Sciences, vol.1, pp.51-66, 2005.
DOI : 10.7150/ijbs.1.51

E. Ross and T. Wilkie, GTPase-Activating Proteins for Heterotrimeric G Proteins: Regulators of G Protein Signaling (RGS) and RGS-Like Proteins, Annual Review of Biochemistry, vol.69, issue.1, pp.795-827, 2000.
DOI : 10.1146/annurev.biochem.69.1.795

T. Cabrera-vera, J. Vanhauwe, T. Thomas, M. Medkova, A. Preininger et al., Insights into G Protein Structure, Function, and Regulation, Endocrine Reviews, vol.24, issue.6, pp.765-81, 2003.
DOI : 10.1210/er.2000-0026

G. Milligan and E. Kostenis, Heterotrimeric G-proteins: a short history, British Journal of Pharmacology, vol.276, issue.S1, pp.46-55, 2006.
DOI : 10.1038/sj.bjp.0706405

R. Navarro-olmos, L. Kawasaki, L. Dominguez-ramirez, L. Ongay-larios, R. Perez-molina et al., The ?? Subunit of the Heterotrimeric G Protein Triggers the Kluyveromyces lactis Pheromone Response Pathway in the Absence of the ?? Subunit, Molecular Biology of the Cell, vol.21, issue.3, pp.489-98, 2010.
DOI : 10.1091/mbc.E09-06-0472

C. Nakai and G. Brooker, Assay for adenylate cyclase and cyclic nucleotide phosphodiesterases and the preparation of high specific activity 3 2P-labeled substrates, Biochimica et Biophysica Acta (BBA) - Enzymology, vol.391, issue.1, pp.222-261, 1975.
DOI : 10.1016/0005-2744(75)90169-2

A. Steiner, D. Kipnis, R. Utiger, and C. Parker, RADIOIMMUNOASSAY FOR THE MEASUREMENT OF ADENOSINE 3',5'-CYCLIC PHOSPHATE, Proceedings of the National Academy of Sciences, vol.64, issue.1, pp.367-73, 1969.
DOI : 10.1073/pnas.64.1.367

V. Nikolaev, C. Hoffmann, M. Bunemann, M. Lohse, and J. Vilardaga, -protein Heterotrimer, Journal of Biological Chemistry, vol.281, issue.34, pp.24506-24517, 2006.
DOI : 10.1074/jbc.M603266200

URL : https://hal.archives-ouvertes.fr/in2p3-00021454

D. Willoughby and D. Cooper, Live-cell imaging of cAMP dynamics, Nature Methods, vol.274, issue.1, pp.29-36, 2008.
DOI : 10.1038/nmeth1135

L. Dipilato, X. Cheng, and J. Zhang, Fluorescent indicators of cAMP and Epac activation reveal differential dynamics of cAMP signaling within discrete subcellular compartments, Proceedings of the National Academy of Sciences, vol.101, issue.47, pp.16513-16521, 2004.
DOI : 10.1073/pnas.0405973101

J. Zhang, Y. Ma, S. Taylor, and R. Tsien, Genetically encoded reporters of protein kinase A activity reveal impact of substrate tethering, Proceedings of the National Academy of Sciences, vol.98, issue.26, pp.14997-5002, 2001.
DOI : 10.1073/pnas.211566798

A. Prinz, M. Diskar, A. Erlbruch, and F. Herberg, Novel, isotype-specific sensors for protein kinase A subunit interaction based on bioluminescence resonance energy transfer (BRET), Cellular Signalling, vol.18, issue.10, pp.1616-1641, 2006.
DOI : 10.1016/j.cellsig.2006.01.013

J. Pantel, S. Williams, M. D. Sebag, J. Corbin, J. Weaver et al., Development of a high throughput screen for allosteric modulators of melanocortin-4 receptor signaling using a real time cAMP assay, European Journal of Pharmacology, vol.660, issue.1, pp.139-186, 2011.
DOI : 10.1016/j.ejphar.2011.01.031

J. Tesmer, R. Sunahara, A. Gilman, and S. Sprang, Crystal structure of the catalytic domains of adenylyl cyclase in a complex with Gsalpha, GTPgammaS Science, vol.278, pp.1907-1923, 1997.

M. Ghahremani, P. Cheng, P. Lembo, and P. Albert, Distinct Roles for G??i2, G??i3, and G???? in Modulation of Forskolin- or Gs-mediated cAMP Accumulation and Calcium Mobilization by Dopamine D2S Receptors, Journal of Biological Chemistry, vol.274, issue.14, pp.9238-9283, 1999.
DOI : 10.1074/jbc.274.14.9238

E. Sutkowski, W. Tang, C. Broome, J. Robbins, and K. Seamon, Regulation of forskolin interactions with type I, II, V, and VI adenylyl cyclases by Gs.alpha., Biochemistry, vol.33, issue.43, pp.12852-12861, 1994.
DOI : 10.1021/bi00209a017

B. Conklin, Z. Farfel, K. Lustig, J. D. Bourne, and H. , Substitution of three amino acids switches receptor specificity of Gq?? to that of Gi??, Nature, vol.363, issue.6426, pp.274-280, 1993.
DOI : 10.1038/363274a0

S. Offermanns and M. Simon, G??15 and G??16 Couple a Wide Variety of Receptors to Phospholipase C, Journal of Biological Chemistry, vol.270, issue.25, pp.15175-80, 1995.
DOI : 10.1074/jbc.270.25.15175

S. Mody, M. Ho, S. Joshi, and Y. Wong, Incorporation of Galpha(z)-specific sequence at the carboxyl terminus increases the promiscuity of galpha(16) toward G(i)-coupled receptors, Mol Pharmacol, vol.57, pp.13-23, 2000.

A. Hazari, V. Lowes, J. Chan, C. Wong, M. Ho et al., Replacement of the ??5 helix of G??16 with G??s-specific sequences enhances promiscuity of G??16 toward Gs-coupled receptors, Cellular Signalling, vol.16, issue.1, pp.51-62, 2004.
DOI : 10.1016/S0898-6568(03)00097-4

A. Liu, M. Ho, C. Wong, J. Chan, A. Pau et al., G?? 16/z Chimeras Efficiently Link a Wide Range of G Protein-- Coupled Receptors to Calcium Mobilization, Journal of Biomolecular Screening, vol.8, issue.1, pp.39-49, 2003.
DOI : 10.1177/1087057102239665

A. Heydorn, R. Ward, R. Jorgensen, M. Rosenkilde, T. Frimurer et al., Identification of a Novel Site within G Protein ?? Subunits Important for Specificity of Receptor-G Protein Interaction, Molecular Pharmacology, vol.66, issue.2, pp.250-259, 2004.
DOI : 10.1124/mol.66.2.250

E. Kostenis, L. Martini, J. Ellis, M. Waldhoer, A. Heydorn et al., A Highly Conserved Glycine within Linker I and the Extreme C Terminus of G Protein ?? Subunits Interact Cooperatively in Switching G Protein-Coupled Receptor-to-Effector Specificity, Journal of Pharmacology and Experimental Therapeutics, vol.313, issue.1, pp.78-87, 2005.
DOI : 10.1124/jpet.104.080424

C. Selvam, N. Oueslati, I. Lemasson, I. Brabet, D. Rigault et al., A Virtual Screening Hit Reveals New Possibilities for Developing Group III Metabotropic Glutamate Receptor Agonists, Journal of Medicinal Chemistry, vol.53, issue.7, pp.2797-813, 2010.
DOI : 10.1021/jm901523t

E. Kostenis, M. Waelbroeck, and G. Milligan, Techniques: Promiscuous G?? proteins in basic research and drug discovery, Trends in Pharmacological Sciences, vol.26, issue.11, pp.595-602, 2005.
DOI : 10.1016/j.tips.2005.09.007

D. Vivo and M. , [10] Assays for G-protein regulation of phospholipase C activity, Methods Enzymol, vol.238, pp.131-171, 1994.
DOI : 10.1016/0076-6879(94)38012-0

T. Mullinax, G. Henrich, P. Kasila, D. Ahern, E. Wenske et al., Monitoring Inositol-Specific Phospholipase C Activity Using a Phospholipid FlashPlate??, Journal of Biomolecular Screening, vol.288, issue.3, pp.151-156, 1999.
DOI : 10.1177/108705719900400309

C. Dangelmaier, J. Daniel, and J. Smith, Determination of basal and stimulated levels of inositol triphosphate in [32P]orthophosphate-labeled platelets, Analytical Biochemistry, vol.154, issue.2, pp.414-423, 1986.
DOI : 10.1016/0003-2697(86)90007-2

C. Barker and P. Berggren, The Role of Inositol and the Principles of Labelling, Extraction, and Analysis of Inositides in Mammalian Cells, Methods Mol Biol, vol.645, pp.1-19, 2010.
DOI : 10.1007/978-1-60327-175-2_1

K. Sauer, Y. Huang, H. Lin, M. Sandberg, and G. Mayr, Phosphoinositide and Inositol Phosphate Analysis in Lymphocyte Activation, Curr Protoc Immunol, vol.6, issue.11Unit11 1, 2009.
DOI : 10.1002/0471142735.im1101s87

R. Eglen, Functional G Protein-Coupled Receptor Assays for Primary and Secondary Screening, Combinatorial Chemistry & High Throughput Screening, vol.8, issue.4, pp.311-319, 2005.
DOI : 10.2174/1386207054020813

E. Trinquet, M. Fink, H. Bazin, F. Grillet, F. Maurin et al., d-myo-Inositol 1-phosphate as a surrogate of d-myo-inositol 1,4,5-tris phosphate to monitor G protein-coupled receptor activation, Analytical Biochemistry, vol.358, issue.1, pp.126-161, 2006.
DOI : 10.1016/j.ab.2006.08.002

URL : https://hal.archives-ouvertes.fr/inserm-00318996

R. Paredes, J. Etzler, L. Watts, W. Zheng, and J. Lechleiter, Chemical calcium indicators, Methods, vol.46, issue.3, pp.143-51, 2008.
DOI : 10.1016/j.ymeth.2008.09.025

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2666335

O. Shimomura, F. Johnson, and Y. Saiga, Extraction, Purification and Properties of Aequorin, a Bioluminescent Protein from the Luminous Hydromedusan,Aequorea, Journal of Cellular and Comparative Physiology, vol.5, issue.3, pp.223-262, 1962.
DOI : 10.1002/jcp.1030590302

P. Pinton, A. Rimessi, A. Romagnoli, A. Prandini, and R. Rizzuto, Biosensors for the Detection of Calcium and pH, Methods Cell Biol, vol.80, pp.297-325, 2007.
DOI : 10.1016/S0091-679X(06)80015-4

A. Miyawaki, J. Llopis, R. Heim, J. Mccaffery, J. Adams et al., Fluorescent indicators for Ca2 based on green fluorescent proteins and calmodulin +, Nature, vol.388, pp.882-889, 1997.

N. Demaurex, Calcium measurements in organelles with Ca2+-sensitive fluorescent proteins, Cell Calcium, vol.38, issue.3-4, pp.213-235, 2005.
DOI : 10.1016/j.ceca.2005.06.026

A. Palmer and R. Tsien, Measuring calcium signaling using genetically targetable fluorescent indicators, Nature Protocols, vol.327, issue.3, pp.1057-65, 2006.
DOI : 10.1038/nprot.2006.172

N. Riobo and D. Manning, Receptors coupled to heterotrimeric G proteins of the G12 family, Trends in Pharmacological Sciences, vol.26, issue.3, pp.146-54, 2005.
DOI : 10.1016/j.tips.2005.01.007

T. Worzfeld, N. Wettschureck, and O. S. , G12/G13-mediated signalling in mammalian physiology and disease, Trends in Pharmacological Sciences, vol.29, issue.11, pp.582-591, 2008.
DOI : 10.1016/j.tips.2008.08.002

J. Regard, H. Kataoka, D. Cano, E. Camerer, L. Yin et al., Probing cell type???specific functions of Gi in vivo identifies GPCR regulators of insulin secretion, Journal of Clinical Investigation, vol.117, pp.4034-4077, 2007.
DOI : 10.1172/JCI32994

T. Higashijima, S. Uzu, T. Nakajima, and E. Ross, Mastoparan, a peptide toxin from wasp venom, mimics receptors by activating GTP-binding regulatory proteins (G proteins), J Biol Chem, vol.263, pp.6491-6495, 1988.

C. Van-dop, M. Tsubokawa, H. Bourne, and J. Ramachandran, Amino acid sequence of retinal transducin at the site ADP-ribosylated by cholera toxin, J Biol Chem, vol.259, pp.696-704, 1984.

J. Northup, P. Sternweis, M. Smigel, L. Schleifer, E. Ross et al., Purification of the regulatory component of adenylate cyclase., Proceedings of the National Academy of Sciences, vol.77, issue.11, pp.6516-6536, 1980.
DOI : 10.1073/pnas.77.11.6516

M. Freissmuth, S. Boehm, W. Beindl, P. Nickel, A. Ijzerman et al., Suramin analogues as subtype-selective G protein inhibitors, Mol Pharmacol, vol.49, pp.602-613, 1996.

M. Hohenegger, M. Waldhoer, W. Beindl, B. Boing, A. Kreimeyer et al., Gs??-selective G protein antagonists, Proceedings of the National Academy of Sciences, vol.95, issue.1, pp.346-51, 1998.
DOI : 10.1073/pnas.95.1.346

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC18220

B. Wilson, X. Zhu, M. Ho, and L. Lu, Pasteurella multocida Toxin Activates the Inositol Triphosphate Signaling Pathway in Xenopus Oocytes via Gq??-coupled Phospholipase C-??1, Journal of Biological Chemistry, vol.272, issue.2, pp.1268-75, 1997.
DOI : 10.1074/jbc.272.2.1268

A. Zywietz, A. Gohla, M. Schmelz, G. Schultz, and S. Offermanns, Pleiotropic Effects of Pasteurella multocida Toxin Are Mediated by Gq-dependent and -independent Mechanisms: INVOLVEMENT OF Gq BUT NOT G11, Journal of Biological Chemistry, vol.276, issue.6, pp.3840-3845, 2001.
DOI : 10.1074/jbc.M007819200

B. Wilson and M. Ho, Pasteurella multocida toxin as a tool for studying Gq signal transduction, Rev Physiol Biochem Pharmacol, vol.276, pp.93-109, 2004.
DOI : 10.1007/s10254-004-0032-6

T. Kawasaki, M. Taniguchi, Y. Moritani, K. Hayashi, T. Saito et al., Antithrombotic and thrombolytic efficacy of YM-254890, a Gq/11 inhibitor, in a rat model of arterial thrombosis, Thrombosis and Haemostasis, vol.90, pp.406-419, 2003.
DOI : 10.1160/TH03-02-0115

A. Nishimura, K. Kitano, J. Takasaki, M. Taniguchi, N. Mizuno et al., Structural basis for the specific inhibition of heterotrimeric Gq protein by a small molecule, Proceedings of the National Academy of Sciences, vol.107, issue.31
DOI : 10.1073/pnas.1003553107

M. Ayoub, M. Damian, C. Gespach, E. Ferrandis, O. Lavergne et al., Inhibition of Heterotrimeric G Protein Signaling by a Small Molecule Acting on G?? Subunit, Journal of Biological Chemistry, vol.284, issue.42, pp.29136-29181, 2009.
DOI : 10.1074/jbc.M109.042333

M. Rasenick, M. Watanabe, M. Lazarevic, S. Hatta, and H. Hamm, Synthetic peptides as probes for G protein function. Carboxyl-terminal G alpha s peptides mimic Gs and evoke high affinity agonist binding to beta-adrenergic receptors, J Biol Chem, vol.269, pp.21519-21544, 1994.

A. Gilchrist, J. Vanhauwe, A. Li, T. Thomas, T. Voyno-yasenetskaya et al., G?? Minigenes Expressing C-terminal Peptides Serve as Specific Inhibitors of Thrombin-mediated Endothelial Activation, Journal of Biological Chemistry, vol.276, issue.28, pp.25672-25681, 2001.
DOI : 10.1074/jbc.M100914200

A. Gilchrist, A. Li, and H. Hamm, G alpha COOH-terminal minigene vectors dissect heterotrimeric G protein signaling, Sci STKE, p.1, 2002.
DOI : 10.1126/stke.2002.118.pl1

B. Atwood, J. Lopez, J. Wager-miller, K. Mackie, and A. Straiker, Expression of G protein-coupled receptors and related proteins in HEK293, AtT20, BV2, and N18 cell lines as revealed by microarray analysis, BMC Genomics, vol.13, issue.6B, p.14, 2011.
DOI : 10.1186/1471-2164-12-14

N. Wettschureck, A. Moers, and S. Offermanns, Mouse models to study G-protein-mediated signaling, Pharmacology & Therapeutics, vol.101, issue.1, pp.75-89, 2004.
DOI : 10.1016/j.pharmthera.2003.10.005

A. Krumins and A. Gilman, Targeted Knockdown of G Protein Subunits Selectively Prevents Receptor-mediated Modulation of Effectors and Reveals Complex Changes in Non-targeted Signaling Proteins, Journal of Biological Chemistry, vol.281, issue.15, pp.10250-62, 2006.
DOI : 10.1074/jbc.M511551200

G. Hilf, P. Gierschik, and K. Jakobs, Muscarinic acetylcholine receptor-stimulated binding of guanosine 5'-O-(3-thiotriphosphate) to guanine-nucleotide-binding proteins in cardiac membranes, European Journal of Biochemistry, vol.12, issue.5, pp.725-756, 1989.
DOI : 10.1006/abio.1976.9999

T. Wieland, K. Liedel, S. Kaldenberg-stasch, M. Zu-heringdorf, D. Schmidt et al., Analysis of receptor-G protein interactions in permeabilized cells, Naunyn-Schmiedeberg's Archives of Pharmacology, vol.351, issue.4
DOI : 10.1007/BF00169072

L. Sim, D. Selley, and S. Childers, In vitro autoradiography of receptor-activated G proteins in rat brain by agonist-stimulated guanylyl 5'-[gamma-[35S]thio]-triphosphate binding., Proceedings of the National Academy of Sciences, vol.92, issue.16, pp.7242-7248, 1995.
DOI : 10.1073/pnas.92.16.7242

C. Harrison and J. Traynor, The [35S]GTP??S binding assay: approaches and applications in pharmacology, Life Sciences, vol.74, issue.4, pp.489-508, 2003.
DOI : 10.1016/j.lfs.2003.07.005

A. Newman-tancredi, C. Conte, C. Chaput, L. Verriele, and M. Millan, Agonist and Inverse Agonist Efficacy at Human Recombinant Serotonin 5-HT1A Receptors as a Function of Receptor:G-protein Stoichiometry, Neuropharmacology, vol.36, issue.4-5, pp.451-460, 1997.
DOI : 10.1016/S0028-3908(97)00022-1

A. Barr, L. Brass, and D. Manning, Reconstitution of receptors and GTP-binding regulatory proteins (G proteins) in Sf9 cells. A direct evaluation of selectivity in receptor

R. Seifert, T. Lee, V. Lam, and B. Kobilka, Reconstitution of beta2-adrenoceptor-GTP-binding-protein interaction in Sf9 cells . High coupling efficiency in a beta2-adrenoceptor-Gsalpha fusion protein, European Journal of Biochemistry, vol.255, issue.2, pp.369-82, 1998.
DOI : 10.1046/j.1432-1327.1998.2550369.x

T. Kenakin, Ligand-selective receptor conformations revisited: the promise and the problem, Trends in Pharmacological Sciences, vol.24, issue.7, pp.346-54, 2003.
DOI : 10.1016/S0165-6147(03)00167-6

P. Pauwels, I. Rauly, T. Wurch, and F. Colpaert, Evidence for protean agonism of RX 831003 at ??2A-adrenoceptors by co-expression with different G?? protein subunits, Neuropharmacology, vol.42, issue.6, pp.855-63, 2002.
DOI : 10.1016/S0028-3908(01)00201-5

R. Windh, M. Lee, T. Hla, S. An, A. Barr et al., Differential coupling of the sphingosine 1-phosphate receptors Edg-1, Edg-3

A. Mclean, F. Zeng, D. Behan, D. Chalmers, and G. Milligan, Generation and Analysis of Constitutively Active and Physically Destabilized Mutants of the Human beta 1-Adrenoceptor, Molecular Pharmacology, vol.62, issue.3, pp.747-55, 2002.
DOI : 10.1124/mol.62.3.747

N. Delapp, The antibody-capture [35S]GTP??S scintillation proximity assay: a powerful emerging technique for analysis of GPCR pharmacology, Trends in Pharmacological Sciences, vol.25, issue.8, pp.400-401, 2004.
DOI : 10.1016/j.tips.2004.06.003

E. Johnson, X. Shi, J. Cassaday, M. Ferrer, B. Strulovici et al., i-Coupled GPCR, ASSAY and Drug Development Technologies, vol.6, issue.3, pp.327-364, 2008.
DOI : 10.1089/adt.2007.113

H. Frang, V. Mukkala, R. Syysto, P. Ollikka, P. Hurskainen et al., Nonradioactive GTP Binding Assay to Monitor Activation of G Protein-Coupled Receptors, ASSAY and Drug Development Technologies, vol.1, issue.2
DOI : 10.1089/15406580360545080

A. Koval, D. Kopein, V. Purvanov, and V. Katanaev, Europium-labeled GTP as a general nonradioactive substitute for [35S]GTP??S in high-throughput G protein studies, Analytical Biochemistry, vol.397, issue.2, pp.202-209, 2010.
DOI : 10.1016/j.ab.2009.10.028

D. Mcewen, K. Gee, H. Kang, and R. Neubig, Fluorescent BODIPY-GTP Analogs: Real-Time Measurement of Nucleotide Binding to G Proteins, Analytical Biochemistry, vol.291, issue.1, pp.109-126, 2001.
DOI : 10.1006/abio.2001.5011

E. Jameson, R. Roof, M. Whorton, H. Mosberg, R. Sunahara et al., Real-time Detection of Basal and Stimulated G Protein GTPase Activity Using Fluorescent GTP Analogues, Journal of Biological Chemistry, vol.280, issue.9, pp.7712-7721, 2005.
DOI : 10.1074/jbc.M413810200

B. Bertin, M. Freissmuth, R. Jockers, A. Strosberg, and S. Marullo, Cellular signaling by an agonist-activated receptor/Gs alpha fusion protein., Proceedings of the National Academy of Sciences, vol.91, issue.19, pp.8827-8858, 1994.
DOI : 10.1073/pnas.91.19.8827

A. Wise, I. Carr, and G. Milligan, fusion protein, Biochemical Journal, vol.325, issue.1, pp.17-21, 1997.
DOI : 10.1042/bj3250017

D. Massotte, K. Brillet, B. Kieffer, and G. Milligan, Agonists activate Gi1?? or Gi2?? fused to the human mu opioid receptor differently, Journal of Neurochemistry, vol.292, issue.6, pp.1372-82, 2002.
DOI : 10.1046/j.1471-4159.2002.00946.x

J. Lane, B. Powney, A. Wise, S. Rees, and G. Milligan, Protean Agonism at the Dopamine D2 Receptor: (S)-3-(3-Hydroxyphenyl)-N-propylpiperidine Is an Agonist for Activation of Go1 but an Antagonist/Inverse Agonist for Gi1,Gi2, and Gi3, Molecular Pharmacology, vol.71, issue.5, pp.1349-59, 2007.
DOI : 10.1124/mol.106.032722

K. Wenzel-seifert and R. Seifert, Molecular analysis of beta(2)-adrenoceptor coupling to G(s)-, G(i)-, and G(q)-proteins, Mol Pharmacol, vol.58, pp.954-66, 2000.

G. Milligan, G. Parenty, L. Stoddart, and J. Lane, Novel pharmacological applications of G-protein-coupled receptor???G protein fusions, Current Opinion in Pharmacology, vol.7, issue.5, pp.521-527, 2007.
DOI : 10.1016/j.coph.2007.06.007

G. Pascal and G. Milligan, Functional complementation and the analysis of opioid receptor homodimerization, Mol Pharmacol, vol.68, pp.905-920, 2005.

Y. Han, I. Moreira, E. Urizar, H. Weinstein, and J. Javitch, Allosteric communication between protomers of dopamine class A GPCR dimers modulates activation, Nature Chemical Biology, vol.455, issue.9, pp.688-95, 2009.
DOI : 10.1038/nchembio.199

Z. Salamon, H. Macleod, and G. Tollin, Coupled plasmon-waveguide resonators: a new spectroscopic tool for probing proteolipid film structure and properties, Biophysical Journal, vol.73, issue.5, pp.2791-2798, 1997.
DOI : 10.1016/S0006-3495(97)78308-5

G. Tollin, Z. Salamon, and V. Hruby, Techniques: Plasmon-waveguide resonance (PWR) spectroscopy as a tool to study ligand???GPCR interactions, Trends in Pharmacological Sciences, vol.24, issue.12, pp.655-664, 2003.
DOI : 10.1016/j.tips.2003.10.010

I. Alves, Z. Salamon, E. Varga, H. Yamamura, G. Tollin et al., Direct Observation of G-protein Binding to the Human ??-Opioid Receptor Using Plasmon-Waveguide Resonance Spectroscopy, Journal of Biological Chemistry, vol.278, issue.49, pp.48890-48897, 2003.
DOI : 10.1074/jbc.M306866200

Z. Salamon, G. Tollin, I. Alves, and V. Hruby, Chapter 6 Plasmon Resonance Methods in Membrane Protein Biology, Methods Enzymol, vol.461, pp.123-169, 2009.
DOI : 10.1016/S0076-6879(09)05406-8

I. Alves, D. Delaroche, B. Mouillac, Z. Salamon, G. Tollin et al., The Two NK-1 Binding Sites Correspond to Distinct, Independent, and Non-Interconvertible Receptor Conformational States As Confirmed by Plasmon-Waveguide Resonance Spectroscopy, Biochemistry, vol.45, issue.16, pp.5309-5327, 2006.
DOI : 10.1021/bi052586d

URL : https://hal.archives-ouvertes.fr/hal-00091764

T. Georgieva, S. Devanathan, D. Stropova, C. Park, Z. Salamon et al., Unique agonist-bound cannabinoid CB1 receptor conformations indicate agonist specificity in signaling, European Journal of Pharmacology, vol.581, issue.1-2, pp.19-29, 2008.
DOI : 10.1016/j.ejphar.2007.11.053

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2279194

N. Boute, R. Jockers, and T. Issad, The use of resonance energy transfer in high-throughput screening: BRET versus FRET, Trends in Pharmacological Sciences, vol.23, issue.8, pp.351-355, 2002.
DOI : 10.1016/S0165-6147(02)02062-X

K. Pfleger and K. Eidne, Monitoring the formation of dynamic G-protein-coupled receptor???protein complexes in living cells, Biochemical Journal, vol.385, issue.3, pp.625-662, 2005.
DOI : 10.1042/BJ20041361

H. Dacres, M. Dumancic, I. Horne, and S. Trowell, Direct comparison of bioluminescence-based resonance energy transfer methods for monitoring of proteolytic cleavage, Analytical Biochemistry, vol.385, issue.2, pp.194-202, 2009.
DOI : 10.1016/j.ab.2008.10.040

A. Loening, A. Wu, and S. Gambhir, Red-shifted Renilla reniformis luciferase variants for imaging in living subjects, Nature Methods, vol.81, issue.8, pp.641-644, 2007.
DOI : 10.1038/nmeth1070

A. De, A. Loening, and S. Gambhir, An Improved Bioluminescence Resonance Energy Transfer Strategy for Imaging Intracellular Events in Single Cells and Living Subjects, Cancer Research, vol.67, issue.15
DOI : 10.1158/0008-5472.CAN-06-4623

C. Gales, R. Rebois, M. Hogue, P. Trieu, A. Breit et al., Real-time monitoring of receptor and G-protein interactions in living cells, Nature Methods, vol.60, issue.3, pp.177-84, 2005.
DOI : 10.1073/pnas.1834247100

P. Hein, M. Frank, C. Hoffmann, M. Lohse, and M. Bunemann, Dynamics of receptor/G protein coupling in living cells, The EMBO Journal, vol.100, issue.23, pp.4106-4120, 2005.
DOI : 10.1038/sj.emboj.7600870

N. Audet, C. Gales, E. Archer-lahlou, M. Vallieres, P. Schiller et al., Bioluminescence Resonance Energy Transfer Assays Reveal Ligand-specific Conformational Changes within Preformed Signaling Complexes Containing ??-Opioid Receptors and Heterotrimeric G Proteins, Journal of Biological Chemistry, vol.283, issue.22, pp.15078-88, 2008.
DOI : 10.1074/jbc.M707941200

URL : https://hal.archives-ouvertes.fr/inserm-00408782

S. Galandrin, G. Oligny-longpre, H. Bonin, K. Ogawa, C. Gales et al., Conformational Rearrangements and Signaling Cascades Involved in Ligand-Biased Mitogen-Activated Protein Kinase Signaling through the ??1-Adrenergic Receptor, Molecular Pharmacology, vol.74, issue.1, pp.162-72, 2008.
DOI : 10.1124/mol.107.043893

M. Ayoub, D. Maurel, V. Binet, M. Fink, L. Prezeau et al., Real-Time Analysis of Agonist-Induced Activation of Protease-Activated Receptor 1/G??i1 Protein Complex Measured by Bioluminescence Resonance Energy Transfer in Living Cells, Molecular Pharmacology, vol.71, issue.5, pp.1329-1369, 2007.
DOI : 10.1124/mol.106.030304

M. Ayoub, E. Trinquet, K. Pfleger, and J. Pin, Differential association modes of the thrombin receptor PAR1 with G??i1, G??12, and ??-arrestin 1, The FASEB Journal, vol.24, issue.9, pp.3522-3557, 2010.
DOI : 10.1096/fj.10-154997

C. Janetopoulos, J. T. Devreotes, and P. , Receptor-Mediated Activation of Heterotrimeric G-Proteins in Living Cells, Science, vol.291, issue.5512, pp.2408-2419, 2001.
DOI : 10.1126/science.1055835

I. Azpiazu and N. Gautam, A Fluorescence Resonance Energy Transfer-based Sensor Indicates that Receptor Access to a G Protein Is Unrestricted in a Living Mammalian Cell, Journal of Biological Chemistry, vol.279, issue.26
DOI : 10.1074/jbc.M403712200

M. Frank, L. Thumer, M. Lohse, and M. G. Bunemann, G Protein Activation without Subunit Dissociation Depends on a G??i-specific Region, Journal of Biological Chemistry, vol.280, issue.26, pp.24584-90, 2005.
DOI : 10.1074/jbc.M414630200

S. Gibson and A. Gilman, Gi?? and G?? subunits both define selectivity of G protein activation by ??2-adrenergic receptors, Proceedings of the National Academy of Sciences, vol.103, issue.1, pp.212-219, 2006.
DOI : 10.1073/pnas.0509763102

M. Nobles, A. Benians, and A. Tinker, Heterotrimeric G proteins precouple with G protein-coupled receptors in living cells, Proceedings of the National Academy of Sciences, vol.102, issue.51, pp.18706-18717, 2005.
DOI : 10.1073/pnas.0504778102

M. Chisari, D. Saini, J. Cho, V. Kalyanaraman, and N. Gautam, G Protein Subunit Dissociation and Translocation Regulate Cellular Response to Receptor Stimulation, PLoS ONE, vol.296, issue.11, p.7797, 2009.
DOI : 10.1371/journal.pone.0007797.s007

URL : http://doi.org/10.1371/journal.pone.0007797

M. Lohse, J. Vilardaga, and M. Bunemann, Direct optical recording of intrinsic efficacy at a G protein-coupled receptor, Life Sciences, vol.74, issue.2-3, pp.397-404, 2003.
DOI : 10.1016/j.lfs.2003.09.026

Z. Gao and K. Jacobson, Translocation of arrestin induced by human A3 adenosine receptor ligands in an engineered cell line: Comparison with G protein-dependent pathways, Pharmacological Research, vol.57, issue.4, pp.303-314, 2008.
DOI : 10.1016/j.phrs.2008.02.008

E. Whalen, S. Rajagopal, and R. Lefkowitz, Therapeutic potential of ??-arrestin- and G protein-biased agonists, Trends in Molecular Medicine, vol.17, issue.3, pp.126-165, 2011.
DOI : 10.1016/j.molmed.2010.11.004

A. Bisello, M. Chorev, M. Rosenblatt, L. Monticelli, D. Mierke et al., Selective Ligand-induced Stabilization of Active and Desensitized Parathyroid Hormone Type 1 Receptor Conformations, Journal of Biological Chemistry, vol.277, issue.41, pp.38524-38554, 2002.
DOI : 10.1074/jbc.M202544200

V. Wehbi, J. Decourtye, V. Piketty, G. Durand, E. Reiter et al., Selective Modulation of Follicle-Stimulating Hormone Signaling Pathways with Enhancing Equine Chorionic Gonadotropin/Antibody Immune Complexes, Endocrinology, vol.151, issue.6, pp.2788-99, 2010.
DOI : 10.1210/en.2009-0892

URL : https://hal.archives-ouvertes.fr/hal-01129424

M. Rafei, Y. Berchiche, E. Birman, M. Boivin, Y. Young et al., An Engineered GM-CSF-CCL2 Fusokine Is a Potent Inhibitor of CCR2-Driven Inflammation As Demonstrated in a Murine Model of Inflammatory Arthritis, The Journal of Immunology, vol.183, issue.3, pp.1759-66, 2009.
DOI : 10.4049/jimmunol.0900523

R. Jorgensen, V. Kubale, M. Vrecl, T. Schwartz, and C. Elling, Oxyntomodulin Differentially Affects Glucagon-Like Peptide-1 Receptor beta-Arrestin Recruitment and Signaling through G??, Journal of Pharmacology and Experimental Therapeutics, vol.322, issue.1, pp.148-54, 2007.
DOI : 10.1124/jpet.107.120006

A. Reversi, V. Rimoldi, T. Marrocco, P. Cassoni, G. Bussolati et al., The Oxytocin Receptor Antagonist Atosiban Inhibits Cell Growth via a "Biased Agonist" Mechanism, Journal of Biological Chemistry, vol.280, issue.16, pp.16311-16319, 2005.
DOI : 10.1074/jbc.M409945200

S. Sensken, C. Staubert, P. Keul, B. Levkau, T. Schoneberg et al., Selective activation of G alpha i mediated signalling of S1P3 by FTY720-phosphate, Cellular Signalling, vol.20, issue.6, pp.1125-1158, 2008.
DOI : 10.1016/j.cellsig.2008.01.019

S. Rajagopal, K. Rajagopal, and R. Lefkowitz, Teaching old receptors new tricks: biasing seven-transmembrane receptors, Nature Reviews Drug Discovery, vol.28, issue.5, pp.373-86, 2010.
DOI : 10.1038/nrd3024

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2902265

H. Wei, S. Ahn, S. Shenoy, S. Karnik, L. Hunyady et al., Independent ??-arrestin 2 and G protein-mediated pathways for angiotensin II activation of extracellular signal-regulated kinases 1 and 2, Proceedings of the National Academy of Sciences, vol.100, issue.19, pp.10782-10789, 2003.
DOI : 10.1073/pnas.1834556100

I. Kim, D. Tilley, J. Chen, N. Salazar, E. Whalen et al., ??-Blockers alprenolol and carvedilol stimulate ??-arrestin-mediated EGFR transactivation, Proceedings of the National Academy of Sciences, vol.105, issue.38, pp.14555-60, 2008.
DOI : 10.1073/pnas.0804745105

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2567217

J. Wisler, S. Dewire, E. Whalen, J. Violin, M. Drake et al., A unique mechanism of beta-blocker action: Carvedilol stimulates beta-arrestin signaling, Proceedings of the National Academy of Sciences, vol.104, issue.42, pp.16657-62, 2007.
DOI : 10.1073/pnas.0707936104

S. Rajagopal, J. Kim, S. Ahn, S. Craig, C. Lam et al., ??-arrestin- but not G protein-mediated signaling by the "decoy" receptor CXCR7, Proceedings of the National Academy of Sciences, vol.107, issue.2, pp.628-660, 2010.
DOI : 10.1073/pnas.0912852107

F. Verkaar, J. Van-rosmalen, M. Blomenrohr, C. Van-koppen, W. Blankesteijn et al., G protein-independent cell-based assays for drug discovery on seven-transmembrane receptors, Biotechnol Annu Rev, vol.14, pp.253-74, 2008.
DOI : 10.1016/S1387-2656(08)00010-0

M. Drake, J. Violin, E. Whalen, J. Wisler, S. Shenoy et al., ??-Arrestin-biased Agonism at the ??2-Adrenergic Receptor, Journal of Biological Chemistry, vol.283, issue.9, pp.5669-76, 2008.
DOI : 10.1074/jbc.M708118200

J. Hansen, M. Aplin, J. Hansen, G. Christensen, M. Bonde et al., The human angiotensin AT1 receptor supports G protein-independent extracellular signal-regulated kinase 1/2 activation and cellular proliferation, European Journal of Pharmacology, vol.590, issue.1-3, pp.255-63, 2008.
DOI : 10.1016/j.ejphar.2008.05.010

K. Luker, M. Gupta, and G. Luker, Imaging CXCR4 Signaling with Firefly Luciferase Complementation, Analytical Chemistry, vol.80, issue.14, pp.5565-73, 2008.
DOI : 10.1021/ac8005457

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC5258185

M. Van-der-lee, M. Bras, C. Van-koppen, and G. Zaman, ??-Arrestin Recruitment Assay for the Identification of Agonists of the Sphingosine 1-Phosphate Receptor EDG1, Journal of Biomolecular Screening, vol.279, issue.1, pp.986-98, 2008.
DOI : 10.1177/1087057108326144

P. Charest, S. Terrillon, and M. Bouvier, Monitoring agonist-promoted conformational changes of ??-arrestin in living cells by intramolecular BRET, EMBO reports, vol.21, issue.4, pp.334-374, 2005.
DOI : 10.1038/nrm976

A. Shukla, J. Violin, E. Whalen, D. Gesty-palmer, S. Shenoy et al., Distinct conformational changes in ??-arrestin report biased agonism at seven-transmembrane receptors, Proceedings of the National Academy of Sciences, vol.105, issue.29, pp.9988-93, 2008.
DOI : 10.1073/pnas.0804246105

L. Luttrell, S. Ferguson, Y. Daaka, W. Miller, S. Maudsley et al., Adrenergic Receptor-Src Protein Kinase Complexes, Science, vol.283, issue.5402, pp.655-61, 1999.
DOI : 10.1126/science.283.5402.655

S. Ahn, S. Shenoy, H. Wei, and R. Lefkowitz, Differential Kinetic and Spatial Patterns of ??-Arrestin and G Protein-mediated ERK Activation by the Angiotensin II Receptor, Journal of Biological Chemistry, vol.279, issue.34, pp.35518-35543, 2004.
DOI : 10.1074/jbc.M405878200

G. Callander, W. Thomas, and R. Bathgate, Prolonged RXFP1 and RXFP2 signaling can be explained by poor internalization and a lack of beta-arrestin recruitment

G. Milligan, Principles: Extending the utility of [35S]GTP??S binding assays, Trends in Pharmacological Sciences, vol.24, issue.2, pp.87-90, 2003.
DOI : 10.1016/S0165-6147(02)00027-5

M. Leduc, B. Breton, C. Gales, L. Gouill, C. Bouvier et al., Functional Selectivity of Natural and Synthetic Prostaglandin EP4 Receptor Ligands, Journal of Pharmacology and Experimental Therapeutics, vol.331, issue.1, pp.297-307, 2009.
DOI : 10.1124/jpet.109.156398

S. Galandrin and M. Bouvier, Distinct Signaling Profiles of beta1 and beta2 Adrenergic Receptor Ligands toward Adenylyl Cyclase and Mitogen-Activated Protein Kinase Reveals the Pluridimensionality of Efficacy, Molecular Pharmacology, vol.70, issue.5, pp.1575-84, 2006.
DOI : 10.1124/mol.106.026716