D. A. Arias, J. Navenot, W. Zhang, J. Broach, and S. C. Peiper, Constitutive Activation of CCR5 and CCR2 Induced by Conformational Changes in the Conserved TXP Motif in Transmembrane Helix 2, J. Biol. Chem, vol.278, pp.36513-36521, 2003.

Y. Artemenko, T. J. Lampert, and P. N. Devreotes, Moving towards a paradigm: common mechanisms of chemotactic signaling in Dictyostelium and mammalian leukocytes, Cell. Mol. Life Sci, vol.71, pp.3711-3747, 2014.

N. Audet, I. Charfi, O. Mnie-filali, M. Amraei, A. Chabot-doré et al., Differential association of receptor-G?? complexes with ?-arrestin2 determines recycling bias and potential for tolerance of ? opioid receptor agonists, J. Neurosci, vol.32, pp.4827-4840, 2012.

J. A. Ballesteros and H. Weinstein, Integrated methods for the construction of threedimensional models and computational probing of structure-function relations in G protein-coupled receptors, Methods in Neurosciences, Receptor Molecular Biology, pp.366-428, 1995.

S. D. Bianco, L. Vandepas, M. Correa-medina, B. Gereben, A. Mukherjee et al., KISS1R intracellular trafficking and degradation: effect of the Arg386Pro disease-associated mutation, Endocrinology, vol.152, pp.1616-1626, 2011.

T. K. Bjarnadóttir, D. E. Gloriam, S. H. Hellstrand, H. Kristiansson, R. Fredriksson et al., Comprehensive repertoire and phylogenetic analysis of the G protein-coupled receptors in human and mouse, Genomics, vol.88, pp.263-273, 2006.

H. Blaser, M. Reichman-fried, I. Castanon, K. Dumstrei, F. L. Marlow et al., Migration of zebrafish primordial germ cells: a role for myosin contraction and cytoplasmic flow, Dev. Cell, vol.11, pp.613-627, 2006.

J. J. Bravo-cordero, L. Hodgson, and J. Condeelis, Directed cell invasion and migration during metastasis, Curr. Opin. Cell Biol, vol.24, pp.277-283, 2012.

C. Brulé, N. Perzo, J. Joubert, X. Sainsily, R. Leduc et al., Biased signaling regulates the pleiotropic effects of the urotensin II receptor to modulate its cellular behaviors, FASEB J, vol.28, pp.5148-5162, 2014.

J. M. Busillo, S. Armando, R. Sengupta, O. Meucci, M. Bouvier et al., Sitespecific phosphorylation of CXCR4 is dynamically regulated by multiple kinases and results in differential modulation of CXCR4 signaling, J. Biol. Chem, vol.285, pp.7805-7817, 2010.

J. M. Busillo and J. L. Benovic, Regulation of CXCR4 signaling, Biochim. Biophys. Acta, vol.1768, pp.952-963, 2007.

X. Cao, J. Yan, S. Shu, J. A. Brzostowski, and T. Jin, Arrestins function in cAR1 GPCRmediated signaling and cAR1 internalization in the development of Dictyostelium discoideum, Mol. Biol. Cell, vol.25, pp.3210-3221, 2014.

J. Cartailler and H. Luecke, Structural and functional characterization of pi bulges and other short intrahelical deformations, Structure, vol.12, pp.133-144, 2004.

L. B. Case, M. A. Baird, G. Shtengel, S. L. Campbell, H. F. Hess et al., Molecular mechanism of vinculin activation and nanoscale spatial organization in focal adhesions, Nat Cell Biol, vol.17, pp.880-892, 2015.

J. P. Castaño, A. J. Martínez-fuentes, E. Gutiérrez-pascual, H. Vaudry, M. Tena-sempere et al., Intracellular signaling pathways activated by kisspeptins through GPR54: do multiple signals underlie function diversity? Peptides, vol.30, pp.10-15, 2009.

M. J. Caterina, P. N. Devreotes, J. Borleis, and D. Hereld, Agonist-induced loss of ligand binding is correlated with phosphorylation of cAR1, a G protein-coupled chemoattractant receptor from Dictyostelium, J. Biol. Chem, vol.270, pp.8667-8672, 1995.

M. Chabbert, H. Castel, J. Pele, J. Deville, R. Legendre et al., Evolution of class A G-protein-coupled receptors: implications for molecular modeling, Curr. Med. Chem, vol.19, pp.1110-1118, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02286182

V. Chantreau, B. Taddese, M. Munier, L. Gourdin, D. Henrion et al., Molecular Insights into the Transmembrane Domain of the Thyrotropin Receptor, PLOS ONE, vol.10, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01397205

V. Cherezov, D. M. Rosenbaum, M. A. Hanson, S. G. Rasmussen, F. S. Thian et al., Highresolution crystal structure of an engineered human beta2-adrenergic G protein-coupled receptor, Science, vol.318, pp.1258-1265, 2007.

H. Chikumi, J. Vázquez-prado, J. Servitja, H. Miyazaki, and J. S. Gutkind, Potent Activation of RhoA by G?q and Gq-coupled Receptors, J. Biol. Chem, vol.277, pp.27130-27134, 2002.

S. Cho, D. Li, K. Tan, S. K. Siwko, and M. Liu, KiSS1 and its G-protein-coupled receptor GPR54 in cancer development and metastasis, Cancer Metastasis Rev, vol.31, pp.585-591, 2012.

C. K. Choi, M. Vicente-manzanares, J. Zareno, L. A. Whitmore, A. Mogilner et al., Actin and alpha-actinin orchestrate the assembly and maturation of nascent adhesions in a myosin II motor-independent manner, Nat. Cell Biol, vol.10, pp.1039-1050, 2008.

C. K. Choi, J. Zareno, M. A. Digman, E. Gratton, and A. R. Horwitz, Cross-correlated fluctuation analysis reveals phosphorylation-regulated paxillin-FAK complexes in nascent adhesions, Biophys. J, vol.100, pp.583-592, 2011.

P. Coly, N. Perzo, V. Le-joncour, C. Lecointre, M. Schouft et al., Chemotactic G protein-coupled receptors control cell migration by repressing autophagosome biogenesis, Autophagy, vol.0, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02345740

R. L. Contento, B. Molon, C. Boularan, T. Pozzan, S. Manes et al., CXCR4-CCR5: a couple modulating T cell functions, Proc. Natl. Acad. Sci. U.S.A, vol.105, pp.10101-10106, 2008.

D. Cvetkovic, M. Dragan, S. J. Leith, Z. M. Mir, H. S. Leong et al., KISS1R induces invasiveness of estrogen receptor-negative human mammary epithelial and breast cancer cells, Endocrinology, vol.154, pp.1999-2014, 2013.

Y. Daaka, L. M. Luttrell, and R. J. Lefkowitz, Switching of the coupling of the beta2-adrenergic receptor to different G proteins by protein kinase A, Nature, vol.390, pp.88-91, 1997.

J. A. Dalton, I. Lans, and J. Giraldo, Quantifying conformational changes in GPCRs: glimpse of a common functional mechanism, BMC Bioinformatics, vol.16, 2015.

K. A. Defea, Stop that cell! Beta-arrestin-dependent chemotaxis: a tale of localized actin assembly and receptor desensitization, Annu. Rev. Physiol, vol.69, pp.535-560, 2007.

X. Deupi, N. Dölker, M. L. López-rodríguez, M. Campillo, J. A. Ballesteros et al., Structural models of class a G protein-coupled receptors as a tool for drug design: insights on transmembrane bundle plasticity, Curr Top Med Chem, vol.7, pp.991-998, 2007.

X. Deupi and B. K. Kobilka, Energy Landscapes as a Tool to Integrate GPCR Structure, Dynamics, and Function, Physiology, vol.25, pp.293-303, 2010.

J. Devillé, J. Rey, and M. Chabbert, An indel in transmembrane helix 2 helps to trace the molecular evolution of class A G-protein-coupled receptors, J. Mol. Evol, vol.68, pp.475-489, 2009.

G. Fenalti, P. M. Giguere, V. Katritch, X. Huang, A. A. Thompson et al., Molecular control of ?-opioid receptor signalling, Nature, vol.506, pp.191-196, 2014.

L. Ge, S. K. Shenoy, R. J. Lefkowitz, and K. Defea, Constitutive protease-activated receptor-2-mediated migration of MDA MB-231 breast cancer cells requires both betaarrestin-1 and -2, J. Biol. Chem, vol.279, pp.55419-55424, 2004.

B. Geiger and K. M. Yamada, Molecular architecture and function of matrix adhesions, Cold Spring Harb Perspect Biol, vol.3, 2011.

A. M. Glodek, M. Honczarenko, Y. Le, J. J. Campbell, and L. E. Silberstein, Sustained activation of cell adhesion is a differentially regulated process in B lymphopoiesis, J. Exp. Med, vol.197, pp.461-473, 2003.

A. Göblyös and A. P. Ijzerman, Allosteric modulation of adenosine receptors, Biochim. Biophys. Acta, vol.1808, pp.1309-1318, 2011.

C. Govaerts, C. Blanpain, X. Deupi, S. Ballet, J. A. Ballesteros et al., The TXP motif in the second transmembrane helix of CCR5. A structural determinant of chemokine-induced activation, J. Biol. Chem, vol.276, pp.13217-13225, 2001.

C. Govaerts, A. Bondue, J. Springael, M. Olivella, X. Deupi et al., Activation of CCR5 by Chemokines Involves an Aromatic Cluster between Transmembrane Helices 2 and 3, J. Biol. Chem, vol.278, pp.1892-1903, 2003.

K. O. Gradel, M. Nørgaard, H. C. Schønheyder, C. Dethlefsen, T. Ejlertsen et al., Salmonella or Campylobacter gastroenteritis prior to a cancer diagnosis does not aggravate the prognosis: a population-based follow-up study, APMIS, vol.118, pp.136-142, 2010.

P. Grieco, R. Franco, G. Bozzuto, L. Toccacieli, A. Sgambato et al., Urotensin II receptor predicts the clinical outcome of prostate cancer patients and is involved in the regulation of motility of prostate adenocarcinoma cells, J. Cell. Biochem, vol.112, pp.341-353, 2011.

H. Gutiérrez-de-terán, A. Massink, D. Rodríguez, W. Liu, G. W. Han et al., The role of a sodium ion binding site in the allosteric modulation, 2013.

R. Horuk, Chemokines beyond inflammation, Nature, vol.393, pp.524-525, 1998.

X. Huang, J. Shen, M. Cui, L. Shen, X. Luo et al., Molecular dynamics simulations on SDF-1alpha: binding with CXCR4 receptor, Biophys. J, vol.84, pp.171-184, 2003.

D. L. Hunton, W. G. Barnes, J. Kim, X. Ren, J. D. Violin et al., Beta-arrestin 2-dependent angiotensin II type 1A receptormediated pathway of chemotaxis, Mol. Pharmacol, vol.67, pp.1229-1236, 2005.

M. Iijima and P. Devreotes, Tumor suppressor PTEN mediates sensing of chemoattractant gradients, Cell, vol.109, pp.599-610, 2002.

R. H. Insall, R. D. Soede, P. Schaap, and P. N. Devreotes, Two cAMP receptors activate common signaling pathways in Dictyostelium, Mol. Biol. Cell, vol.5, pp.703-711, 1994.

A. A. Jarjour, M. Durko, T. L. Luk, N. Marçal, M. Shekarabi et al., Autocrine netrin function inhibits glioma cell motility and promotes focal adhesion formation, PLoS ONE, vol.6, 2011.

M. Jarry, M. Diallo, C. Lecointre, L. Desrues, T. Tokay et al., The vasoactive peptides urotensin II and urotensin II-related peptide regulate astrocyte activity through common and distinct mechanisms: involvement in cell proliferation, Biochem. J, vol.428, pp.113-124, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00479207

K. Ji, L. Ye, M. D. Mason, and W. G. Jiang, The Kiss-1/Kiss-1R complex as a negative regulator of cell motility and cancer metastasis (Review), Int. J. Mol. Med, vol.32, pp.747-754, 2013.

T. H. Ji, M. Grossmann, and I. Ji, G protein-coupled receptors. I. Diversity of receptorligand interactions, J. Biol. Chem, vol.273, pp.17299-17302, 1998.

T. Jiang, S. Zhang, B. Lin, L. Meng, and H. Gao, Expression and clinical significance of KISS-1 and GPR54 mRNA in endometrial carcinoma, 2005.

, Zhonghua Zhong Liu Za Zhi, vol.27, pp.229-231

T. Jin, X. Xu, and D. Hereld, Chemotaxis, chemokine receptors and human disease, Cytokine, vol.44, pp.1-8, 2008.

H. Katoh, J. Aoki, Y. Yamaguchi, Y. Kitano, A. Ichikawa et al., Constitutively Active G?12, G?13, and G?q Induce Rho-dependent Neurite Retraction through Different Signaling Pathways, J. Biol. Chem, vol.273, pp.28700-28707, 1998.

C. M. Kenific, S. J. Stehbens, J. Goldsmith, A. M. Leidal, N. Faure et al., NBR1 enables autophagy-dependent focal adhesion turnover, J. Cell Biol, vol.212, pp.577-590, 2016.

L. F. Kolakowski, GCRDb: a G-protein-coupled receptor database, Recept. Channels, vol.2, pp.1-7, 1994.

M. Kotani, M. Detheux, A. Vandenbogaerde, D. Communi, J. M. Vanderwinden et al., The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54, J. Biol. Chem, vol.276, pp.34631-34636, 2001.

C. Lawson, S. Lim, S. Uryu, X. L. Chen, D. A. Calderwood et al., FAK promotes recruitment of talin to nascent adhesions to control cell motility, J. Cell Biol, vol.196, pp.223-232, 2012.

C. Lecointre, L. Desrues, J. E. Joubert, N. Perzo, P. Guichet et al., Signaling switch of the urotensin II vasosactive peptide GPCR: prototypic chemotaxic mechanism in glioma, Oncogene, vol.34, pp.5080-5094, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02367872

J. H. Lee, M. E. Miele, D. J. Hicks, K. K. Phillips, J. M. Trent et al., KiSS-1, a novel human malignant melanoma metastasis-suppressor gene, J. Natl. Cancer Inst, vol.88, pp.1731-1737, 1996.

J. Lee and D. R. Welch, Identification of highly expressed genes in metastasissuppressed chromosome 6/human malignant melanoma hybrid cells using subtractive hybridization and differential display, Int. J. Cancer, vol.71, pp.1035-1044, 1997.

L. Leloup, H. Shao, Y. H. Bae, B. Deasy, D. Stolz et al., 2010. m-Calpain activation is regulated by its membrane localization and by its binding to phosphatidylinositol 4,5-bisphosphate, J. Biol. Chem, vol.285, pp.33549-33566

A. Levoye, K. Balabanian, F. Baleux, F. Bachelerie, and B. Lagane, CXCR7 heterodimerizes with CXCR4 and regulates CXCL12-mediated G protein signaling, Blood, vol.113, pp.6085-6093, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-00415320

Q. Li, A. Roa, I. J. Clarke, and J. T. Smith, Seasonal variation in the gonadotropinreleasing hormone response to kisspeptin in sheep: possible kisspeptin regulation of the kisspeptin receptor, Neuroendocrinology, vol.96, pp.212-221, 2012.

H. Lin, M. F. Sassano, B. L. Roth, and B. K. Shoichet, A pharmacological organization of G protein-coupled receptors, Nat. Methods, vol.10, pp.140-146, 2013.

J. Liu, Y. Zhao, Y. Sun, B. He, C. Yang et al., Exo70 stimulates the Arp2/3 complex for lamellipodia formation and directional cell migration, Curr. Biol, vol.22, pp.1510-1515, 2012.

F. Magnani, Y. Shibata, M. J. Serrano-vega, and C. G. Tate, Co-evolving stability and conformational homogeneity of the human adenosine A2a receptor, Proc. Natl. Acad. Sci. U.S.A, vol.105, pp.10744-10749, 2008.

M. Mellado, J. M. Rodríguez-frade, A. J. Vila-coro, S. Fernández, A. Martín-de-ana et al., Chemokine receptor homo-or heterodimerization activates distinct signaling pathways, EMBO J, vol.20, pp.2497-2507, 2001.

N. Mittal, K. Roberts, K. Pal, L. A. Bentolila, E. Fultz et al., Select G-protein-coupled receptors modulate agonist-induced signaling via a ROCK, LIMK, and ?-arrestin 1 pathway, Cell Rep, vol.5, pp.1010-1021, 2013.

S. Miura and S. S. Karnik, Constitutive Activation of Angiotensin II Type 1 Receptor Alters the Orientation of Transmembrane Helix-2, J. Biol. Chem, vol.277, pp.24299-24305, 2002.

S. Miura, J. Zhang, J. Boros, and S. S. Karnik, TM2-TM7 Interaction in Coupling Movement of Transmembrane Helices to Activation of the Angiotensin II Type-1, 2003.

, Receptor. J. Biol. Chem, vol.278, pp.3720-3725

M. Monterrubio, M. Mellado, A. C. Carrera, and J. M. Rodríguez-frade, PI3Kgamma activation by CXCL12 regulates tumor cell adhesion and invasion, Biochem. Biophys. Res. Commun, vol.388, pp.199-204, 2009.

J. Navenot, N. Fujii, and S. C. Peiper, KiSS1 Metastasis Suppressor Gene Product Induces Suppression of Tyrosine Kinase Receptor Signaling to Akt, Tumor Necrosis Factor Family Ligand Expression, and Apoptosis, Mol Pharmacol, vol.75, pp.1074-1083, 2009.

T. Ohtaki, Y. Shintani, S. Honda, H. Matsumoto, A. Hori et al., Metastasis suppressor gene KiSS-1 encodes peptide ligand of a G-protein-coupled receptor, Nature, vol.411, pp.613-617, 2001.

M. Pampillo, N. Camuso, J. E. Taylor, J. M. Szereszewski, M. R. Ahow et al., Regulation of GPR54 signaling by GRK2 and {beta}-arrestin, Mol. Endocrinol, vol.23, pp.2060-2074, 2009.

M. S. Parker, Y. Y. Wong, and S. L. Parker, An ion-responsive motif in the second transmembrane segment of rhodopsin-like receptors, Amino Acids, vol.35, pp.1-15, 2008.

J. Pelé, H. Abdi, M. Moreau, D. Thybert, and M. Chabbert, Multidimensional scaling reveals the main evolutionary pathways of class A G-protein-coupled receptors, PLoS ONE, vol.6, 2011.

R. J. Petrie, A. D. Doyle, and K. M. Yamada, Random versus directionally persistent cell migration, Nat. Rev. Mol. Cell Biol, vol.10, pp.538-549, 2009.

T. D. Pollard and G. G. Borisy, Cellular motility driven by assembly and disassembly of actin filaments, Cell, vol.112, pp.453-465, 2003.

T. Pozzobon, G. Goldoni, A. Viola, and B. Molon, CXCR4 signaling in health and disease, Immunol. Lett, vol.177, pp.6-15, 2016.

V. V. Prabhu, K. M. Sakthivel, and C. Guruvayoorappan, Kisspeptins (KiSS-1): essential players in suppressing tumor metastasis. Asian Pac, J. Cancer Prev, vol.14, pp.6215-6220, 2013.

A. A. Pradhan, J. Perroy, W. M. Walwyn, M. L. Smith, A. Vicente-sanchez et al., Agonist-Specific Recruitment of Arrestin Isoforms Differentially Modify Delta Opioid Receptor Function, J. Neurosci, vol.36, pp.3541-3551, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01936373

S. G. Rasmussen, H. Choi, D. M. Rosenbaum, T. S. Kobilka, F. S. Thian et al., Crystal structure of the human ?2 adrenergic G-protein-coupled receptor, Nature, vol.450, pp.383-387, 2007.

R. I. Reis, E. L. Santos, J. B. Pesquero, L. Oliveira, J. P. Schanstra et al., Participation of transmembrane proline 82 in angiotensin II AT1 receptor signal transduction, Regulatory Peptides, vol.140, pp.32-36, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00409738

X. Ren, E. Reiter, S. Ahn, J. Kim, W. Chen et al., Different G protein-coupled receptor kinases govern G protein and beta-arrestin-mediated signaling of V2 vasopressin receptor, Proc. Natl. Acad. Sci. U.S.A, vol.102, pp.1448-1453, 2005.

X. Ren, E. Reiter, S. Ahn, J. Kim, W. Chen et al., Different G protein-coupled receptor kinases govern G protein and beta-arrestin-mediated signaling of V2 vasopressin receptor, Proc. Natl. Acad. Sci. U.S.A, vol.102, pp.1448-1453, 2005.

J. Rey, J. Deville, and M. Chabbert, Structural determinants stabilizing helical distortions related to proline, J. Struct. Biol, vol.171, pp.266-276, 2010.

B. E. Richardson and R. Lehmann, Mechanisms guiding primordial germ cell migration: strategies from different organisms, Nat. Rev. Mol. Cell Biol, vol.11, pp.37-49, 2010.

W. J. Rossowski, B. Cheng, J. E. Taylor, R. Datta, and D. H. Coy, Human urotensin IIinduced aorta ring contractions are mediated by protein kinase C, tyrosine kinases and Rho-kinase: inhibition by somatostatin receptor antagonists, Eur. J. Pharmacol, vol.438, pp.159-170, 2002.

E. T. Roussos, J. S. Condeelis, and A. Patsialou, Chemotaxis in cancer, Nat. Rev. Cancer, vol.11, pp.573-587, 2011.

J. B. Rubin, Chemokine Signaling in Cancer: One Hump or Two? Seminars in cancer biology 19, p.116, 2009.

C. D. Sadik and A. D. Luster, Lipid-cytokine-chemokine cascades orchestrate leukocyte recruitment in inflammation, J. Leukoc. Biol, vol.91, pp.207-215, 2012.

J. Segain, M. Rolli-derkinderen, N. Gervois, D. Raingeard-de-la-blétière, G. Loirand et al., Urotensin II is a new chemotactic factor for UT receptor-expressing monocytes, J. Immunol, vol.179, pp.901-909, 2007.

A. Van-keymeulen, K. Wong, Z. A. Knight, C. Govaerts, K. M. Hahn et al., To stabilize neutrophil polarity, PIP3 and Cdc42 augment RhoA activity at the back as well as signals at the front, J. Cell Biol, vol.174, pp.437-445, 2006.

S. Vogt, R. Grosse, G. Schultz, and S. Offermanns, Receptor-dependent RhoA Activation in G12/G13-deficient Cells GENETIC EVIDENCE FOR AN INVOLVEMENT OF Gq/G11, J. Biol. Chem, vol.278, pp.28743-28749, 2003.

S. Wang, J. Hong, C. Hsueh, and C. Chiang, Tumor-secreted SDF-1 promotes glioma invasiveness and TAM tropism toward hypoxia in a murine astrocytoma model, Lab. Invest, vol.92, pp.151-162, 2012.

T. Warne, M. J. Serrano-vega, J. G. Baker, R. Moukhametzianov, P. C. Edwards et al., Structure of a ?1-adrenergic G protein-coupled receptor, Nature, vol.454, pp.486-491, 2008.

D. J. Webb, K. Donais, L. A. Whitmore, S. M. Thomas, C. E. Turner et al., FAK-Src signalling through paxillin, ERK and MLCK regulates adhesion disassembly, Nat. Cell Biol, vol.6, pp.154-161, 2004.

D. J. Webb, J. T. Parsons, and A. F. Horwitz, Adhesion assembly, disassembly and turnover in migrating cells --over and over and over again, Nat. Cell Biol, vol.4, pp.97-100, 2002.

O. D. Weiner, W. A. Marganski, L. F. Wu, S. J. Altschuler, and M. W. Kirschner, An actinbased wave generator organizes cell motility, PLoS Biol, vol.5, p.221, 2007.

H. Wolfenson, I. Lavelin, and B. Geiger, Dynamic regulation of the structure and functions of integrin adhesions, Dev. Cell, vol.24, pp.447-458, 2013.

B. Wu, E. Y. Chien, C. D. Mol, G. Fenalti, W. Liu et al., Structures of the CXCR4 chemokine GPCR with small-molecule and cyclic peptide antagonists, Science, vol.330, pp.1066-1071, 2010.

Z. Xiao, N. Zhang, D. B. Murphy, and P. N. Devreotes, Dynamic distribution of chemoattractant receptors in living cells during chemotaxis and persistent stimulation, J. Cell Biol, vol.139, pp.365-374, 1997.

Y. Xiong, C. Kabacoff, J. Franca-koh, P. N. Devreotes, D. N. Robinson et al., Automated characterization of cell shape changes during amoeboid motility by skeletonization, BMC Syst Biol, vol.4, p.33, 2010.

, Références bibliographiques -A

A. Adebiyi, RGS2 regulates urotensin II-induced intracellular Ca2+ elevation and contraction in glomerular mesangial cells, J Cell Physiol, vol.229, pp.502-511, 2014.

R. P. Ahlquist, A study of the adrenotropic receptors, Am. J. Physiol, vol.153, pp.586-600, 1948.

A. Aiuti, I. J. Webb, C. Bleul, T. Springer, and J. C. Gutierrez-ramos, The Chemokine SDF-1 Is a Chemoattractant for Human CD34+ Hematopoietic Progenitor Cells and Provides a New Mechanism to Explain the Mobilization of CD34+ Progenitors to Peripheral Blood, J. Exp. Med, vol.185, p.111, 1997.

G. Albertin, V. Casale, A. Ziolkowska, R. Spinazzi, L. K. Malendowicz et al., Urotensin-II and UII-receptor expression and function in the rat adrenal cortex, International journal of molecular medicine, vol.17, pp.1111-1115, 2006.

R. S. Ames, H. M. Sarau, J. K. Chambers, R. N. Willette, N. V. Aiyar et al., Human urotensin-II is a potent vasoconstrictor and agonist for the orphan receptor GPR14, Nature, vol.401, pp.282-286, 1999.

H. Andersson, A. M. D'antona, D. A. Kendall, G. Von-heijne, and C. Chin, Membrane assembly of the cannabinoid receptor 1: impact of a long N-terminal tail, Mol. Pharmacol, vol.64, pp.570-577, 2003.

J. Andreev, M. L. Galisteo, O. Kranenburg, S. K. Logan, E. S. Chiu et al., Src and Pyk2 mediate G-proteincoupled receptor activation of epidermal growth factor receptor (EGFR) but are not required for coupling to the mitogen-activated protein (MAP) kinase signaling cascade, J. Biol. Chem, vol.276, pp.20130-20135, 2001.

H. Arai, K. Nakao, K. Takaya, K. Hosoda, Y. Ogawa et al., The human endothelin-B receptor gene. Structural organization and chromosomal assignment, J. Biol. Chem, vol.268, pp.3463-3470, 1993.

D. A. Arias, J. Navenot, W. Zhang, J. Broach, and S. C. Peiper, Constitutive Activation of CCR5 and CCR2 Induced by Conformational Changes in the Conserved TXP Motif in Transmembrane Helix 2, J. Biol. Chem, vol.278, pp.36513-36521, 2003.

Y. Artemenko, T. J. Lampert, and P. N. Devreotes, Moving towards a paradigm: common mechanisms of chemotactic signaling in Dictyostelium and mammalian leukocytes, Cell. Mol. Life Sci. CMLS, vol.71, pp.3711-3747, 2014.

T. K. Attwood and J. B. Findlay, Fingerprinting G-protein-coupled receptors, Protein Eng, vol.7, pp.195-203, 1994.

N. Audet, I. Charfi, O. Mnie-filali, M. Amraei, A. Chabot-doré et al., Differential association of receptor-G?? complexes with ?-arrestin2 determines recycling bias and potential for tolerance of ? opioid receptor agonists, J. Neurosci. Off. J. Soc. Neurosci, vol.32, pp.4827-4840, 2012.

M. Azzi, P. G. Charest, S. Angers, G. Rousseau, T. Kohout et al., ?-Arrestin-mediated activation of MAPK by inverse agonists reveals distinct active conformations for G protein-coupled receptors, Proc. Natl. Acad. Sci, vol.100, pp.11406-11411, 2003.

A. Balat, M. Karakok, K. Yilmaz, and Y. Kibar, Urotensin-II immunoreactivity in children with chronic glomerulonephritis, Renal failure, vol.29, pp.573-578, 2007.

F. Balkwill, Cancer and the chemokine network, Nat. Rev. Cancer, vol.4, pp.540-550, 2004.

J. A. Ballesteros and H. Weinstein, Integrated methods for the construction of threedimensional models and computational probing of structure-function relations in G protein-coupled receptors, pp.366-428, 1995.

J. Ballesteros, S. Kitanovic, F. Guarnieri, P. Davies, B. J. Fromme et al., Functional microdomains in G-protein-coupled receptors. The conserved arginine-cage motif in the gonadotropinreleasing hormone receptor, J. Biol. Chem, vol.273, pp.10445-10453, 1998.

J. A. Ballesteros, L. Shi, and J. A. Javitch, Structural Mimicry in G Protein-Coupled Receptors: Implications of the High-Resolution Structure of Rhodopsin for Structure-Function Analysis of Rhodopsin-Like Receptors, vol.60, pp.1-19, 2001.

Y. Ban, T. Watanabe, T. Suguro, T. A. Matsuyama, Y. Iso et al., Increased plasma urotensin-II and carotid atherosclerosis are associated with vascular dementia, J. Atheroscler. Thromb, vol.16, pp.179-187, 2009.

G. Bansal, K. M. Druey, and Z. Xie, R4 RGS proteins: regulation of G-protein signaling and beyond, Pharmacol. Ther, vol.116, pp.473-495, 2007.

O. K. Baryshnikova and B. D. Sykes, Backbone dynamics of SDF-1alpha determined by NMR: interpretation in the presence of monomer-dimer equilibrium, Protein Sci. Publ. Protein Soc, vol.15, pp.2568-2578, 2006.

M. Bastepe, Y. Gunes, B. Perez-villamil, J. Hunzelman, L. S. Weinstein et al., Receptor-Mediated Adenylyl Cyclase Activation Through XL?s, the Extra-Large Variant of the Stimulatory G Protein ?-Subunit, Mol. Endocrinol, vol.16, pp.1912-1919, 2002.

L. A. Begley, S. Kasina, R. B. Shah, and J. A. Macoska, Signaling mechanisms coupled to CXCL12/CXCR4-mediated cellular proliferation are PTEN-dependent, Am. J. Clin. Exp. Urol, vol.3, pp.91-99, 2015.

D. J. Behm, N. V. Aiyar, A. R. Olzinski, J. J. Mcatee, M. A. Hilfiker et al., ) GSK1562590, a slowly dissociating urotensin-II receptor antagonist, exhibits prolonged pharmacodynamic activity ex vivo, Br. J Pharmacol161, pp.207-228, 2010.

D. J. Behm, C. P. Doe, D. G. Johns, K. Maniscalco, G. P. Stankus et al., Urotensin-II: a novel systemic hypertensive factor in the cat, Naunyn Schmiedebergs Arch of Pharmacol, vol.369, pp.274-280, 2004.

D. J. Behm, C. L. Herold, V. Camarda, N. V. Aiyar, and S. A. Douglas, Differential agonistic and antagonistic effects of the urotensin-II ligand SB-710411 at rodent and primate UT receptors, Eur J Pharmacol, vol.492, pp.113-116, 2004.

D. J. Behm, C. L. Herold, E. H. Ohlstein, S. D. Knight, D. Dhanak et al., Pharmacological characterization of SB-710411 (Cpa-c[D-Cys-Pal-D-Trp-Lys-Val-Cys]-Cpa-amide), a novel peptidic urotensin-II receptor antagonist, Br. J Pharmacol, vol.137, pp.449-458, 2002.

D. J. Behm, G. Stankus, C. P. Doe, R. N. Willette, H. M. Sarau et al., The peptidic urotensin-II receptor ligand GSK248451 possesses less intrinsic activity than the low-efficacy partial agonists SB-710411 and urantide in native mammalian tissues and recombinant cell systems, Br. J Pharmacol, vol.148, pp.173-190, 2006.

I. Ben-ami, Z. Yao, Z. Naor, and R. Seger, Gq protein-induced apoptosis is mediated by AKT kinase inhibition that leads to protein kinase C-induced c-Jun Nterminal kinase activation, J. Biol. Chem, vol.286, pp.31022-31031, 2011.

J. L. Benovic, C. Staniszewski, F. Mayor, M. G. Caron, and R. J. Lefkowitz, beta-Adrenergic receptor kinase. Activity of partial agonists for stimulation of adenylate cyclase correlates with ability to promote receptor phosphorylation, J. Biol. Chem, vol.263, pp.3893-3897, 1988.

H. A. Bern and K. Lederis, A reference preparation for the study of active substances in the caudal neurosecretory system of teleosts, J. Endoc, vol.45, 1969.

D. Bian, C. Mahanivong, J. Yu, S. M. Frisch, Z. K. Pan et al., The G12/13-RhoA signaling pathway contributes to efficient lysophosphatidic acidstimulated cell migration, Oncogene, vol.25, pp.2234-2244, 2006.

S. D. Bianco, L. Vandepas, M. Correa-medina, B. Gereben, A. Mukherjee et al., KISS1R intracellular trafficking and degradation: effect of the Arg386Pro disease-associated mutation, Endocrinology, vol.152, pp.1616-1626, 2011.

J. Bockaert and J. P. Pin, Molecular tinkering of G protein-coupled receptors: an evolutionary success, EMBO J, vol.18, pp.1723-1729, 1999.

G. Boerrigter, D. G. Soergel, J. D. Violin, M. W. Lark, and J. C. Burnett, , 2012.

, Novel ?-Arrestin Biased Ligand at the Angiotensin II Type I Receptor, Unloads the Heart and Maintains Renal Function When Added to Furosemide in Experimental Heart Failure, TRV120027, a, vol.5, pp.627-634

L. M. Bohn, R. J. Lefkowitz, R. R. Gainetdinov, K. Peppel, M. G. Caron et al., Enhanced Morphine Analgesia in Mice Lacking ?-Arrestin 2, Science, vol.286, pp.2495-2498, 1999.

S. Boivin, L. Guilhaudis, I. Milazzo, H. Oulyadi, D. Davoust et al., Characterization of urotensin-II receptor structural domains involved in the recognition of U-II, URP, and urantide, Biochemistry (Mosc.), vol.45, pp.5993-6002, 2006.
URL : https://hal.archives-ouvertes.fr/pasteur-00820020

A. Boudot, G. Kerdivel, D. Habauzit, J. Eeckhoute, F. L. Dily et al., Differential Estrogen-Regulation of CXCL12 Chemokine Receptors, CXCR4 and CXCR7, Contributes to the Growth Effect of Estrogens in Breast Cancer Cells, PLoS ONE, vol.6, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00605459

N. Bousette, L. Patel, S. A. Douglas, E. H. Ohlstein, and A. Giaid, Increased expression of urotensin II and its cognate receptor GPR14 in atherosclerotic lesions of the human aorta, Atherosclerosis, vol.176, pp.117-123, 2004.

D. Brancaccio, F. Merlino, A. Limatola, A. M. Yousif, I. Gomez-monterrey et al., An investigation into the origin of the biased agonism associated with the urotensin II receptor activation, J. Pept. Sci, vol.21, pp.392-399, 2015.

M. R. Bratton, J. W. Antoon, B. N. Duong, D. E. Frigo, S. Tilghman et al., G?o potentiates estrogen receptor ? activity via the ERK signaling pathway, J. Endocrinol, vol.214, pp.45-54, 2012.

A. Brkovic, A. Hattenberger, E. Kostenis, T. Klabunde, S. Flohr et al., Functional and binding characterizations of urotensin II-related peptides in human and rat urotensin II-receptor assay, J Pharmacol Exp The, vol.306, pp.1200-1209, 2003.

C. Brulé, N. Perzo, J. Joubert, X. Sainsily, R. Leduc et al., Biased signaling regulates the pleiotropic effects of the urotensin II receptor to modulate its cellular behaviors, FASEB J, vol.28, pp.5148-5162, 2014.

A. M. Buhl, N. L. Johnson, N. Dhanasekaran, J. , and G. L. , G?12 and G?13 Stimulate Rho-dependent Stress Fiber Formation and Focal Adhesion Assembly, J. Biol. Chem, vol.270, pp.24631-24634, 1995.

J. R. Bunzow, H. H. Tol, D. K. Grandy, P. Albert, J. Salon et al., Cloning and expression of a rat D2 dopamine receptor cDNA, Nature, vol.336, pp.783-787, 1988.

J. M. Busillo and J. L. Benovic, Regulation of CXCR4 signaling, Biochim. Biophys. Acta, vol.1768, pp.952-963, 2007.

J. M. Busillo, S. Armando, R. Sengupta, O. Meucci, M. Bouvier et al., , 2010.

, Site-specific phosphorylation of CXCR4 is dynamically regulated by multiple kinases and results in differential modulation of CXCR4 signaling, J. Biol. Chem, vol.285, pp.7805-7817

J. Cabana, B. Holleran, R. Leduc, E. Escher, G. Guillemette et al., Identification of Distinct Conformations of the Angiotensin-II Type 1 Receptor Associated with the Gq/11 Protein Pathway and the ?-Arrestin Pathway Using Molecular Dynamics Simulations, J. Biol. Chem, vol.290, pp.15835-15854, 2015.

D. Calebiro, V. O. Nikolaev, M. C. Gagliani, T. De-filippis, C. Dees et al., Persistent cAMP-signals triggered by internalized Gprotein-coupled receptors, PLoS Biol, vol.7, p.1000172, 2009.

V. Camarda, R. Guerrini, E. Kostenis, A. Rizzi, G. Calò et al., A new ligand for the urotensin II receptor, Br. J. Pharmacol, vol.137, pp.311-314, 2002.

V. Camarda, W. Song, E. Marzola, M. Spagnol, R. Guerrini et al., Urantide mimics urotensin-II induced calcium release in cells expressing recombinant UT receptors, Eur J Pharmacol, vol.498, pp.83-86, 2004.

V. Camarda, M. Spagnol, W. Song, R. Vergura, A. L. Roth et al., In vitro and in vivo pharmacological characterization of the novel UT receptor ligand, Br. J. Pharmacol, vol.147, pp.92-100, 2006.

A. Carotenuto, P. Grieco, P. Campiglia, E. Novellino, and P. Rovero, Unraveling the active conformation of urotensin II, J Med Chem, vol.47, pp.1652-1661, 2004.

J. Cartailler and H. Luecke, Structural and Functional Characterization of ? Bulges and Other Short Intrahelical Deformations, Structure, vol.12, pp.133-144, 2004.

L. B. Case, M. A. Baird, G. Shtengel, S. L. Campbell, H. F. Hess et al., Molecular mechanism of vinculin activation and nanoscale spatial organization in focal adhesions, Nat. Cell Biol, vol.17, pp.880-892, 2015.

H. Castel, M. Diallo, D. Chatenet, J. Leprince, L. Desrues et al., Biochemical and functional characterization of high-affinity urotensin II receptors in rat cortical astrocytes, J. Neurochem, vol.99, pp.582-595, 2006.
URL : https://hal.archives-ouvertes.fr/hal-01960748

M. J. Caterina, P. N. Devreotes, J. Borleis, and D. Hereld, Agonist-induced loss of ligand binding is correlated with phosphorylation of cAR1, a G protein-coupled chemoattractant receptor from Dictyostelium, J. Biol. Chem, vol.270, pp.8667-8672, 1995.

P. Chakrabarti and S. Chakrabarti, C-H?O hydrogen bond involving proline residues in ?-helices1, J. Mol. Biol, vol.284, pp.867-873, 1998.

J. Changeux and S. Edelstein, Conformational selection or induced fit? 50 years of debate resolved, Biol. Rep, vol.3, 1000.

N. Chartrel, J. M. Conlon, F. Collin, B. Braun, D. Waugh et al., Urotensin II in the central nervous system of the frog Rana ridibunda: immunohistochemical localization and biochemical characterization, J Comp Neurol, vol.364, pp.324-339, 1996.

D. Chatenet, B. Folch, D. Feytens, M. Letourneau, D. Tourwe et al., Development and pharmacological characterization of conformationally constrained urotensin II-related peptide agonists, J Med Chem, vol.56, pp.9612-9622, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-01130814

D. Chatenet, C. Dubessy, J. Leprince, C. Boularan, L. Carlier et al., Structure-activity relationships and structural conformation of a novel urotensin II-related peptide, Peptides, vol.25, pp.1819-1830, 2004.
URL : https://hal.archives-ouvertes.fr/hal-01960815

D. Chatenet, T. M. Nguyen, M. Létourneau, and A. Fournier, Update on the urotensinergic system: new trends in receptor localization, activation, and drug design, Front. Endocrinol, vol.3, p.174, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-01001636

J. Chen, M. Devivo, J. Dingus, A. Harry, J. Li et al., A region of adenylyl cyclase 2 critical for regulation by G protein beta gamma subunits, Science, vol.268, pp.1166-1169, 1995.

Y. Chen, M. Zhao, X. Liu, W. Yao, J. Yang et al., Urotensin II receptor in the rat airway smooth muscle and its effect on the rat airway smooth muscle cells proliferation, Chin Med Sci J, vol.16, pp.231-235, 2001.

L. Chen, J. C. Liu, S. H. Loh, C. H. Chen, C. Y. Hong et al., Involvement of reactive oxygen species in urotensin II-induced proliferation of cardiac fibroblasts, Eur J Pharmacol, vol.593, pp.24-29, 2008.

X. Chen, J. Fang, S. Wang, H. Liu, X. Du et al., A new mosaic pattern in glioma vascularization: exogenous endothelial progenitor cells integrating into the vessels containing tumor-derived endothelial cells, Oncotarget, vol.5, pp.1955-1968, 2014.

Z. Chen, J. Xu, Y. Ye, Y. Li, H. Gong et al., Urotensin II inhibited the proliferation of cardiac side population cells in mice during pressure overload by JNK-LRP6 signalling, J Cel Mol Med, vol.18, pp.852-862, 2014.

V. Cherezov, D. M. Rosenbaum, M. A. Hanson, S. G. Rasmussen, F. S. Thian et al., High-resolution crystal structure of an engineered human beta2-adrenergic G protein-coupled receptor, Science, vol.318, pp.1258-1265, 2007.

H. Chikumi, J. Vázquez-prado, J. Servitja, H. Miyazaki, and J. S. Gutkind, Potent Activation of RhoA by G?q and Gq-coupled Receptors, J. Biol. Chem, vol.277, pp.27130-27134, 2002.

H. Choe, Y. J. Kim, J. H. Park, T. Morizumi, E. F. Pai et al., Crystal structure of metarhodopsin II, Nature, vol.471, pp.651-655, 2011.

O. Civelli, Y. Saito, Z. Wang, H. Nothacker, and R. K. Reinscheid, Orphan GPCRs and their ligands, Pharmacol. Ther, vol.110, pp.525-532, 2006.

M. J. Clark, C. Harrison, H. Zhong, R. R. Neubig, and J. R. Traynor, Endogenous RGS protein action modulates mu-opioid signaling through Galphao. Effects on adenylyl cyclase, extracellular signal-regulated kinases, and intracellular calcium pathways, J. Biol. Chem, vol.278, pp.9418-9425, 2003.

S. D. Clark, H. P. Nothacker, Z. Wang, Y. Saito, F. M. Leslie et al., The urotensin II receptor is expressed in the cholinergic mesopontine tegmentum of the rat, Brain Res, vol.923, pp.120-127, 2001.

M. K. Clements, T. P. Mcdonald, R. Wang, G. Xie, B. F. O'dowd et al., FMRFamide-Related Neuropeptides Are Agonists of the Orphan G-Protein-Coupled Receptor GPR54, Biochem. Biophys. Res. Commun, vol.284, pp.1189-1193, 2001.

M. Clozel, C. Binkert, M. Birker-robaczewska, C. Boukhadra, S. S. Ding et al., -methyl-quinolin-4-yl)-urea sulfate salt): first demonstration of a pathophysiological role of the urotensin System, Pharmacology of the urotensin, vol.311, issue.2, pp.204-212, 2004.

M. Cojoc, C. Peitzsch, F. Trautmann, L. Polishchuk, G. D. Telegeev et al., Emerging targets in cancer management: role of the CXCL12/CXCR4 axis, OncoTargets Ther, vol.6, p.1347, 2013.

P. Coly, N. Perzo, V. Le-joncour, C. Lecointre, M. Schouft et al., Chemotactic G protein-coupled receptors control cell migration by repressing autophagosome biogenesis, Autophagy, vol.12, pp.1-19, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02345740

J. M. Conlon, F. O'harte, D. D. Smith, M. C. Tonon, and H. Vaudry, Isolation and primary structure of urotensin II from the brain of a tetrapod, the frog Rana ridibunda, Biochem. Biophys. Res. Commun, vol.188, pp.578-583, 1992.

M. Conner, S. R. Hawtin, J. Simms, D. Wootten, Z. Lawson et al., Systematic analysis of the entire second extracellular loop of the V(1a) vasopressin receptor: key residues, conserved throughout a G-proteincoupled receptor family, identified, J. Biol. Chem, vol.282, pp.17405-17412, 2007.

R. L. Contento, B. Molon, C. Boularan, T. Pozzan, S. Manes et al., CXCR4-CCR5: a couple modulating T cell functions, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.10101-10106, 2008.

F. S. Cordes, J. N. Bright, and M. S. Sansom, Proline-induced Distortions of Transmembrane Helices, J. Mol. Biol, vol.323, pp.951-960, 2002.

O. A. Coso, H. Teramoto, W. F. Simonds, and J. S. Gutkind, Signaling from G protein-coupled receptors to c-Jun kinase involves beta gamma subunits of heterotrimeric G proteins acting on a Ras and Rac1-dependent pathway, J. Biol. Chem, vol.271, pp.3963-3966, 1996.

C. M. Costa-neto, -. Parreiras, L. T. Silva, and M. Bouvier, A Pluridimensional View of Biased Agonism, Mol. Pharmacol, vol.90, pp.587-595, 2016.

Y. Coulouarn, I. Lihrmann, S. Jegou, Y. Anouar, H. Tostivint et al., Cloning of the cDNA encoding the urotensin II precursor in frog and human reveals intense expression of the urotensin II gene in motoneurons of the spinal cord, PNAS, vol.95, pp.15803-15808, 1998.

E. Cowley, J. P. Thompson, P. Sharpe, J. Waugh, N. Ali et al., Effects of preeclampsia on maternal plasma, cerebrospinal fluid, and umbilical cord urotensin II concentrations: a pilot study, Br J Anaesth, vol.95, pp.495-499, 2005.

C. J. Crasto, Hydrophobicity profiles in G protein-coupled receptor transmembrane helical domains, J. Recept. Ligand Channel Res, pp.123-133, 2010.

M. P. Crump, J. Gong, P. Loetscher, K. Rajarathnam, A. Amara et al., Solution structure and basis for functional activity of stromal cell-derived factor-1; dissociation of CXCR4 activation from binding and inhibition of HIV-1, EMBO J, vol.16, pp.6996-7007, 1997.

K. P. Currie, G protein modulation of CaV2 voltage-gated calcium channels, Channels Austin Tex, vol.4, pp.497-509, 2010.

D. Cvetkovi?, M. Dragan, S. J. Leith, Z. M. Mir, H. S. Leong et al., KISS1R Induces Invasiveness of Estrogen Receptor-Negative Human Mammary Epithelial and Breast Cancer Cells, Endocrinology, vol.154, pp.1999-2014, 2013.

V. Cvicek, W. A. Iii, A. , and R. , Structure-Based Sequence Alignment of the Transmembrane Domains of All Human GPCRs: Phylogenetic, Structural and Functional Implications, PLOS Comput Biol, vol.12, 2016.

Y. Daaka, L. M. Luttrell, and R. J. Lefkowitz, Switching of the coupling of the beta2-adrenergic receptor to different G proteins by protein kinase A, Nature, vol.390, pp.88-91, 1997.

H. Y. Dai, X. G. Guo, Z. M. Ge, Z. H. Li, X. J. Yu et al., Elevated expression of urotensin II and its receptor in diabetic cardiomyopathy, J. Diabetes Complicat, vol.22, pp.137-143, 2008.

J. A. Dalton, I. Lans, G. , and J. , Quantifying conformational changes in GPCRs: glimpse of a common functional mechanism, BMC Bioinformatics, vol.16, 2015.

H. Daub, F. U. Weiss, C. Wallasch, and A. Ullrich, Role of transactivation of the EGF receptor in signalling by G-protein-coupled receptors, Nature, vol.379, pp.557-560, 1996.

F. M. Décaillot, K. Befort, D. Filliol, S. Yue, P. Walker et al., Opioid receptor random mutagenesis reveals a mechanism for G protein-coupled receptor activation, Nat. Struct. Mol. Biol, vol.10, pp.629-636, 2003.

K. A. Defea, Stop that cell! Beta-arrestin-dependent chemotaxis: a tale of localized actin assembly and receptor desensitization, Annu. Rev. Physiol, vol.69, pp.535-560, 2007.

M. Y. Degtyarev, A. M. Spiegel, and T. L. Jones, Palmitoylation of a G protein alpha i subunit requires membrane localization not myristoylation, J. Biol. Chem, vol.269, pp.30898-30903, 1994.

L. Desrues, T. Lefebvre, C. Lecointre, M. Schouft, J. Leprince et al., Down-Regulation of GABA(A) Receptor via Promiscuity with the Vasoactive Peptide Urotensin II Receptor. Potential Involvement in Astrocyte Plasticity, PloS One, vol.7, p.36319, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02294501

X. Deupi and B. K. Kobilka, Energy Landscapes as a Tool to Integrate GPCR Structure, Dynamics, and Function, Physiology, vol.25, pp.293-303, 2010.

X. Deupi, M. Olivella, C. Govaerts, J. A. Ballesteros, M. Campillo et al., Ser and Thr residues modulate the conformation of pro-kinked transmembrane alphahelices, Biophys. J, vol.86, pp.105-115, 2004.

J. Devillé, J. Rey, and M. Chabbert, An indel in transmembrane helix 2 helps to trace the molecular evolution of class A G-protein-coupled receptors, J. Mol. Evol, vol.68, pp.475-489, 2009.

M. Diallo, . M. Jarry, L. Desrues, H. Castel, D. Chatenet et al., Orn5]URP acts as a pure antagonist of urotensinergic receptors in rat cortical astrocytes, Peptides, vol.29, pp.813-819, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01962707

I. Diebold, A. Petry, M. Burger, J. Hess, and A. Gorlach, NOX4 mediates activation of FoxO3a and matrix metalloproteinase-2 expression by urotensin-II, Mol. Biol. Cell, vol.22, pp.4424-4434, 2011.

S. Diel, K. Klass, B. Wittig, and C. Kleuss, Gbetagamma activation site in adenylyl cyclase type II. Adenylyl cyclase type III is inhibited by Gbetagamma, J. Biol. Chem, vol.281, pp.288-294, 2006.

G. Dietrich, G. Gaibelet, R. Capeyrou, J. L. Butour, F. Pontet et al., Implication of the first and third extracellular loops of the mu-opioid receptor in the formation of the ligand binding site: a study using chimeric mu-opioid/angiotensin receptors, J. Neurochem, vol.70, pp.2106-2111, 1998.

R. A. Dixon, B. K. Kobilka, D. J. Strader, J. L. Benovic, H. G. Dohlman et al., Cloning of the gene and cDNA for mammalian ?-adrenergic receptor and homology with rhodopsin, Nature, vol.321, pp.75-79, 1986.

T. Djordjevic, R. S. Belaiba, S. Bonello, J. Pfeilschifter, J. Hess et al., Human urotensin II is a novel activator of NADPH oxidase in human pulmonary artery smooth muscle cells, Arterios. Scler. Thromb. Vasc. Biol, vol.25, pp.519-525, 2005.

N. D. Doan, T. T. Nguyen, M. Letourneau, K. Turcotte, A. Fournier et al., Biochemical and pharmacological characterization of nuclear urotensin-II binding sites in rat heart, Br. J. Pharmacol, vol.166, pp.243-257, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-00818552

S. A. Douglas, L. Tayara, E. H. Ohlstein, N. Halawa, and A. Giaid, Congestive heart failure and expression of myocardial urotensin II, Lancet, vol.359, pp.1990-1997, 2002.

S. A. Douglas, D. Naselsky, Z. Ao, J. Disa, C. L. Herold et al., Identification and pharmacological characterization of native, functional human urotensin-II receptors in rhabdomyosarcoma cell lines, Br. J. Pharmacol, vol.142, pp.921-932, 2004.

S. A. Douglas, D. J. Behm, N. V. Aiyar, D. Naselsky, J. Disa et al., Nonpeptidic urotensin-II receptor antagonists I: in vitro pharmacological characterization of SB-Jane-Eileen Joubert 706375, Br. J. Pharmacol, vol.145, pp.620-635, 2005.

S. A. Douglas, D. Dhanak, and D. G. Johns, From 'gills to pills': urotensin-II as a regulator of mammalian cardiorenal function, Trends Pharmacol. Sci, vol.25, pp.76-85, 2004.

G. W. Dorn, GRK mythology: G-protein receptor kinases in cardiovascular disease, J. Mol. Med, vol.87, pp.455-463, 2009.

L. J. Drury, J. J. Ziarek, S. Gravel, C. T. Veldkamp, T. Takekoshi et al., Monomeric and dimeric CXCL12 inhibit metastasis through distinct CXCR4 interactions and signaling pathways, Proc. Natl. Acad. Sci. U. S. A, vol.108, pp.17655-17660, 2011.

T. Dschietzig, C. Bartsch, R. Pregla, H. R. Zurbrugg, F. P. Armbruster et al., Plasma levels and cardiovascular gene expression of urotensin-II in human heart failure, Regul. Pept, vol.110, pp.33-38, 2002.

C. Dubessy, D. Cartier, B. Lectez, C. Bucharles, N. Chartrel et al., Characterization of urotensin II, distribution of urotensin II, urotensin II-related peptide and UT receptor mRNAs in mouse: evidence of urotensin II at the neuromuscular junction, J. Neurochem, vol.107, pp.361-374, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01962726

S. L. Dun, G. C. Brailoiu, J. Yang, J. K. Chang, and N. J. Dun, Urotensin IIimmunoreactivity in the brainstem and spinal cord of the rat, Neurosci. Lett, vol.305, pp.9-12, 2001.

J. H. Dunham, R. C. Meyer, E. L. Garcia, and R. A. Hall, GPR37 surface expression enhancement via N-terminal truncation or protein-protein interactions, Biochemistry (Mosc.), vol.48, pp.10286-10297, 2009.

J. G. Egginger, C. Parmentier, G. Garrel, J. Cohen-tannoudji, A. Camus et al., Direct evidence for the co-expression of URP and GnRH in a sub-population of rat hypothalamic neurones: anatomical and functional correlation, PloS one, vol.6, 2011.

H. M. El-shewy, S. A. Abdel-samie, A. M. Qalam, M. Lee, K. Kitatani et al., Phospholipase C and protein kinase C-? 2 mediate insulin-like growth factor II-dependent sphingosine kinase 1 activation, Mol. Endocrinol. Baltim. Md, vol.25, pp.2144-2156, 2011.

N. A. Elshourbagy, S. A. Douglas, U. Shabon, S. Harrison, G. Duddy et al., Molecular and pharmacological characterization of genes encoding urotensin-II peptides and their cognate G-protein-coupled receptors from the mouse and monkey, Br J Pharmacol, vol.136, pp.9-22, 2002.

. Di-villa, R. Bianca, G. Cirino, E. Mitidieri, C. Coletta et al., Urotensin II: a novel target in human corpus cavernosum, J. Sex. Med, vol.7, pp.1778-1786, 2010.

O. P. Ernst, C. Bieri, H. Vogel, and K. P. Hofmann, Intrinsic biophysical monitors of transducin activation: Fluorescence, UV-visible spectroscopy, light scattering, and evanescent field techniques, pp.471-489, 2000.

G. Esposito, C. Perrino, A. Cannavo, G. G. Schiattarella, F. Borgia et al., EGFR trans-activation by urotensin II receptor is mediated by ?-arrestin recruitment and confers cardioprotection in pressure overload-induced cardiac hypertrophy, Basic Res. Cardiol, vol.106, pp.577-589, 2011.

A. Federico, S. Zappavigna, M. Romano, P. Grieco, A. Luce et al., Urotensin-II receptor is over-expressed in colon cancer cell lines and in colon carcinoma in humans, European journal of clinical investigation, vol.44, pp.285-294, 2014.

G. Fenalti, P. M. Giguere, V. Katritch, X. Huang, A. A. Thompson et al., Molecular control of ?-opioid receptor signalling, Nature, vol.506, pp.191-196, 2014.

Y. Feng, C. C. Broder, P. E. Kennedy, and E. A. Berger, HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor, Science, vol.272, pp.872-877, 1996.

A. Fisher, E. Heldman, D. Gurwitz, R. Haring, D. Barak et al., Selective Signaling via Unique Ml Muscarinic Agonistsa, Ann. N. Y. Acad. Sci, vol.695, pp.300-303, 1993.

S. Flohr, M. Kurz, E. Kostenis, A. Brkovich, A. Fournier et al., Identification of nonpeptidic urotensin II receptor antagonists by virtual screening based on a pharmacophore model derived from structure-activity relationships and nuclear magnetic resonance studies on urotensin II, Journal of medicinal chemistry, vol.45, pp.1799-1805, 2002.

S. Foister, L. L. Taylor, J. J. Feng, W. L. Chen, A. Lin et al., Design and synthesis of potent cystine-free cyclic hexapeptide agonists at the human urotensin receptor, Organic letters, vol.8, pp.1799-1802, 2006.

M. Fokkelman, H. E. Balc?o?lu, J. E. Klip, K. Yan, F. J. Verbeek et al., Cellular adhesome screen identifies critical modulators of focal adhesion dynamics, cellular traction forces and cell migration behaviour, Sci. Rep, vol.6, 2016.

A. P. Fontes-sousa, C. Bras-silva, A. L. Pires, D. Monteiro-sousa, and A. F. Leite-moreira, Urotensin II acutely increases myocardial length and distensibility: potential implications for diastolic function and ventricular remodeling, Naunyn-Schmiedeberg's archives of pharmacology, vol.376, pp.107-115, 2007.

M. Franco, P. Chardin, M. Chabre, P. , and S. , Myristoylation-facilitated binding of the G protein ARF1GDP to membrane phospholipids is required for its activation by a soluble nucleotide exchange factor, J. Biol. Chem, vol.271, pp.1573-1578, 1996.

R. Franco, E. Martínez-pinilla, J. L. Lanciego, and G. Navarro, Basic Pharmacological and Structural Evidence for Class A G-Protein-Coupled Receptor Heteromerization, Front. Pharmacol, vol.7, p.76, 2016.

R. Franco, S. Zappavigna, V. Gigantino, A. Luce, M. Cantile et al., Urotensin II receptor determines prognosis of bladder cancer regulating cell motility/invasion, Journal of experimental & clinical cancer research, vol.33, p.48, 2014.

R. Fredriksson and H. B. Schiöth, The repertoire of G-protein-coupled receptors in fully sequenced genomes, Mol. Pharmacol, vol.67, pp.1414-1425, 2005.

R. Fredriksson, M. C. Lagerström, L. Lundin, and H. B. Schiöth, The G-proteincoupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints, Mol. Pharmacol, vol.63, pp.1256-1272, 2003.

C. Galés, R. V. Rebois, M. Hogue, P. Trieu, A. Breit et al., Real-time monitoring of receptor and G-protein interactions in living cells, Nat. Methods, vol.2, pp.177-184, 2005.

S. Gao, Y. B. Oh, B. M. Park, W. H. Park, and S. H. Kim, Urotensin II protects ischemic reperfusion injury of hearts through ROS and antioxidant pathway, Peptides, vol.36, pp.199-205, 2012.

S. Gao, Y. B. Oh, A. Shah, W. H. Park, M. J. Chung et al., Urotensin II receptor antagonist attenuates monocrotaline-induced cardiac hypertrophy in rats, Am J Physiol Heart Circ Physiol, vol.299, pp.1782-1789, 2010.

J. Gartlon, F. Parker, D. C. Harrison, S. A. Douglas, T. E. Ashmeade et al., Central effects of urotensin-II following ICV administration in rats, Psychopharmacology, vol.155, pp.426-433, 2001.

L. Ge, S. K. Shenoy, R. J. Lefkowitz, and K. Defea, Constitutive protease-activated receptor-2-mediated migration of MDA MB-231 breast cancer cells requires both beta-arrestin-1 and -2, J. Biol. Chem, vol.279, pp.55419-55424, 2004.

D. Gesty-palmer, L. Yuan, B. Martin, W. H. Wood, M. Lee et al., ?-arrestin-selective G proteincoupled receptor agonists engender unique biological efficacy in vivo, Mol. Endocrinol. Baltim. Md, vol.27, pp.296-314, 2013.

A. G. Gilman, G Proteins: Transducers of Receptor-Generated Signals, Annu. Rev. Biochem, vol.56, pp.615-649, 1987.

L. Giuliani, L. Lenzini, M. Antonello, E. Aldighieri, A. S. Belloni et al., Expression and functional role of urotensin-II and its receptor in the adrenal cortex and medulla: novel insights for the pathophysiology of primary aldosteronism, J Clin Endocrinol Metab, vol.94, pp.684-690, 2009.

A. Göblyös and A. P. Ijzerman, Allosteric modulation of adenosine receptors, Biochim. Biophys. Acta, vol.1808, pp.1309-1318, 2011.

A. Gohla, R. Harhammer, and G. Schultz, The G-protein G13 but Not G12 Mediates Signaling from Lysophosphatidic Acid Receptor via Epidermal Growth Factor Receptor to Rho, J. Biol. Chem, vol.273, pp.4653-4659, 1998.

F. Golzar, S. H. Javanmard, V. Bahrambeigi, R. , and L. , The effect of Kisspeptin-10 on mesenchymal stem cells migration in vitro and in vivo, Adv. Biomed. Res, vol.4, p.20, 2015.

H. Gong, Y. X. Wang, Y. Z. Zhu, W. W. Wang, M. J. Wang et al., Cellular distribution of GPR14 and the positive inotropic role of urotensin II in the myocardium in adult rat, J Applied Physiology, vol.97, pp.2228-2235, 2004.

A. González, T. Perez-acle, L. Pardo, and X. Deupi, Molecular basis of ligand dissociation in ?-adrenergic receptors, PloS One, vol.6, 2011.

C. Govaerts, C. Blanpain, X. Deupi, S. Ballet, J. A. Ballesteros et al., The TXP motif in the second transmembrane helix of CCR5. A structural determinant of chemokine-induced activation, J. Biol. Chem, vol.276, pp.13217-13225, 2001.

G. C. Gonzalez, M. Martinez-padron, K. Lederis, and K. Lukowiak, Distribution and coexistence of urotensin I and urotensin II peptides in the cerebral ganglia of Aplysia Jane-Eileen Joubert californica, Peptides, vol.13, pp.695-703, 1992.

C. Govaerts, A. Lefort, S. Costagliola, S. J. Wodak, J. A. Ballesteros et al., A Conserved Asn in Transmembrane Helix 7 Is an On/Off Switch in the Activation of the Thyrotropin Receptor, J. Biol. Chem, vol.276, pp.22991-22999, 2001.

T. M. Gray and B. W. Matthews, Intrahelical hydrogen bonding of serine, threonine and cysteine residues within alpha-helices and its relevance to membrane-bound proteins, J. Mol. Biol, vol.175, pp.75-81, 1984.

P. Grieco, A. Carotenuto, P. Campiglia, E. Zampelli, R. Patacchini et al., A new, potent urotensin II receptor peptide agonist containing a Pen residue at the disulfide bridge, J Med. Chem, vol.45, pp.4391-4394, 2002.

P. Grieco, R. Franco, G. Bozzuto, L. Toccacieli, A. Sgambato et al., Urotensin II receptor predicts the clinical outcome of prostate cancer patients and is involved in the regulation of motility of prostate adenocarcinoma cells, J. Cell. Biochem, vol.112, pp.341-353, 2011.

C. E. Groer, K. Tidgewell, R. A. Moyer, W. W. Harding, R. B. Rothman et al., An Opioid Agonist that Does Not Induce µ-Opioid Receptor-Arrestin Interactions or Receptor Internalization, Mol. Pharmacol, vol.71, pp.549-557, 2007.

D. Gruson, A. Ginion, N. Decroly, P. Lause, J. L. Vanoverschelde et al., Urotensin II induction of adult cardiomyocytes hypertrophy involves the Akt/GSK-3beta signaling pathway, Peptides, vol.31, pp.1326-1333, 2010.

J. Gsponer and M. M. Babu, The rules of disorder or why disorder rules, Prog. Biophys. Mol. Biol, vol.99, pp.94-103, 2009.

R. Guerrini, V. Camarda, E. Marzola, M. Arduin, G. Calo et al., Structure-activity relationship study on human urotensin II, J. Pept. Sci. Off. Publ. Eur. Pept. Soc, vol.11, pp.85-90, 2005.

D. Guo, T. Mulder-krieger, A. P. Ijzerman, and L. H. Heitman, Functional efficacy of adenosine A?A receptor agonists is positively correlated to their receptor residence time, Br. J. Pharmacol, vol.166, pp.1846-1859, 2012.

J. Guo, X. Yu, J. Gu, Z. Lin, G. Zhao et al., Regulation of CXCR4/AKT-signaling-induced cell invasion and tumor metastasis by RhoA, Rac-1, and Cdc42 in human esophageal cancer, Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med, vol.37, pp.6371-6378, 2016.

R. Guo, L. Chai, L. Chen, W. Chen, L. Ge et al., Stromal cell-derived factor 1 (SDF-1) accelerated skin wound healing by promoting the migration and proliferation of epidermal stem cells, In Vitro Cell. Dev. Biol. Anim, vol.51, pp.578-585, 2015.

J. Joubert,

D. Gurwitz, R. Haring, E. Heldman, C. M. Fraser, D. Manor et al., Discrete activation of transduction pathways associated with acetylcholine m1 receptor by several muscarinic ligands, Eur. J. Pharmacol. Mol. Pharmacol, vol.267, pp.21-31, 1994.

H. Gutiérrez-de-terán, A. Massink, D. Rodríguez, W. Liu, G. W. Han et al., The role of a sodium ion binding site in the allosteric modulation of the A(2A) adenosine G protein-coupled receptor, Struct. Lond. Engl, vol.21, pp.2175-2185, 1993.

E. Gutiérrez-pascual, J. Leprince, A. J. Martínez-fuentes, I. Ségalas-milazzo, R. Pineda et al., Vivo and in Vitro Structure-Activity Relationships and Structural Conformation of Kisspeptin-10-Related Peptides, vol.76, pp.58-67, 2009.

C. Hague, Z. Chen, A. S. Pupo, N. A. Schulte, M. L. Toews et al., , 2004.

, The N Terminus of the Human ?1D-Adrenergic Receptor Prevents Cell Surface Expression, J. Pharmacol. Exp. Ther, vol.309, pp.388-397

S. Hautaniemi, S. Kharait, A. Iwabu, A. Wells, and D. A. Lauffenburger, Modeling of signal-response cascades using decision tree analysis, Bioinforma. Oxf. Engl, vol.21, pp.2027-2035, 2005.

M. Heringlake, T. Kox, O. Uzun, B. Will, L. Bahlmann et al., The relationship between urotensin II plasma immunoreactivity and left ventricular filling pressures in coronary artery disease, Reg. Pep, vol.121, pp.129-136, 2004.

N. Heveker, M. Montes, L. Germeroth, A. Amara, A. Trautmann et al., Dissociation of the signalling and antiviral properties of SDF-1-derived small peptides, Curr. Biol. CB, vol.8, pp.369-376, 1998.

B. Hille, G protein-coupled mechanisms and nervous signaling, Neuron, vol.9, pp.187-195, 1992.

T. Hirose, K. Takahashi, N. Mori, T. Nakayama, M. Kikuya et al., Increased expression of urotensin II, urotensin II-related peptide and urotensin II receptor mRNAs in the cardiovascular organs of hypertensive rats: comparison with endothelin-1, Peptides, vol.30, pp.1124-1129, 2009.

B. J. Holleran, I. Domazet, M. Beaulieu, L. P. Yan, G. Guillemette et al., Identification of transmembrane domain 6 & 7 residues that contribute to the binding pocket of the urotensin II receptor, Biochem. Pharmacol, vol.77, pp.1374-1382, 2009.

S. Hollinger and J. R. Hepler, Cellular regulation of RGS proteins: modulators and integrators of G protein signaling, Pharmacol. Rev, vol.54, pp.527-559, 2002.

A. C. Holloway, H. Qian, L. Pipolo, J. Ziogas, S. Miura et al., Side-Chain Substitutions within Angiotensin II Reveal Different Requirements for Signaling, Internalization, and Phosphorylation of Type 1A Angiotensin Receptors, Mol. Pharmacol, vol.61, pp.768-777, 2002.

J. Hu, Y. Wang, X. Zhang, J. R. Lloyd, J. H. Li et al., Structural basis of G protein-coupled receptor-G protein interactions, Nat. Chem. Biol, vol.6, pp.541-548, 2010.

X. Huang, J. Shen, M. Cui, L. Shen, X. Luo et al., Molecular Dynamics Simulations on SDF-1?: Binding with CXCR4 Receptor, Biophys. J, vol.84, pp.171-184, 2003.

T. Huma, Z. Wang, J. Rizak, F. Ahmad, M. Shahab et al., Kisspeptin-10 modulates the proliferation and differentiation of the rhesus monkey derived stem cell line, p.135470, 2013.

D. L. Hunton, W. G. Barnes, J. Kim, X. Ren, J. D. Violin et al., Beta-arrestin 2-dependent angiotensin II type 1A receptor-mediated pathway of chemotaxis, Mol. Pharmacol, vol.67, pp.1229-1236, 2005.

E. H. Hurowitz, J. M. Melnyk, Y. J. Chen, H. Kouros-mehr, M. I. Simon et al., Genomic characterization of the human heterotrimeric G protein alpha, beta, and gamma subunit genes, DNA Res. Int. J. Rapid Publ. Rep. Genes Genomes, vol.7, pp.111-120, 2000.

M. Iglewski and S. R. Grant, Urotensin II-induced signaling involved in proliferation of vascular smooth muscle cells, Vascular health and risk management, vol.6, pp.723-734, 2010.

N. C. Information, B. Pike, U. S. , R. Md, B. et al., A highly efficacious lymphocyte chemoattractant, p.20894, 1996.

, J. Exp. Med, vol.184, p.1101

Y. Itoh, K. Cai, and H. G. Khorana, Mapping of contact sites in complex formation between light-activated rhodopsin and transducin by covalent crosslinking: Use of a chemically preactivated reagent, Proc. Natl. Acad. Sci, vol.98, pp.4883-4887, 2001.

F. Jaffré, A. E. Friedman, Z. Hu, N. Mackman, and B. C. Blaxall, ?-Adrenergic Receptor Stimulation Transactivates Protease-Activated Receptor 1 via Matrix Metalloproteinase 13 in Cardiac CellsClinical Perspective, Circulation, vol.125, pp.2993-3003, 2012.

J. M. Janz and D. L. Farrens, Rhodopsin Activation Exposes a Key Hydrophobic Binding Site for the Transducin ?-Subunit C Terminus, J. Biol. Chem, vol.279, pp.29767-29773, 2004.

M. B. Jarpe, C. Knall, F. M. Mitchell, A. M. Buhl, E. Duzic et al., Substance P Acts as a Biased Agonist toward Neuropeptide and Chemokine Receptors, J. Biol. Chem, vol.273, pp.3097-3104, 1998.

M. Jarry, M. Diallo, C. Lecointre, L. Desrues, T. Tokay et al., The vasoactive peptides urotensin II and urotensin II-related peptide regulate astrocyte activity through common and distinct mechanisms: involvement in cell proliferation, Biochem. J, vol.428, pp.113-124, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00479207

B. Jastrzebska, Y. Tsybovsky, and K. Palczewski, Complexes between photoactivated rhodopsin and transducin: progress and questions, Biochem. J, vol.428, pp.1-10, 2010.

D. G. Johns, Z. Ao, D. Naselsky, C. L. Herold, K. Maniscalco et al., Urotensin-II-mediated cardiomyocyte hypertrophy: effect of receptor antagonism and role of inflammatory mediators, Naunyn-Schmiedeberg's archives of pharmacology, vol.370, pp.238-250, 2004.

S. Jegou, D. Cartier, C. Dubessy, B. J. Gonzalez, D. Chatenet et al., Localization of the urotensin II receptor in the rat central nervous system, J. Comp. Neurol, vol.495, pp.21-36, 2006.
URL : https://hal.archives-ouvertes.fr/hal-01962761

D. Joyal, T. Huynh, N. Aiyar, B. Guida, S. Douglas et al., Urotensin-II levels in acute coronary syndromes, Int. J. Cardiol, vol.108, pp.31-35, 2006.

T. Jiang, S. Zhang, B. Lin, L. Meng, and H. Gao, , 2005.

, Zhonghua Zhong Liu Za Zhi, vol.27, pp.229-231

A. W. Kahsai, K. Xiao, S. Rajagopal, S. Ahn, A. K. Shukla et al., Multiple ligand-specific conformations of the ?2-adrenergic receptor, Nat. Chem. Biol, vol.7, pp.692-700, 2011.

H. Kalamarz-kubiak, I. M. Ashkenazi, A. Kleszczynska, and H. Rosenfeld, , 2014.

, Urotensin II inhibits arginine vasotocin and stimulates isotocin release from nerve endings in the pituitary of gilthead sea bream (Sparus aurata), J. Exp. Zool. A Ecol. Genet. Physiol, vol.321, pp.467-471

I. Kalatskaya, S. Schüssler, A. Blaukat, W. Müller-esterl, M. Jochum et al., Mutation of Tyrosine in the Conserved NPXXY Sequence Leads to Constitutive Phosphorylation and Internalization, but Not Signaling, of the Human B2 Bradykinin Receptor, J. Biol. Chem, vol.279, pp.31268-31276, 2004.

Y. Kang, X. E. Zhou, X. Gao, Y. He, W. Liu et al., Crystal structure of rhodopsin bound to arrestin by femtosecond X-ray laser, Nature, vol.523, pp.561-567, 2015.

T. Kanno, A. Gotoh, Y. Fujita, T. Nakano, and T. Nishizaki, A(3) adenosine receptor mediates apoptosis in 5637 human bladder cancer cells by G(q) protein/PKCdependent AIF upregulation, Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol, vol.30, pp.1159-1168, 2012.

R. Van-der-kant and G. Vriend, Alpha-Bulges in G Protein-Coupled Receptors, Int. J. Mol. Sci, vol.15, pp.7841-7864, 2014.

S. Kasina, P. A. Scherle, C. L. Hall, and J. A. Macoska, ADAM-mediated amphiregulin shedding and EGFR transactivation, Cell Prolif, vol.42, pp.799-812, 2009.

H. Katoh, J. Aoki, Y. Yamaguchi, Y. Kitano, A. Ichikawa et al., , 1998.

, Constitutively Active G?12, G?13, and G?q Induce Rho-dependent Neurite Retraction through Different Signaling Pathways, J. Biol. Chem, vol.273, pp.28700-28707

V. Katritch, V. Cherezov, and R. C. Stevens, Diversity and Modularity of G Protein-Coupled Receptor Structures, Trends Pharmacol. Sci, vol.33, pp.17-27, 2012.

K. Kaupmann, B. Malitschek, V. Schuler, J. Heid, W. Froestl et al., GABA(B)-receptor subtypes assemble into functional heteromeric complexes, Nature, vol.396, pp.683-687, 1998.

T. Kenakin, Agonist-receptor efficacy II: agonist trafficking of receptor signals, Trends Pharmacol. Sci, vol.16, pp.232-238, 1995.

D. S. Kim, .. Li, D. Y. Park, P. C. Abrol, D. R. Goddard et al., Prediction of the 3D structure for the rat urotensin II receptor and comparison of the antagonist binding sites and binding selectivity between human and rat from atomistic simulations, ChemMedChem, vol.5, pp.1594-1608, 2010.

J. Kim, S. Ahn, K. Rajagopal, and R. J. Lefkowitz, Independent beta-arrestin2 and Gq/protein kinase Czeta pathways for ERK stimulated by angiotensin type 1A receptors in vascular smooth muscle cells converge on transactivation of the epidermal growth factor receptor, J. Biol. Chem, vol.284, pp.11953-11962, 2009.

W. A. Kinney, H. R. Almond, J. Qi, C. E. Smith, R. J. Santulli et al., Structure-function analysis of urotensin II and its use in the construction of a ligand-receptor working model, Angew. Chem, vol.41, pp.2940-2944, 2002.

B. K. Kobilka, R. A. Dixon, T. Frielle, H. G. Dohlman, M. A. Bolanowski et al., cDNA for the human beta 2-adrenergic receptor: a protein with multiple membrane-spanning domains and encoded by a gene whose chromosomal location is shared with that of the receptor for platelet-derived growth factor, Proc. Natl. Acad. Sci, vol.84, pp.46-50, 1987.

W. J. Koch, B. E. Hawes, L. F. Allen, and R. J. Lefkowitz, Direct evidence that Gicoupled receptor stimulation of mitogen-activated protein kinase is mediated by G beta gamma activation of p21ras, Proc. Natl. Acad. Sci. U. S. A, vol.91, pp.12706-12710, 1994.

M. R. Koelle and H. R. Horvitz, EGL-10 Regulates G Protein Signaling in the C. elegans Nervous System and Shares a Conserved Domain with Many Mammalian Proteins, Cell, vol.84, pp.115-125, 1996.

M. Kojima, R. Haruno, M. Nakazato, Y. Date, N. Murakami et al., Purification and identification of neuromedin U as an endogenous ligand for an orphan receptor GPR66 (FM3), Biochem. Biophys. Res. Commun, vol.276, pp.435-438, 2000.

L. F. Kolakowski, GCRDb: a G-protein-coupled receptor database, Receptors Channels, vol.2, pp.1-7, 1994.

S. Kosugi and T. Mori, The third exoplasmic loop of the thyrotropin receptor is partially involved in signal transduction, FEBS Lett, vol.349, pp.89-92, 1994.

M. Kotani, M. Detheux, A. Vandenbogaerde, D. Communi, J. M. Vanderwinden et al., The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54, J. Biol. Chem, vol.276, pp.34631-34636, 2001.

C. Krasel, M. Bünemann, K. Lorenz, and M. J. Lohse, Beta-arrestin binding to the beta2-adrenergic receptor requires both receptor phosphorylation and receptor activation, J. Biol. Chem, vol.280, pp.9528-9535, 2005.

A. S. Kristof, Z. You, Y. S. Han, and A. Giaid, Protein expression of urotensin II, urotensin-related peptide and their receptor in the lungs of patients with lymphangioleiomyomatosis, Peptides, vol.31, pp.1511-1516, 2010.

B. E. Krumm and R. Grisshammer, Peptide ligand recognition by G protein-coupled receptors, Exp. Pharmacol. Drug Discov, vol.16, p.48, 2015.

I. Kufareva, B. S. Stephens, L. G. Holden, L. Qin, C. Zhao et al., Stoichiometry and geometry of the CXC chemokine receptor 4 complex with CXC ligand 12: molecular modeling and experimental validation, Proc. Natl. Acad. Sci. U. S. A, vol.111, pp.5363-5372, 2014.

A. Kumar, K. N. Kremer, D. Dominguez, M. Tadi, and K. E. Hedin, G?13 and Rho mediate endosomal trafficking of CXCR4 into Rab11+ vesicles upon SDF-1 stimulation, J. Immunol. Baltim. Md, vol.186, pp.951-958, 1950.

P. Labarrere, D. Chatenet, J. Leprince, C. Marionneau, G. Loirand et al., , 2003.

, Structure-activity relationships of human urotensin II and related analogues on rat aortic ring contraction, J. Enzyme Inhib. Med. Chem, vol.18, pp.77-88

M. Lachance, N. Ethier, G. Wolbring, P. P. Schnetkamp, and T. E. Hébert, Stable association of G proteins with beta 2AR is independent of the state of receptor activation, Cell. Signal, vol.11, pp.523-533, 1999.

M. C. Lagerström and H. B. Schiöth, Structural diversity of G protein-coupled receptors and significance for drug discovery, Nat. Rev. Drug Discov, vol.7, pp.339-357, 2008.

E. S. Lander, L. M. Linton, B. Birren, C. Nusbaum, M. C. Zody et al., Initial sequencing and analysis of the human genome, Nature, vol.409, pp.860-921, 2001.

R. G. Langham, D. J. Kelly, R. M. Gow, Y. Zhang, J. K. Dowling et al., Increased expression of urotensin II and urotensin II receptor in human diabetic nephropathy, Am. J. Kidney Dis, vol.44, pp.826-831, 2004.

J. N. Langley, On the reaction of cells and of nerve-endings to certain poisons, chiefly as regards the reaction of striated muscle to nicotine and to curari, J. Physiol, vol.33, pp.374-413, 1905.

S. A. Laporte, R. H. Oakley, J. Zhang, J. A. Holt, S. S. Ferguson et al., The ?2-adrenergic receptor/?arrestin complex recruits the clathrin adaptor AP-2 during endocytosis, Proc. Natl. Acad. Sci, vol.96, pp.3712-3717, 1999.

H. Lapp, G. Boerrigter, L. C. Costello-boerrigter, K. Jaekel, T. Scheffold et al., Elevated plasma human urotensin-IIlike immunoreactivity in ischemic cardiomyopathy, Int. J. Cardiol, vol.94, pp.93-97, 2004.

A. D. Lean, J. M. Stadel, and R. J. Lefkowitz, A ternary complex model explains the agonist-specific binding properties of the adenylate cyclase-coupled beta-adrenergic receptor, J. Biol. Chem, vol.255, pp.7108-7117, 1980.

C. Lecointre, L. Desrues, J. E. Joubert, N. Perzo, P. Guichet et al., Signaling switch of the urotensin II vasosactive peptide GPCR: prototypic chemotaxic mechanism in glioma, Oncogene, vol.34, pp.5080-5094, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02367872

J. Lee and D. R. Welch, Identification of highly expressed genes in metastasissuppressed chromosome 6/human malignant melanoma hybrid cells using subtractive hybridization and differential display, Int. J. Cancer, vol.71, pp.1035-1044, 1997.

D. K. Lee, T. Nguyen, G. P. O'neill, R. Cheng, Y. Liu et al., Discovery of a receptor related to the galanin receptors, FEBS Lett, vol.446, pp.103-107, 1999.

J. H. Lee, M. E. Miele, D. J. Hicks, K. K. Phillips, J. M. Trent et al., KiSS-1, a novel human malignant melanoma metastasis-suppressor gene, J. Natl. Cancer Inst, vol.88, pp.1731-1737, 1996.

J. Y. Lee, J. S. Moon, Y. Eu, C. W. Lee, S. Yang et al., Molecular interaction between kisspeptin decapeptide analogs and a lipid membrane, Arch. Biochem. Biophys, vol.485, pp.109-114, 2009.

C. Y. Lee, Y. T. Tsai, S. H. Loh, J. C. Liu, T. H. Chen et al., Urotensin II induces interleukin 8 expression in human umbilical vein endothelial cells, PloS One, vol.9, p.90278, 2014.

U. Lehner, A. Velic, R. Schroter, E. Schlatter, and A. Sindic, Ligands and signaling of the G-protein-coupled receptor GPR14, expressed in human kidney cells, Cell Physiol. Biochem, vol.20, pp.181-192, 2007.

L. Leloup, H. Shao, Y. H. Bae, B. Deasy, D. Stolz et al., m-Calpain activation is regulated by its membrane localization and by its binding to phosphatidylinositol 4,5-bisphosphate, J. Biol. Chem, vol.285, pp.33549-33566, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01740965

A. D. Leonard, J. P. Thompson, E. L. Hutchinson, S. P. Young, J. Mcdonald et al., Urotensin II receptor expression in human right atrium and aorta: effects of ischaemic heart disease, Br. J. Anaesth, vol.102, pp.477-484, 2009.

J. Leprince, D. Chatenet, C. Dubessy, A. Fournier, B. Pfeiffer et al., Structure-activity relationships of urotensin II and URP, vol.29, pp.658-673, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01972605

M. Leserer, A. Gschwind, and A. Ullrich, Epidermal growth factor receptor signal transactivation, IUBMB Life, vol.49, pp.405-409, 2000.

A. Levoye, K. Balabanian, F. Baleux, F. Bachelerie, and B. Lagane, CXCR7 heterodimerizes with CXCR4 and regulates CXCL12-mediated G protein signaling, Blood, vol.113, pp.6085-6093, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-00415320

B. Li, S. Zhang, H. Zhang, W. Nu, L. Cai et al., Fluoxetinemediated 5-HT2B receptor stimulation in astrocytes causes EGF receptor transactivation and ERK phosphorylation, Psychopharmacology (Berl.), vol.201, pp.443-458, 2008.

Q. Li, A. Roa, I. J. Clarke, and J. T. Smith, Seasonal variation in the gonadotropinreleasing hormone response to kisspeptin in sheep: possible kisspeptin regulation of the kisspeptin receptor, Neuroendocrinology, vol.96, pp.212-221, 2012.

F. Libert, G. Vassart, and M. Parmentier, Current developments in G-proteincoupled receptors, Curr. Opin. Cell Biol, vol.3, pp.218-223, 1991.

F. T. Lin, K. M. Krueger, H. E. Kendall, Y. Daaka, Z. L. Fredericks et al., Clathrin-mediated endocytosis of the beta-adrenergic receptor is regulated by phosphorylation/dephosphorylation of beta-arrestin1, J. Biol. Chem, vol.272, pp.31051-31057, 1997.

Y. Lin, . T. Tsuchihashi, K. Matsumura, I. Abe, and M. Iida, Central cardiovascular action of urotensin II in conscious rats, J. Hypertens, vol.21, pp.159-165, 2003.

Y. Lin, T. Tsuchihashi, K. Matsumura, M. Fukuhara, Y. Ohya et al., Central cardiovascular action of urotensin II in spontaneously hypertensive rats, Hypertension Res, vol.26, pp.839-845, 2003.

H. Lin, M. F. Sassano, B. L. Roth, and B. K. Shoichet, A pharmacological organization of G protein-coupled receptors, Nat. Methods, vol.10, pp.140-146, 2013.

K. Ling, P. Wang, J. Zhao, Y. Wu, Z. Cheng et al., Five-transmembrane domains appear sufficient for a G protein-coupled receptor: Functional five-transmembrane domain chemokine receptors, Proc. Natl. Acad. Sci, vol.96, pp.7922-7927, 1999.

P. J. Little, C. B. Neylon, V. A. Tkachuk, and A. Bobik, Endothelin-1 and endothelin-3 stimulate calcium mobilization by different mechanisms in vascular smooth muscle, Biochem. Biophys. Res. Commun, vol.183, pp.694-700, 1992.

Q. Liu, S. S. Pong, Z. Zeng, Q. Zhang, A. D. Howard et al., Identification of urotensin II as the endogenous ligand for the orphan G-protein-coupled receptor GPR14, Biochem. Biophys. Res. Commun, vol.266, pp.174-178, 1999.

J. C. Liu, C. H. Chen, J. J. Chen, and T. H. Cheng, Urotensin II induces rat cardiomyocyte hypertrophy via the transient oxidization of Src homology 2-containing tyrosine phosphatase and transactivation of epidermal growth factor receptor, Mol. Pharmacol, vol.76, pp.1186-1195, 2009.

J. Liu, Y. Zhao, Y. Sun, B. He, C. Yang et al., Exo70 stimulates the Arp2/3 complex for lamellipodia formation and directional cell migration, Curr. Biol. CB, vol.22, pp.1510-1515, 2012.

J. J. Liu, R. Horst, V. Katritch, R. C. Stevens, and K. Wüthrich, Biased signaling pathways in ?2-adrenergic receptor characterized by 19F-NMR, Science, vol.335, pp.1106-1110, 2012.

P. Liu, P. Long, Y. Huang, F. Sun, W. et al., CXCL12/CXCR4 axis induces proliferation and invasion in human endometrial cancer, Am. J. Transl. Res, vol.8, pp.1719-1729, 2016.

Q. Liu, S. S. Pong, Z. Zeng, Q. Zhang, A. D. Howard et al., Identification of urotensin II as the endogenous ligand for the orphan G-protein-coupled receptor GPR14, Biochem. Biophys. Res. Commun, vol.266, pp.174-178, 1999.

M. Loetscher, T. Geiser, T. O'reilly, R. Zwahlen, M. Baggiolini et al., Cloning of a human seven-transmembrane domain receptor, LESTR, that is highly expressed in leukocytes, J. Biol. Chem, vol.269, pp.232-237, 1994.

D. E. Logothetis, Y. Kurachi, J. Galper, E. J. Neer, and D. E. Clapham, The ?? subunits of GTP-binding proteins activate the muscarinic K+ channel in heart, Nature, vol.325, pp.321-326, 1987.

M. J. Lohse, J. L. Benovic, J. Codina, M. G. Caron, and R. J. Lefkowitz, beta-Arrestin: a protein that regulates beta-adrenergic receptor function, Science, vol.248, pp.1547-1550, 1990.

M. J. Lohse, S. Andexinger, J. Pitcher, S. Trukawinski, J. Codina et al., Receptor-specific desensitization with purified proteins. Kinase dependence and receptor specificity of beta-arrestin and arrestin in the beta 2-adrenergic receptor and rhodopsin systems, J. Biol. Chem, vol.267, pp.8558-8564, 1992.

X. Luo, S. Popov, A. K. Bera, T. M. Wilkie, and S. Muallem, RGS Proteins Provide Biochemical Control of Agonist-Evoked [Ca2+]i Oscillations, Mol. Cell, vol.7, pp.651-660, 2001.

L. M. Luttrell, Minireview: More than just a hammer: ligand "bias" and pharmaceutical discovery, Mol. Endocrinol. Baltim. Md, vol.28, pp.281-294, 2014.

S. Lutz, A. Freichel-blomquist, Y. Yang, U. Rümenapp, K. H. Jakobs et al., The Guanine Nucleotide Exchange Factor p63RhoGEF, a Specific Link between Gq/11-coupled Receptor Signaling and RhoA, J. Biol. Chem, vol.280, pp.11134-11139, 2005.

Q. Ma, D. Jones, P. R. Borghesani, R. A. Segal, T. Nagasawa et al., Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4-and SDF-1-deficient mice, Proc. Natl. Acad. Sci. U. S. A, vol.95, p.9448, 1998.

A. A. Maghazachi, Role of the Heterotrimeric G Proteins in Stromal-Derived Factor-1?-Induced Natural Killer Cell Chemotaxis and Calcium Mobilization, Biochem. Biophys. Res. Commun, vol.236, pp.270-274, 1997.

F. Magnani, Y. Shibata, M. J. Serrano-vega, and C. G. Tate, Co-evolving stability and conformational homogeneity of the human adenosine A2a receptor, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.10744-10749, 2008.

J. J. Maguire, R. E. Kuc, M. J. Kleinz, and A. P. Davenport, Immunocytochemical localization of the urotensin-II receptor, UT, to rat and human tissues: relevance to function, Peptides, vol.29, pp.735-742, 2008.

J. J. Maguire, R. E. Kuc, K. E. Wiley, M. J. Kleinz, and A. P. Davenport, Cellular distribution of immunoreactive urotensin-II in human tissues with evidence of increased expression in atherosclerosis and a greater constrictor response of small compared to large coronary arteries, Peptides, vol.25, pp.1767-1774, 2004.

A. Manglik, A. C. Kruse, T. S. Kobilka, F. S. Thian, J. M. Mathiesen et al., Crystal structure of the µ-opioid receptor bound to a morphinan antagonist, Nature, vol.485, pp.321-326, 2012.

A. Marchese, M. Heiber, T. Nguyen, H. H. Heng, V. R. Saldivia et al., Cloning and Chromosomal Mapping of Three Novel Genes, GPR9, GPR10, and GPR14, Encoding Receptors Related to Interleukin 8, Neuropeptide Y, and Somatostatin Receptors. Genomics, vol.29, pp.335-344, 1995.

J. Marco, E. M. Egido, R. Hernandez, and R. A. Silvestre, Evidence for endogenous urotensin-II as an inhibitor of insulin secretion. Study in the perfused rat pancreas, Peptides, vol.29, pp.852-858, 2008.

M. J. Marinissen, J. Servitja, S. Offermanns, M. I. Simon, and J. S. Gutkind, Thrombin Protease-activated Receptor-1 Signals through Gq-and G13-initiated MAPK Cascades Regulating c-Jun Expression to Induce Cell Transformation, J. Biol. Chem, vol.278, pp.46814-46825, 2003.

A. J. Martínez-fuentes, M. Molina, R. Vázquez-martínez, M. D. Gahete, L. Jiménez-reina et al., Expression of functional KISS1 and KISS1R system is altered in human pituitary adenomas: evidence for apoptotic action of kisspeptin-10, Eur. J. Endocrinol, vol.164, pp.355-362, 2011.

T. Masui, R. Doi, T. Mori, E. Toyoda, M. Koizumi et al., Metastin and its variant forms suppress migration of pancreatic cancer cells, Biochem. Biophys. Res. Commun, vol.315, pp.85-92, 2004.

M. Matsushita, M. Shichiri, T. Imai, M. Iwashina, H. Tanaka et al., Co-expression of urotensin II and its receptor (GPR14) in human cardiovascular and renal tissues, J. Hypertens, vol.19, pp.2185-2190, 2001.

M. Matsushita, M. Shichiri, N. Fukai, N. Ozawa, T. Yoshimoto et al., Urotensin II is an autocrine/paracrine growth factor for the porcine renal epithelial cell line, LLCPK1. Endocrinology, vol.144, pp.1825-1831, 2003.

P. H. Mcdonald, C. W. Chow, W. E. Miller, S. A. Laporte, M. E. Field et al., Beta-arrestin 2: a receptor-regulated MAPK scaffold for the activation of JNK3, Science, vol.290, pp.1574-1577, 2000.

J. Meunier, C. Mollereau, L. Toll, C. Suaudeau, C. Moisand et al., Isolation and structure of the endogenous agonist of opioid receptor-like ORL1 receptor, Nature, vol.377, pp.532-535, 1995.

S. Miura and S. S. Karnik, Constitutive Activation of Angiotensin II Type 1 Receptor Alters the Orientation of Transmembrane Helix-2, J. Biol. Chem, vol.277, pp.24299-24305, 2002.

S. Miura, J. Zhang, J. Boros, and S. S. Karnik, TM2-TM7 Interaction in Coupling Movement of Transmembrane Helices to Activation of the Angiotensin II Type-1, 2003.

, Receptor. J. Biol. Chem, vol.278, pp.3720-3725

C. E. Mona, É. Besserer-offroy, J. Cabana, M. Lefrançois, P. E. Boulais et al., Structure-Activity Relationship and Signaling of New Chimeric CXCR4 Agonists, J. Med. Chem, vol.59, pp.7512-7524, 2016.

C. A. Moore, S. K. Milano, and J. L. Benovic, Regulation of Receptor Trafficking by GRKs and Arrestins, Annu. Rev. Physiol, vol.69, pp.451-482, 2007.

I. S. Moreira, Structural features of the G-protein/GPCR interactions, Biochim. Biophys. Acta BBA -Gen. Subj, vol.1840, pp.16-33, 2014.

M. Mori, T. Sugo, M. Abe, Y. Shimomura, M. Kurihara et al., Urotensin II is the endogenous ligand of a G-protein-coupled orphan receptor, SENR (GPR14), Biochem. Biophys. Res. Commun, vol.265, pp.123-129, 1999.

N. Mori, T. Hirose, T. Nakayama, O. Ito, M. Kanazawa et al., Increased expression of urotensin II-related peptide and its receptor in kidney with hypertension or renal failure, Peptides, vol.30, pp.400-408, 2009.

R. Morimoto, F. Satoh, O. Murakami, K. Totsune, Y. Arai et al., Immunolocalization of urotensin II and its receptor in human adrenal tumors and attached non-neoplastic adrenal tissues, Peptides, vol.29, pp.873-880, 2008.

A. I. Muir, L. Chamberlain, N. A. Elshourbagy, D. Michalovich, D. J. Moore et al., AXOR12, a Novel Human G Protein-coupled Receptor, Activated by the Peptide KiSS-1, J. Biol. Chem, vol.276, pp.28969-28975, 2001.

C. Mukherjee, M. G. Caron, M. Coverstone, and R. J. Lefkowitz, Identification of adenylate cyclase-coupled beta-adrenergic receptors in frog erythrocytes with (minus)-[3-H] alprenolol, J. Biol. Chem, vol.250, pp.4869-4876, 1975.

S. Mukherjee, K. Palczewski, V. Gurevich, J. L. Benovic, J. P. Banga et al., A direct role for arrestins in desensitization of the luteinizing hormone/choriogonadotropin receptor in porcine ovarian follicular membranes, Proc. Natl. Acad. Sci, vol.96, pp.493-498, 1999.

T. Nagasawa, H. Kikutani, and T. Kishimoto, Molecular cloning and structure of a pre-B-cell growth-stimulating factor, Proc. Natl. Acad. Sci. U. S. A, vol.91, p.2305, 1994.

T. Nakayama, T. Hirose, K. Totsune, N. Mori, Y. Maruyama et al., Increased gene expression of urotensin II-related peptide in the hearts of rats with congestive heart failure, Peptides, vol.29, pp.801-808, 2008.

C. Nanoff and G. L. Stiles, Solubilization and Characterization of the A2-Adenosine Receptor, J. Recept. Res, vol.13, pp.961-973, 1993.

J. Nathans and D. S. Hogness, Isolation, sequence analysis, and intron-exon arrangement of the gene encoding bovine rhodopsin, Cell, vol.34, pp.807-814, 1983.

J. Navenot, N. Fujii, and S. C. Peiper, KiSS1 Metastasis Suppressor Gene Product Induces Suppression of Tyrosine Kinase Receptor Signaling to Akt, Tumor Necrosis Factor Family Ligand Expression, and Apoptosis, Mol. Pharmacol, vol.75, pp.1074-1083, 2009.

J. Navenot, N. Fujii, and S. C. Peiper, Activation of Rho and Rho-Associated Kinase by GPR54 and KiSS1 Metastasis Suppressor Gene Product Induces Changes of Cell Morphology and Contributes to Apoptosis, Mol. Pharmacol, vol.75, pp.1300-1306, 2009.

A. Niida, Z. Wang, K. Tomita, S. Oishi, H. Tamamura et al., Design and synthesis of downsized metastin (45-54) analogs with maintenance of high GPR54 agonistic activity, Bioorg. Med. Chem. Lett, vol.16, pp.134-137, 2006.

L. L. Ng, I. Loke, R. J. O'brien, I. B. Squire, and J. E. Davies, Plasma urotensin in human systolic heart failure, Circulation, vol.106, pp.2877-2880, 2002.

T. T. Nguyen, M. Letourneau, D. Chatenet, and A. Fournier, Presence of urotensin-II receptors at the cell nucleus: specific tissue distribution and hypoxia-induced modulation, Int. J. Biochem. Cell Biol, vol.44, pp.639-647, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-00817600

J. Niu, Y. Huang, and L. Zhang, CXCR4 silencing inhibits invasion and migration of human laryngeal cancer Hep-2 cells, Int. J. Clin. Exp. Pathol, vol.8, pp.6255-6261, 2015.

K. N. Nobles, Z. Guan, K. Xiao, T. G. Oas, and R. J. Lefkowitz, The active conformation of beta-arrestin1: direct evidence for the phosphate sensor in the Ndomain and conformational differences in the active states of beta-arrestins1 and -2, J. Biol. Chem, vol.282, pp.21370-21381, 2007.

K. N. Nobles, K. Xiao, S. Ahn, A. K. Shukla, C. M. Lam et al., Distinct phosphorylation sites on the ?(2)-adrenergic receptor establish a barcode that encodes differential functions of ?-arrestin, Sci. Signal, vol.4, p.51, 2011.

J. P. Noel, H. E. Hamm, and P. B. Sigler, The 2.2 A crystal structure of transducinalpha complexed with GTP gamma S, Nature, vol.366, pp.654-663, 1993.
URL : https://hal.archives-ouvertes.fr/jpa-00210878

K. J. Nordstrom, M. Sallman-almen, M. M. Edstam, R. Fredriksson, and H. B. Schioth, Independent HHsearch, Needleman-Wunsch-Based, and Motif Analyses Reveal the Overall Hierarchy for Most of the G Protein-Coupled Receptor Families, Mol. Biol. Evol, vol.28, pp.2471-2480, 2011.

H. Nothacker, Z. Wang, A. M. Mcneill, Y. Saito, S. Merten et al., Identification of the natural ligand of an orphan G-proteincoupled receptor involved in the regulation of vasoconstriction, Nat. Cell Biol, vol.1, pp.383-385, 1999.

R. Nygaard, Y. Zou, R. O. Dror, T. J. Mildorf, D. H. Arlow et al., The Dynamic Process of ?2-Adrenergic Receptor Activation, Cell, vol.152, pp.532-542, 2013.

R. H. Oakley, S. A. Laporte, J. A. Holt, L. S. Barak, C. et al., Association of ?-Arrestin with G Protein-coupled Receptors during Clathrin-mediated Endocytosis Dictates the Profile of Receptor Resensitization, J. Biol. Chem, vol.274, pp.32248-32257, 1999.

R. H. Oakley, S. A. Laporte, J. A. Holt, M. G. Caron, and L. S. Barak, Differential affinities of visual arrestin, beta arrestin1, and beta arrestin2 for G protein-coupled receptors delineate two major classes of receptors, J. Biol. Chem, vol.275, pp.17201-17210, 2000.

J. Joubert,

R. H. Oakley, S. A. Laporte, J. A. Holt, L. S. Barak, C. et al., Molecular Determinants Underlying the Formation of Stable Intracellular G Protein-coupled Receptor-?-Arrestin Complexes after Receptor Endocytosis*, J. Biol. Chem, vol.276, pp.19452-19460, 2001.

E. Oberlin, A. Amara, F. Bachelerie, C. Bessia, J. L. Virelizier et al., The CXC chemokine SDF-1 is the ligand for LESTR/fusin and prevents infection by T-cell-lineadapted HIV-1, Nature, vol.382, pp.833-835, 1996.

T. Ohtaki, Y. Shintani, S. Honda, H. Matsumoto, A. Hori et al., Metastasis suppressor gene KiSS-1 encodes peptide ligand of a G-protein-coupled receptor, Nature, vol.411, pp.613-617, 2001.

T. Olbrich, E. Ziegler, G. Türk, A. Schubert, G. Emons et al., Kisspeptin-10 inhibits bone-directed migration of GPR54-positive breast cancer cells: Evidence for a dose-window effect, Gynecol. Oncol, vol.119, pp.571-578, 2010.

W. M. Oldham and H. E. Hamm, Heterotrimeric G protein activation by G-proteincoupled receptors, Nat. Rev. Mol. Cell Biol, vol.9, pp.60-71, 2008.

W. M. Oldham, N. Van-eps, A. M. Preininger, W. L. Hubbell, and H. E. Hamm, Mechanism of the receptor-catalyzed activation of heterotrimeric G proteins, Nat. Struct. Mol. Biol, vol.13, pp.772-777, 2006.

D. Onan, R. D. Hannan, and W. G. Thomas, Urotensin II: the old kid in town, Trends Endocrinol. Metab. TEM, vol.15, pp.175-182, 2004.

A. M. Onat, Y. Pehlivan, I. H. Turkbeyler, T. Demir, D. S. Kaplan et al., Urotensin inhibition with palosuran could be a promising alternative in pulmonary arterial hypertension, Inflammation, vol.36, pp.405-412, 2013.

A. M. Onat, I. H. Turkbeyler, Y. Pehlivan, T. Demir, D. S. Kaplan et al., The efficiency of a urotensin II antagonist in an experimental lung fibrosis model, Inflammation, vol.35, pp.1138-1143, 2012.

O. S. Opgaard, H. Nothacker, F. J. Ehlert, and D. N. Krause, Human urotensin II mediates vasoconstriction via an increase in inositol phosphates, Eur. J. Pharmacol, vol.406, pp.265-271, 2000.

J. Joubert,

K. Palczewski, J. Buczy?ko, M. W. Kaplan, A. S. Polans, and J. W. Crabb, Mechanism of rhodopsin kinase activation, J. Biol. Chem, vol.266, pp.12949-12955, 1991.

K. Palczewski, T. Kumasaka, T. Hori, C. A. Behnke, H. Motoshima et al., Crystal Structure of Rhodopsin: A G Protein-Coupled Receptor. Science, vol.289, pp.739-745, 2000.

M. Pampillo, N. Camuso, J. E. Taylor, J. M. Szereszewski, M. R. Ahow et al., Regulation of GPR54 Signaling by GRK2 and ?-Arrestin, Mol. Endocrinol, vol.23, pp.2060-2074, 2009.

C. Parmentier, E. Hameury, C. Dubessy, F. B. Quan, D. Habert et al., Occurrence of two distinct urotensin II-related peptides in zebrafish provides new insight into the evolutionary history of the urotensin II gene family, Endocrinology, vol.152, pp.2330-2341, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02179710

C. H. Park, J. H. Lee, M. Y. Lee, J. H. Lee, B. H. Lee et al., A novel role of G protein-coupled receptor kinase 5 in urotensin II-stimulated cellular hypertrophy in H9c2UT cells, Mol. Cell. Biochem, vol.422, pp.151-160, 2016.

J. H. Park, P. Scheerer, K. P. Hofmann, H. Choe, and O. P. Ernst, Crystal structure of the ligand-free G-protein-coupled receptor opsin, Nature, vol.454, pp.183-187, 2008.

M. S. Parker, Y. Y. Wong, and S. L. Parker, An ion-responsive motif in the second transmembrane segment of rhodopsin-like receptors, Amino Acids, vol.35, pp.1-15, 2008.

R. Patacchini, P. Santicioli, S. Giuliani, P. Grieco, E. Novellino et al., Urantide: an ultrapotent urotensin II antagonist peptide in the rat aorta, Br. J. Pharmacol, vol.140, pp.1155-1158, 2003.

D. Pearson, J. E. Shively, B. R. Clark, I. I. Geschwind, M. Barkley et al., Urotensin II: a somatostatin-like peptide in the caudal neurosecretory system of fishes, Proc. Natl. Acad. Sci. U. S. A, vol.77, pp.5021-5024, 1980.

Y. Pehlivan, R. Dokuyucu, T. Demir, D. S. Kaplan, I. Koc et al., Palosuran treatment effective as bosentan in the treatment model of pulmonary arterial hypertension, Inflammation, vol.37, pp.1280-1288, 2014.

J. Pelé, H. Abdi, M. Moreau, D. Thybert, and M. Chabbert, Multidimensional Scaling Reveals the Main Evolutionary Pathways of Class A G-Protein-Coupled Receptors, PLoS ONE, vol.6, 2011.

G. Pelletier, I. Lihrmann, and H. Vaudry, Role of androgens in the regulation of urotensin II precursor mRNA expression in the rat brainstem and spinal cord, Neuroscience, vol.115, pp.525-532, 2002.

S. Peng, X. Zhang, D. Paul, L. M. Kays, M. Ye et al., Inhibition of CXCR4 by LY2624587, a Fully Humanized Anti-CXCR4 Antibody Induces Apoptosis of Hematologic Malignancies, PloS One, vol.11, 2016.

J. M. Perez-aguilar, J. Shan, M. V. Levine, G. Khelashvili, and H. Weinstein, A Functional Selectivity Mechanism at the Serotonin-2A GPCR Involves Ligand-Dependent Conformations of Intracellular Loop 2, J. Am. Chem. Soc, vol.136, pp.16044-16054, 2014.

T. D. Perkins, S. Bansal, and D. J. Barlow, Molecular modelling and design of analogues of the peptide hormone urotensin II, Biochem. Soc. transactions, vol.18, pp.918-919, 1990.

J. H. Perlman, W. Wang, D. R. Nussenzveig, and M. C. Gershengorn, A Disulfide Bond between Conserved Extracellular Cysteines in the Thyrotropin-releasing Hormone Receptor Is Critical for Binding, J. Biol. Chem, vol.270, pp.24682-24685, 1995.

F. C. Peterson, J. A. Thorpe, A. G. Harder, B. F. Volkman, and S. R. Schwarze, Structural determinants involved in the regulation of CXCL14/BRAK expression by the 26 S proteasome, J. Mol. Biol, vol.363, pp.813-822, 2006.

R. J. Petrie, A. D. Doyle, and K. M. Yamada, Random versus directionally persistent cell migration, Nat. Rev. Mol. Cell Biol, vol.10, pp.538-549, 2009.

W. J. Phillips and R. A. Cerione, Rhodopsin/transducin interactions. I. Characterization of the binding of the transducin-beta gamma subunit complex to rhodopsin using fluorescence spectroscopy, J. Biol. Chem, vol.267, pp.17032-17039, 1992.

J. Pin, J. Kniazeff, C. Goudet, A. Bessis, J. Liu et al., The activation mechanism of class-C G-protein coupled receptors, Biol. Cell, vol.96, pp.335-342, 2004.

L. Pinilla, E. Aguilar, C. Dieguez, R. P. Millar, and M. Tena-sempere, Kisspeptins and Reproduction: Physiological Roles and Regulatory Mechanisms, Physiol. Rev, vol.92, pp.1235-1316, 2012.

J. A. Pitcher, J. Inglese, J. B. Higgins, J. L. Arriza, P. J. Casey et al., Role of beta gamma subunits of G proteins in targeting the beta-adrenergic receptor kinase to membrane-bound receptors, Science, vol.257, pp.1264-1267, 1992.

T. Pozzobon, G. Goldoni, A. Viola, and B. Molon, CXCR4 signaling in health and disease, Immunol. Lett, vol.177, pp.6-15, 2016.

A. A. Pradhan, J. Perroy, W. M. Walwyn, M. L. Smith, A. Vicente-sanchez et al., Agonist-Specific Recruitment of Arrestin Isoforms Differentially Modify Delta Opioid Receptor Function, J. Neurosci. Off. J. Soc. Neurosci, vol.36, pp.3541-3551, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01936373

A. M. Preininger, N. Van-eps, N. Yu, M. Medkova, W. L. Hubbell et al., The myristoylated amino terminus of Galpha(i)(1) plays a critical role in the structure and function of Galpha(i)(1) subunits in solution, Biochemistry (Mosc.), vol.42, pp.7931-7941, 2003.

N. Prenzel, E. Zwick, H. Daub, M. Leserer, R. Abraham et al., EGF receptor transactivation by G-protein-coupled receptors requires metalloproteinase cleavage of proHB-EGF, Nature, vol.402, pp.884-888, 1999.

C. Prioleau, I. Visiers, B. J. Ebersole, H. Weinstein, and S. C. Sealfon, Conserved Helix 7 Tyrosine Acts as a Multistate Conformational Switch in the 5HT2C Receptor. Identification of a novel "locked-on" phenotype and double reversant mutations, J. Biol. Chem, vol.277, pp.36577-36584, 2002.

H. C. Prosser, M. E. Forster, A. M. Richards, and C. J. Pemberton, Urotensin II and urotensin II-related peptide (URP) in cardiac ischemia-reperfusion injury, Peptides, vol.29, pp.770-777, 2008.

C. D. Proulx, M. Simaan, E. Escher, S. A. Laporte, G. Guillemette et al., Involvement of a cytoplasmic-tail serine cluster in urotensin II receptor internalization, Biochem. J, vol.385, pp.115-123, 2005.

C. D. Proulx, B. J. Holleran, P. Lavigne, E. Escher, G. Guillemette et al., Biological properties and functional determinants of the urotensin II receptor, Peptides, vol.29, pp.691-699, 2008.

C. D. Proulx, B. J. Holleran, A. A. Boucard, E. Escher, G. Guillemette et al., Mutational Analysis of the Conserved Asp2.50 and ERY Motif Reveals Signaling Bias of the Urotensin II Receptor, Mol. Pharmacol, vol.74, pp.552-561, 2008.

J. Qi, J. Du, X. Tang, J. Li, B. Wei et al., The upregulation of endothelial nitric oxide synthase and urotensin-II is associated with pulmonary hypertension and vascular diseases in rats produced by aortocaval shunting, Heart and vessels, vol.19, pp.81-88, 2004.

J. S. Qi, R. Schulingkamp, T. J. Parry, R. Colburn, D. Stone et al., Urotensin-II induces ear flushing in rats, Br. J. Pharmacol, vol.150, pp.415-423, 2007.

F. Qiu, Y. Li, Q. Fu, Y. Fan, C. Zhu et al., Stromal Cell-Derived Factor 1 Increases Tetrodotoxin-Resistant Sodium Currents Nav1.8 and Nav1.9 in Rat Dorsal Root Ganglion Neurons via Different Mechanisms, Neurochem. Res, vol.41, pp.1587-1603, 2016.

J. Joubert,

J. Quoyer, J. M. Janz, J. Luo, Y. Ren, S. Armando et al., Pepducin targeting the C-X-C chemokine receptor type 4 acts as a biased agonist favoring activation of the inhibitory G protein, Proc Natl Acad Sci U A E5088-97, 2013.

K. M. Raehal, J. K. Walker, and L. M. Bohn, Morphine Side Effects in ?-Arrestin 2 Knockout Mice, J. Pharmacol. Exp. Ther, vol.314, pp.1195-1201, 2005.

E. Rakowski, G. S. Hassan, D. Dhanak, E. H. Ohlstein, S. A. Douglas et al., A role for urotensin II in restenosis following balloon angioplasty: use of a selective UT receptor blocker, J. Mol. Cell. Cardiol, vol.39, pp.785-791, 2005.

M. Rask-andersen, M. S. Almén, and H. B. Schiöth, Trends in the exploitation of novel drug targets, Nat. Rev. Drug Discov, vol.10, pp.579-590, 2011.

S. G. Rasmussen, A. D. Jensen, G. Liapakis, P. Ghanouni, J. A. Javitch et al., Mutation of a highly conserved aspartic acid in the beta2 adrenergic receptor: constitutive activation, structural instability, and conformational rearrangement of transmembrane segment 6, Mol. Pharmacol, vol.56, pp.175-184, 1999.

S. G. Rasmussen, H. Choi, D. M. Rosenbaum, T. S. Kobilka, F. S. Thian et al., Crystal structure of the human ?2 adrenergic G-protein-coupled receptor, Nature, vol.450, pp.383-387, 2007.

S. G. Rasmussen, B. T. Devree, Y. Zou, A. C. Kruse, K. Y. Chung et al., Crystal structure of the ?2 adrenergic receptor-Gs protein complex, Nature, vol.477, pp.549-555, 2011.

R. I. Reis, E. L. Santos, J. B. Pesquero, L. Oliveira, J. P. Schanstra et al., Participation of transmembrane proline 82 in angiotensin II AT1 receptor signal transduction, Regul. Pept, vol.140, pp.32-36, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00409738

X. Ren, E. Reiter, S. Ahn, J. Kim, W. Chen et al., Different G protein-coupled receptor kinases govern G protein and beta-arrestin-mediated signaling of V2 vasopressin receptor, Proc. Natl. Acad. Sci. U.S.A, vol.102, pp.1448-1453, 2005.

J. Rey, J. Deville, and M. Chabbert, Structural determinants stabilizing helical distortions related to proline, J. Struct. Biol, vol.171, pp.266-276, 2010.

A. M. Richards, M. G. Nicholls, J. G. Lainchbury, S. Fisher, and T. G. Yandle, Plasma urotensin II in heart failure, Lancet, vol.360, pp.545-546, 2002.

E. V. Romanova, K. Sasaki, V. Alexeeva, F. S. Vilim, J. Jing et al., Urotensin II in invertebrates: from structure to function in Aplysia californica, PloS one, vol.7, p.48764, 2012.

D. M. Rosenbaum, S. G. Rasmussen, and B. K. Kobilka, The structure and function of G-protein-coupled receptors, Nature, vol.459, pp.356-363, 2009.

D. M. Rosenbaum, C. Zhang, J. Lyons, R. Holl, D. Aragao et al., Structure and Function of an Irreversible Agonist-?2 Adrenoceptor complex, Nature, vol.469, pp.236-240, 2011.

A. K. Roseweir, A. A. Katz, and R. P. Millar, Kisspeptin-10 inhibits cell migration in vitro via a receptor-GSK3 beta-FAK feedback loop in HTR8SVneo cells, Placenta, vol.33, pp.408-415, 2012.

E. M. Ross and T. M. Wilkie, GTPase-Activating Proteins for Heterotrimeric G Proteins: Regulators of G Protein Signaling (RGS) and RGS-Like Proteins, Annu. Rev. Biochem, vol.69, pp.795-827, 2000.

B. Ross, K. Mckendy, and A. Giaid, Role of urotensin II in health and disease, Am. J. Physiol. Regul. Integr. Comp. Physiol, vol.298, pp.1156-1172, 2010.

W. J. Rossowski, B. Cheng, J. E. Taylor, R. Datta, and D. H. Coy, Human urotensin II-induced aorta ring contractions are mediated by protein kinase C, tyrosine kinases and Rho-kinase: inhibition by somatostatin receptor antagonists, Eur. J. Pharmacol, vol.438, pp.159-170, 2002.

E. T. Roussos, J. S. Condeelis, and A. Patsialou, Chemotaxis in cancer, Nat. Rev. Cancer, vol.11, pp.573-587, 2011.

A. A. Roy, C. Nunn, H. Ming, M. Zou, J. Penninger et al., Up-regulation of endogenous RGS2 mediates crossdesensitization between Gs and Gq signaling in osteoblasts, J. Biol. Chem, vol.281, pp.32684-32693, 2006.

J. B. Rubin, Chemokine Signaling in Cancer: One Hump or Two? Semin, Cancer Biol, vol.19, p.116, 2009.

F. D. Russell, P. Molenaar, and D. M. O'brien, Cardiostimulant effects of urotensin-II in human heart in vitro, Br. J. Pharmacol, vol.132, pp.5-9, 2001.

F. D. Russell, D. Meyers, A. J. Galbraith, N. Bett, I. Toth et al., Elevated plasma levels of human urotensin-II immunoreactivity in congestive heart failure, Am. J. Physiol. Heart Circ. Physiol, vol.285, pp.1576-1581, 2003.

F. D. Russell and P. Molenaar, Investigation of signaling pathways that mediate the inotropic effect of urotensin-II in human heart, Cardiovasc. Res, vol.63, pp.673-681, 2004.

E. K. Ryu, T. G. Kim, T. H. Kwon, I. D. Jung, D. Ryu et al., Crystal structure of recombinant human stromal cell-derived factor-1?, Proteins Struct. Funct. Bioinforma, vol.67, pp.1193-1197, 2007.

X. Sainsily, J. Cabana, P. E. Boulais, B. J. Holleran, E. Escher et al., Identification of transmembrane domain 3, 4 & 5 residues that contribute to the formation of the ligand-binding pocket of the urotensin-II receptor, Biochem. Pharmacol, vol.86, pp.1584-1593, 2013.

X. Sainsily, J. Cabana, B. J. Holleran, E. Escher, P. Lavigne et al., Identification of transmembrane domain 1 & 2 residues that contribute to the formation of the ligand-binding pocket of the urotensin-II receptor, Biochem. Pharmacol, vol.92, pp.280-288, 2014.

G. Sánchez-fernández, S. Cabezudo, C. García-hoz, A. B. Tobin, F. Mayor et al., ERK5 activation by Gq-coupled muscarinic receptors is independent of receptor internalization and ?-arrestin recruitment, PloS One, vol.8, p.84174, 2013.

A. Saulière, M. Bellot, H. Paris, C. Denis, F. Finana et al., Deciphering biased-agonism complexity reveals a new active AT1 receptor entity, Nat. Chem. Biol, vol.8, pp.622-630, 2012.

V. Sauzeau, E. Le-mellionnec, J. Bertoglio, E. Scalbert, P. Pacaud et al., Human urotensin II-induced contraction and arterial smooth muscle cell proliferation are mediated by RhoA and Rho-kinase, Circ. Res, vol.88, pp.1102-1104, 2001.

T. M. Savarese, C. D. Wang, and C. M. Fraser, Site-directed mutagenesis of the rat m1 muscarinic acetylcholine receptor. Role of conserved cysteines in receptor function, J. Biol. Chem, vol.267, pp.11439-11448, 1992.

P. Scheerer, J. H. Park, P. W. Hildebrand, Y. J. Kim, N. Krauss et al., Crystal structure of opsin in its G-protein-interacting conformation, Nature, vol.455, pp.497-502, 2008.

O. Schoots, J. M. Wilson, N. Ethier, E. Bigras, T. E. Hebert et al., Coexpression of human Kir3 subunits can yield channels with different functional properties, Cell. Signal, vol.11, pp.871-883, 1999.

J. Segain, M. Rolli-derkinderen, N. Gervois, D. Raingeard-de-la-blétière, G. Loirand et al., Urotensin II is a new chemotactic factor for UT receptorexpressing monocytes, J. Immunol. Baltim. Md, vol.179, pp.901-909, 1950.

J. Joubert,

J. Shan, H. Weinstein, and E. L. Mehler, Probing the Structural Determinants for the Function of Intracellular Loop 2 in Structurally Cognate G-Protein-Coupled Receptors, Biochemistry (Mosc.), vol.49, pp.10691-10701, 2010.

H. Shao, J. Chou, C. J. Baty, N. A. Burke, S. C. Watkins et al., Spatial localization of m-calpain to the plasma membrane by phosphoinositide biphosphate binding during epidermal growth factor receptor-mediated activation, Mol. Cell. Biol, vol.26, pp.5481-5496, 2006.

D. A. Shapiro, K. Kristiansen, D. M. Weiner, W. K. Kroeze, and B. L. Roth, Evidence for a Model of Agonist-induced Activation of 5-Hydroxytryptamine 2A Serotonin Receptors That Involves the Disruption of a Strong Ionic Interaction between Helices 3 and 6, J. Biol. Chem, vol.277, pp.11441-11449, 2002.

A. Shenouda, S. A. Douglas, E. H. Ohlstein, and A. Giaid, Localization of urotensin-II immunoreactivity in normal human kidneys and renal carcinoma, J. Histochem. Cytochem, vol.50, pp.885-889, 2002.

L. Shi, G. Liapakis, R. Xu, F. Guarnieri, J. A. Ballesteros et al., , p.2, 2002.

, Adrenergic Receptor Activation modulation of the proline kink in transmembrane 6 by a rotamer toggel switch, J. Biol. Chem, vol.277, pp.40989-40996

L. Shi, G. Liapakis, R. Xu, F. Guarnieri, J. A. Ballesteros et al., Beta2 adrenergic receptor activation. Modulation of the proline kink in transmembrane 6 by a rotamer toggle switch, J. Biol. Chem, vol.277, pp.40989-40996, 2002.

T. Shimamura, M. Shiroishi, S. Weyand, H. Tsujimoto, G. Winter et al., Structure of the human histamine H1 receptor complex with doxepin, Nature, vol.475, pp.65-70, 2011.

R. Shin, D. R. Welch, V. K. Mishra, K. T. Nash, D. R. Hurst et al., Nuclear magnetic resonance and circular dichroism study of metastin (Kisspeptin-54) structure in solution, Clin. Exp. Metastasis, vol.26, pp.527-533, 2009.

A. K. Shukla, A. Manglik, A. C. Kruse, K. Xiao, R. I. Reis et al., Structure of active ?-arrestin-1 bound to a G-protein-coupled receptor phosphopeptide, Nature, vol.497, pp.137-141, 2013.

A. K. Shukla, G. H. Westfield, K. Xiao, R. I. Reis, L. Huang et al., Visualization of arrestin recruitment by a G-protein-coupled receptor, Nature, vol.512, pp.218-222, 2014.

P. N. Sidharta, F. D. Wagner, H. Bohnemeier, A. Jungnik, A. Halabi et al., Pharmacodynamics and pharmacokinetics of the urotensin II receptor antagonist palosuran in macroalbuminuric, diabetic patients, Clin. Pharmacol. Ther, vol.80, pp.246-256, 2006.

R. A. Silvestre, E. M. Egido, R. Hernandez, J. Leprince, D. Chatenet et al., Urotensin-II is present in pancreatic extracts and inhibits insulin release in the perfused rat pancreas, Eur. J. Endocrinol, 2004.
URL : https://hal.archives-ouvertes.fr/hal-01974193

J. , , vol.151, pp.803-809

M. I. Simon, M. P. Strathmann, and N. Gautam, Diversity of G proteins in signal transduction, Science, vol.252, pp.802-808, 1991.

H. Smith, C. Whittall, B. Weksler, and J. Middleton, Chemokines stimulate bidirectional migration of human mesenchymal stem cells across bone marrow endothelial cells, Stem Cells Dev, vol.21, pp.476-486, 2012.

W. Song, J. Mcdonald, V. Camarda, G. Calo, R. Guerrini et al., Cell and tissue responses of a range of Urotensin II analogs at cloned and native urotensin II receptors. Evidence for coupling promiscuity, Naunyn. Schmiedebergs Arch. Pharmacol, vol.373, pp.148-157, 2006.

S. R. Sprang, G protein mechanisms:Insights from Structural Analysis, Annu. Rev. Biochem, vol.66, pp.639-678, 1997.

A. Srivastava, B. Gupta, C. Gupta, and A. K. Shukla, Emerging Functional Divergence of ?-Arrestin Isoforms in GPCR Function, Trends Endocrinol. Metab. TEM, vol.26, pp.628-642, 2015.

J. Standfuss, P. C. Edwards, A. D'antona, M. Fransen, G. Xie et al., The structural basis of agonist-induced activation in constitutively active rhodopsin, Nature, vol.471, pp.656-660, 2011.

M. Stathaki, A. Armakolas, A. Dimakakos, L. Kaklamanis, I. Vlachos et al., Kisspeptin effect on endothelial monocyte activating polypeptide II (EMAP-II)-associated lymphocyte cell death and metastases in colorectal cancer patients, Mol. Med. Camb. Mass, vol.20, pp.80-92, 2014.

A. Steen, S. Thiele, D. Guo, L. S. Hansen, T. M. Frimurer et al., Biased and constitutive signaling in the CC-chemokine receptor CCR5 by manipulating the interface between transmembrane helices 6 and 7, J. Biol. Chem, vol.288, pp.12511-12521, 2013.

Y. M. Sue, C. H. Chen, Y. H. Hsu, C. C. Hou, C. Y. Cheng et al., Urotensin II induces transactivation of the epidermal growth factor receptor via transient oxidation of SHP-2 in the rat renal tubular cell line NRK-52E, Growth factors, vol.27, pp.155-162, 2009.

T. Sugo and M. Mori, Another ligand fishing for G protein-coupled receptor 14. Discovery of urotensin II-related peptide in the rat brain, Peptides, vol.29, pp.809-812, 2008.

T. Sugo, Y. Murakami, Y. Shimomura, M. Harada, M. Abe et al., Identification of urotensin IIrelated peptide as the urotensin II-immunoreactive molecule in the rat brain, Biochem. Biophys. Res. Commun, vol.310, pp.860-868, 2003.

Y. Sun, Z. Cheng, L. Ma, P. , and G. , ?-Arrestin2 Is Critically Involved in CXCR4-mediated Chemotaxis, and This Is Mediated by Its Enhancement of p38 MAPK Activation, J. Biol. Chem, vol.277, pp.49212-49219, 2002.

J. M. Szereszewski, M. Pampillo, M. R. Ahow, S. Offermanns, M. Bhattacharya et al., GPR54 Regulates ERK1/2 Activity and Hypothalamic Gene Expression in a G? q/11 and ?-Arrestin-Dependent Manner, Plos One, vol.5, 2010.

M. Tal, D. A. Ammar, M. Karpuj, V. Krizhanovsky, M. Naim et al., A Novel Putative Neuropeptide Receptor Expressed in Neural Tissue, Including Sensory Epithelia, Biochem. Biophys. Res. Commun, vol.209, pp.752-759, 1995.

K. Tan, S. Cho, W. Luo, T. Yi, X. Wu et al., , 2014.

, KiSS1-induced GPR54 signaling inhibits breast cancer cell migration and epithelialmesenchymal transition via protein kinase D1, Curr. Mol. Med, vol.14, pp.652-662

Q. Tan, Y. Zhu, J. Li, Z. Chen, G. W. Han et al., Structure of the CCR5 chemokine receptor-HIV entry inhibitor maraviroc complex, Science, vol.341, pp.1387-1390, 2013.

W. Tan, D. Martin, and J. S. Gutkind, The G?13-Rho Signaling Axis Is Required for SDF-1-induced Migration through CXCR4, J. Biol. Chem, vol.281, pp.39542-39549, 2006.

X. Tang, Y. Wang, D. Li, J. Luo, and M. Liu, Orphan G protein-coupled receptors (GPCRs): biological functions and potential drug targets, Acta Pharmacol. Sin, vol.33, pp.363-371, 2012.

X. Tang, R. Jin, G. Qu, X. Wang, Z. Li et al., GPR116, an adhesion G-protein-coupled receptor, promotes breast cancer metastasis via the G?q-p63RhoGEF-Rho GTPase pathway, Cancer Res, vol.73, pp.6206-6218, 2013.

K. Tashiro, H. Tada, R. Heilker, M. Shirozu, T. Nakano et al., Signal sequence trap: a cloning strategy for secreted proteins and type I membrane proteins, Science, vol.261, pp.600-603, 1993.

C. S. Tautermann, GPCR structures in drug design, emerging opportunities with new structures, Bioorg. Med. Chem. Lett, vol.24, pp.4073-4079, 2014.

J. M. Taylor and R. R. Neubig, Peptides as probes for G protein signal transduction, Cell. Signal, vol.6, pp.841-849, 1994.

J. Joubert,

B. Trzaskowski, D. Latek, S. Yuan, U. Ghoshdastider, A. Debinski et al., Action of Molecular Switches in GPCRs -Theoretical and Experimental Studies, Curr. Med. Chem, vol.19, pp.1090-1109, 2012.

C. S. Tsai, S. H. Loh, J. C. Liu, J. W. Lin, Y. L. Chen et al., Urotensin II-induced endothelin-1 expression and cell proliferation via epidermal growth factor receptor transactivation in rat aortic smooth muscle cells, Atherosclerosis, vol.206, pp.86-94, 2009.

A. Tzanidis, R. D. Hannan, W. G. Thomas, D. Onan, D. J. Autelitano et al., Direct actions of urotensin II on the heart: implications for cardiac fibrosis and hypertrophy, Circ. Res, vol.93, pp.246-253, 2003.

Y. Uchi, H. Takeuchi, S. Matsuda, Y. Saikawa, H. Kawakubo et al., CXCL12 expression promotes esophageal squamous cell carcinoma proliferation and worsens the prognosis, BMC Cancer, vol.16, 2016.

J. Van-unen, N. R. Reinhard, T. Yin, Y. I. Wu, M. Postma et al., Plasma membrane restricted RhoGEF activity is sufficient for RhoAmediated actin polymerization, Sci. Rep, vol.5, p.14693, 2015.

C. T. Veldkamp, F. C. Peterson, A. J. Pelzek, and B. F. Volkman, The monomer-dimer equilibrium of stromal cell-derived factor-1 (CXCL 12) is altered by pH, phosphate, sulfate, and heparin, Protein Sci. Publ. Protein Soc, vol.14, pp.1071-1081, 2005.

C. T. Veldkamp, C. Seibert, F. C. Peterson, N. B. De-la-cruz, J. C. Haugner et al., Structural Basis of CXCR4 Sulfotyrosine Recognition by the Chemokine SDF-1/CXCL12, Sci. Signal, vol.1, pp.4-4, 2008.

A. J. Venkatakrishnan, X. Deupi, G. Lebon, C. G. Tate, G. F. Schertler et al., Molecular signatures of G-protein-coupled receptors, Nature, vol.494, pp.185-194, 2013.

R. Vergura, V. Camarda, A. Rizzi, M. Spagnol, R. Guerrini et al., Urotensin II stimulates plasma extravasation in mice via UT receptor activation, Naunyn. Schmiedebergs Arch. Pharmacol, vol.370, pp.347-352, 2004.

J. Joubert,

A. J. Vila-coro, J. M. Rodríguez-frade, A. M. Ana, M. C. Moreno-ortíz, C. Martínez-a et al., The chemokine SDF-1? triggers CXCR4 receptor dimerization and activates the JAK/STAT pathway, FASEB J, vol.13, pp.1699-1710, 1999.

A. Viola and A. D. Luster, Chemokines and their receptors: drug targets in immunity and inflammation, Annu. Rev. Pharmacol. Toxicol, vol.48, pp.171-197, 2008.

J. D. Violin and R. J. Lefkowitz, ?-Arrestin-biased ligands at seven-transmembrane receptors, Trends Pharmacol. Sci, vol.28, pp.416-422, 2007.

J. D. Violin, S. M. Dewire, D. Yamashita, D. H. Rominger, L. Nguyen et al., Selectively Engaging ?-Arrestins at the Angiotensin II Type 1 Receptor Reduces Blood Pressure and Increases Cardiac Performance, J. Pharmacol. Exp. Ther, vol.335, pp.572-579, 2010.

I. Visiers, J. A. Ballesteros, and H. Weinstein, Three-dimensional representations of G protein-coupled receptor structures and mechanisms, Methods Enzymol, vol.343, pp.329-371, 2002.

S. Vogt, R. Grosse, G. Schultz, and S. Offermanns, Receptor-dependent RhoA activation in G12/G13-deficient cells genetic evidence for an involvement of Gq/G11, J. Biol. Chem, vol.278, pp.28743-28749, 2003.

P. Voigt, C. Brock, B. Nürnberg, and M. Schaefer, Assigning Functional Domains within the p101 Regulatory Subunit of Phosphoinositide 3-Kinase ?, J. Biol. Chem, vol.280, pp.5121-5127, 2005.

J. L. Wacker, D. B. Feller, X. Tang, M. C. Defino, Y. Namkung et al., Disease-causing Mutation in GPR54 Reveals the Importance of the Second Intracellular Loop for Class A G-proteincoupled Receptor Function, J. Biol. Chem, vol.283, pp.31068-31078, 2008.

M. A. Wall, D. E. Coleman, E. Lee, J. A. Iñiguez-lluhi, B. A. Posner et al., The structure of the G protein heterotrimer Gi?1?1?2, Cell, vol.83, pp.1047-1058, 1995.

T. G. Walsh, M. T. Harper, and A. W. Poole, SDF-1? is a novel autocrine activator of platelets operating through its receptor CXCR4, Cell. Signal, vol.27, pp.37-46, 2015.

T. Wang, S. Pentyala, J. T. Elliott, L. Dowal, E. Gupta et al., Selective interaction of the C2 domains of phospholipase C-beta1 and -beta2 with activated Galphaq subunits: an alternative function for C2-signaling modules, Proc. Natl. Acad. Sci. U. S. A, vol.96, pp.7843-7846, 1999.

H. Wang, K. Dong, X. Xue, P. Feng, and X. Wang, Elevated expression of urotensin II and its receptor in diethylnitrosamine-mediated precancerous lesions in rat liver, Peptides, vol.32, pp.382-387, 2011.

H. X. Wang, X. J. Zeng, Y. Liu, J. Wang, L. Q. Lu et al., Elevated expression of urotensin II and its receptor in skeletal muscle of diabetic mouse, Regul. Pept, vol.154, pp.85-90, 2009.

T. Warne, M. J. Serrano-vega, J. G. Baker, R. Moukhametzianov, P. C. Edwards et al., Structure of a ?1-adrenergic G protein-coupled receptor, Nature, vol.454, pp.486-491, 2008.

T. Watanabe, R. Pakala, T. Katagiri, and C. R. Benedict, Synergistic effect of urotensin II with serotonin on vascular smooth muscle cell proliferation, J. Hypertens, vol.19, pp.2191-2196, 2001.

D. J. Webb, K. Donais, L. A. Whitmore, S. M. Thomas, C. E. Turner et al., FAK-Src signalling through paxillin, ERK and MLCK regulates adhesion disassembly, Nat. Cell Biol, vol.6, pp.154-161, 2004.

M. Weber, L. Tome, D. Otzen, and D. Schneider, A Ser residue influences the structure and stability of a Pro-kinked transmembrane helix dimer, Biochim. Biophys. Acta BBA -Biomembr, vol.1818, pp.2103-2107, 2012.

H. Wei, S. Ahn, S. K. Shenoy, S. S. Karnik, L. Hunyady et al., Independent beta-arrestin 2 and G protein-mediated pathways for angiotensin II activation of extracellular signal-regulated kinases 1 and 2, Proc. Natl. Acad. Sci. U. S. A, vol.100, pp.10782-10787, 2003.

L. Wei, B. Zhang, W. Cao, H. Xing, X. Yu et al., Inhibition of CXCL12/CXCR4 suppresses pulmonary arterial smooth muscle cell proliferation and cell cycle progression via PI3K/Akt pathway under hypoxia, J. Recept. Signal Transduct. Res, vol.35, pp.329-339, 2015.

M. P. Wescott, I. Kufareva, C. Paes, J. R. Goodman, Y. Thaker et al., Signal transmission through the CXC chemokine receptor 4 (CXCR4) transmembrane helices, Proc. Natl. Acad. Sci. U. S. A, vol.113, pp.9928-9933, 2016.

N. Wettschureck and S. Offermanns, Mammalian G proteins and their cell type specific functions, Physiol. Rev, vol.85, pp.1159-1204, 2005.

C. D. White, M. Coetsee, K. Morgan, C. A. Flanagan, R. P. Millar et al., A crucial role for Galphaq/11, but not Galphai/o or Galphas, in gonadotropin-releasing hormone receptor-mediated cell growth inhibition, Mol. Endocrinol. Baltim. Md, vol.22, pp.2520-2530, 2008.

R. S. Williams and R. J. Lefkowitz, Alpha-adrenergic receptors in rat myocardium. Identification by binding of [3H]dihydroergocryptine, Circ. Res, vol.43, pp.721-727, 1978.

C. Wittpoth, K. Scholich, Y. Yigzaw, T. M. Stringfield, and T. B. Patel, Regions on adenylyl cyclase that are necessary for inhibition of activity by beta gamma and G(ialpha) subunits of heterotrimeric G proteins, Proc. Natl. Acad. Sci. U. S. A, vol.96, pp.9551-9556, 1999.

B. L. Wolfe and J. Trejo, Clathrin-Dependent Mechanisms of G Protein-coupled Receptor Endocytosis, Traffic, vol.8, pp.462-470, 2007.

B. Wu, E. Y. Chien, C. D. Mol, G. Fenalti, W. Liu et al., Structures of the CXCR4 chemokine GPCR with small-molecule and cyclic peptide antagonists, Science, vol.330, pp.1066-1071, 2010.

K. Xiao, S. K. Shenoy, K. Nobles, and R. J. Lefkowitz, Activation-dependent conformational changes in {beta}-arrestin 2, J. Biol. Chem, vol.279, pp.55744-55753, 2004.

Z. Xiao, N. Zhang, D. B. Murphy, and P. N. Devreotes, Dynamic distribution of chemoattractant receptors in living cells during chemotaxis and persistent stimulation, J. Cell Biol, vol.139, pp.365-374, 1997.

H. Xu, J. S. Partilla, X. Wang, J. M. Rutherford, K. Tidgewell et al., A comparison of noninternalizing (herkinorin) and internalizing (DAMGO) µ-opioid agonists on cellular markers related to opioid tolerance and dependence, Synapse, vol.61, pp.166-175, 2007.

S. Xu, H. Jiang, B. Wu, J. Yang, and S. Chen, Urotensin II induces migration of endothelial progenitor cells via activation of the RhoA/Rho kinase pathway, The Tohoku J. Exp. Med, vol.219, pp.283-288, 2009.

S. Xu, H. Wen, and H. Jiang, Urotensin II promotes the proliferation of endothelial progenitor cells through p38 and p44/42 MAPK activation, Mol. Med. Rep, vol.6, pp.197-200, 2012.

X. Xu, W. Zeng, S. Popov, D. M. Berman, I. Davignon et al., RGS Proteins Determine Signaling Specificity of Gq-coupled Receptors, J. Biol. Chem, vol.274, pp.3549-3556, 1999.

H. Yagi, W. Tan, P. Dillenburg-pilla, S. Armando, P. Amornphimoltham et al., A Synthetic Biology Approach Reveals a CXCR4-G13-Rho Signaling Axis Driving Transendothelial Migration of Metastatic Breast Cancer Cells, Sci. Signal, vol.4, p.60, 2011.

J. Yamauchi, M. Nagao, Y. Kaziro, and H. Itoh, Activation of p38 Mitogenactivated Protein Kinase by Signaling through G Protein-coupled Receptors INVOLVEMENT OF G?? AND G?q/11 SUBUNITS, J. Biol. Chem, vol.272, pp.27771-27777, 1997.

K. Yi, M. Yu, L. Wu, and X. Tan, Effects of urotensin II on functional activity of late endothelial progenitor cells, Peptides, vol.33, pp.87-91, 2012.

S. Yohannan, S. Faham, D. Yang, J. P. Whitelegge, and J. U. Bowie, The evolution of transmembrane helix kinks and the structural diversity of G protein-coupled receptors, Proc. Natl. Acad. Sci. U.S.A, vol.101, pp.959-963, 2004.

W. Yuan, Y. Guo, X. Li, M. Deng, Z. Shen et al., MicroRNA-126 inhibits colon cancer cell proliferation and invasion by targeting the chemokine (C-X-C motif) receptor 4 and Ras homolog gene family, 2016.

C. R. Yulis and K. Lederis, Extraurophyseal distribution of urotensin II immunoreactive neuronal perikarya and their processes, Proc. Natl. Acad. Sci. U.S.A, vol.83, pp.7079-7083, 1986.

M. Zajac, J. Law, D. D. Cvetkovic, M. Pampillo, L. Mccoll et al., GPR54 (KISS1R) Transactivates EGFR to Promote Breast Cancer Cell Invasiveness, PLoS ONE, vol.6, p.21599, 2011.

Z. P. Zeng, G. Q. Liu, H. Z. Li, X. R. Fan, D. M. Liu et al., The effects of urotensin-II on proliferation of pheochromocytoma cells and mRNA expression of urotensin-II and its receptor in pheochromocytoma tissues, Proc. Natl. Acad. Sci. U.S.A, vol.1073, pp.284-289, 2006.

F. L. Zhang and P. J. Casey, Protein prenylation: molecular mechanisms and functional consequences, Annu. Rev. Biochem, vol.65, pp.241-269, 1996.

L. Zhang, X. Wang, H. Cao, Y. Chen, X. Chen et al., Vasopressin V1A receptor mediates cell proliferation through GRK2-EGFR-ERK1/2 pathway in A7r5 cells, Eur. J. Pharmacol, vol.792, pp.15-25, 2016.

Y. Zhang, J. Ying, D. Jiang, Z. Chang, H. Li et al., Urotensin-II receptor stimulation of cardiac L-type Ca2+ channels requires the ?? subunits of Gi/o-protein and phosphatidylinositol 3-kinase-dependent protein kinase C ?1 isoform, J. Biol. Chem, vol.290, pp.8644-8655, 2015.

Y. G. Zhang, J. Li, Y. G. Li, and R. H. Wei, Urotensin II induces phenotypic differentiation, migration, and collagen synthesis of adventitial fibroblasts from rat aorta, J. Hypertens, vol.26, pp.1119-1126, 2008.

W. X. Zhang, Y. F. Liang, X. M. Wang, Y. Nie, L. Chong et al., Urotensin upregulates transforming growth factor-beta1 expression of asthma airway through ERK-dependent pathway, vol.364, pp.291-298, 2012.

H. Zheng, E. A. Pearsall, D. P. Hurst, Y. Zhang, J. Chu et al., Palmitoylation and membrane cholesterol stabilize µ-opioid receptor homodimerization and G protein coupling, BMC Cell Biol, vol.13, p.6, 2012.

H. Zhou and H. H. Tai, Expression and functional characterization of mutant human CXCR4 in insect cells: role of cysteinyl and negatively charged residues in ligand binding, Arch. Biochem. Biophys, vol.373, pp.211-217, 2000.

H. Zhou, J. Yang, T. Xin, T. Zhang, S. Hu et al., Exendin-4 enhances the migration of adipose-derived stem cells to neonatal rat ventricular cardiomyocyte-derived conditioned medium via the phosphoinositide 3-kinase/Akt-stromal cell-derived factor-1?/CXC chemokine receptor 4 pathway, Mol. Med. Rep, vol.11, pp.4063-4072, 2015.

Y. Zhou, J. Sondek, and T. K. Harden, Activation of human phospholipase C-eta2 by Gbetagamma, Biochemistry (Mosc.), vol.47, pp.4410-4417, 2008.

J. J. Ziarek, A. E. Getschman, S. J. Butler, D. Taleski, B. Stephens et al., Sulfopeptide probes of the CXCR4/CXCL12 interface reveal oligomer-specific contacts and chemokine allostery, ACS Chem. Biol, vol.8, pp.1955-1963, 2013.

E. Ziegler, T. Olbrich, G. Emons, and C. Gründker, Antiproliferative effects of kisspeptin-10 depend on artificial GPR54 (KISS1R) expression levels, Oncol. Rep, vol.29, pp.549-554, 2013.

P. Ziltener, C. Mueller, B. Haenig, M. W. Scherz, and O. Nayler, Urotensin Ii Mediates Erk1/2 Phosphorylation and Proliferation in Gpr14-Transfected Cell Lines, J. Recept. Signal Transduct, vol.22, pp.155-168, 2002.

B. Zimmerman, A. Beautrait, B. Aguila, R. Charles, E. Escher et al., Differential ?-arrestin-dependent conformational signaling and cellular responses revealed by angiotensin analogs, Sci. Signal, vol.5, p.33, 2012.

Y. Zou, A. H. Kottmann, M. Kuroda, I. Taniuchi, and D. R. Littman, Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development, Nature, vol.393, pp.595-599, 1998.