Browse > Article
http://dx.doi.org/10.4062/biomolther.2013.080

Intercellular Lipid Mediators and GPCR Drug Discovery  

Im, Dong-Soon (Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of Pharmacy, Pusan National University)
Publication Information
Biomolecules & Therapeutics / v.21, no.6, 2013 , pp. 411-422 More about this Journal
Abstract
G-protein-coupled receptors (GPCR) are the largest superfamily of receptors responsible for signaling between cells and tissues, and because they play important physiological roles in homeostasis, they are major drug targets. New technologies have been developed for the identification of new ligands, new GPCR functions, and for drug discovery purposes. In particular, intercellular lipid mediators, such as, lysophosphatidic acid and sphingosine 1-phosphate have attracted much attention for drug discovery and this has resulted in the development of fingolimod (FTY-720) and AM095. The discovery of new intercellular lipid mediators and their GPCRs are discussed from the perspective of drug development. Lipid GPCRs for lysophospholipids, including lysophosphatidylserine, lysophosphatidylinositol, lysophosphatidylcholine, free fatty acids, fatty acid derivatives, and other lipid mediators are reviewed.
Keywords
Lipid mediator; GPCR; Lipid; Lysophospholipid; Fatty acid; Drug discovery;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Jenkins, L., Alvarez-Curto, E., Campbell, K., de Munnik, S., Canals, M., Schlyer, S. and Milligan, G. (2011) Agonist activation of the G protein-coupled receptor GPR35 involves transmembrane domain III and is transduced via Galpha(1)(3) and beta-arrestin-2. Br. J. Pharmacol. 162, 733-748.   DOI   ScienceOn
2 Johns, D. G., Behm, D. J., Walker, D. J., Ao, Z., Shapland, E. M., Daniels, D. A., Riddick, M., Dowell, S., Staton, P. C., Green, P., Shabon, U., Bao, W., Aiyar, N., Yue, T. L., Brown, A. J., Morrison, A. D. and Douglas, S. A. (2007) The novel endocannabinoid receptor GPR55 is activated by atypical cannabinoids but does not mediate their vasodilator effects. Br. J. Pharmacol. 152, 825-831.
3 Johnson, L. E., Elias, M. S., Bolick, D. T., Skafl en, M. D., Green, R. M. and Hedrick, C. C. (2008) The G protein-coupled receptor G2A: involvement in hepatic lipid metabolism and gallstone formation in mice. Hepatology 48, 1138-1148.   DOI   ScienceOn
4 Jones, C. E., Holden, S., Tenaillon, L., Bhatia, U., Seuwen, K., Tranter, P., Turner, J., Kettle, R., Bouhelal, R., Charlton, S., Nirmala, N. R., Jarai, G. and Finan, P. (2003) Expression and characterization of a 5-oxo-6E,8Z,11Z,14Z-eicosatetraenoic acid receptor highly expressed on human eosinophils and neutrophils. Mol. Pharmacol. 63, 471-477.   DOI   ScienceOn
5 Kabarowski, J. H. (2009) G2A and LPC: regulatory functions in immunity. Prostaglandins Other Lipid Mediat. 89, 73-81.   DOI   ScienceOn
6 Kaur, B., Cork, S. M., Sandberg, E. M., Devi, N. S., Zhang, Z., Klenotic, P. A., Febbraio, M., Shim, H., Mao, H., Tucker-Burden, C., Silverstein, R. L., Brat, D. J., Olson, J. J. and Van Meir, E. G. (2009) Vasculostatin inhibits intracranial glioma growth and negatively regulates in vivo angiogenesis through a CD36-dependent mechanism. Cancer Res. 69, 1212-1220.   DOI   ScienceOn
7 Kawamata, Y., Fujii, R., Hosoya, M., Harada, M., Yoshida, H., Miwa, M., Fukusumi, S., Habata, Y., Itoh, T., Shintani, Y., Hinuma, S., Fujisawa, Y. and Fujino, M. (2003) A G protein-coupled receptor responsive to bile acids. J. Biol. Chem. 278, 9435-9440.   DOI   ScienceOn
8 Uhlenbrock, K., Gassenhuber, H. and Kostenis, E. (2002) Sphingosine 1-phosphate is a ligand of the human gpr3, gpr6 and gpr12 family of constitutively active G protein-coupled receptors. Cell. Signal. 14, 941-953.   DOI   ScienceOn
9 Valverde, O., Celerier, E., Baranyi, M., Vanderhaeghen, P., Maldonado, R., Sperlagh, B., Vassart, G. and Ledent, C. (2009) GPR3 receptor, a novel actor in the emotional-like responses. PloS one 4, e4704.   DOI   ScienceOn
10 Vassilatis, D. K., Hohmann, J. G., Zeng, H., Li, F., Ranchalis, J. E., Mortrud, M. T., Brown, A., Rodriguez, S. S., Weller, J. R., Wright, A. C., Bergmann, J. E. and Gaitanaris, G. A. (2003) The G proteincoupled receptor repertoires of human and mouse. Proc. Natl. Acad Sci. U.S.A. 100, 4903-4908.   DOI   ScienceOn
11 Vassileva, G., Golovko, A., Markowitz, L., Abbondanzo, S. J., Zeng, M., Yang, S., Hoos, L., Tetzloff, G., Levitan, D., Murgolo, N. J., Keane, K., Davis, H. R., Jr., Hedrick, J. and Gustafson, E. L. (2006) Targeted deletion of Gpbar1 protects mice from cholesterol gallstone formation. Biochem. J. 398, 423-430.   DOI   ScienceOn
12 Venkataraman, C. and Kuo, F. (2005) The G-protein coupled receptor, GPR84 regulates IL-4 production by T lymphocytes in response to CD3 crosslinking. Immunol. Lett. 101, 144-153.   DOI   ScienceOn
13 Wang, J., Simonavicius, N., Wu, X., Swaminath, G., Reagan, J., Tian, H. and Ling, L. (2006a) Kynurenic acid as a ligand for orphan G protein-coupled receptor GPR35. J. Biol. Chem. 281, 22021-22028.   DOI   ScienceOn
14 Wang, J., Wu, X., Simonavicius, N., Tian, H. and Ling, L. (2006b) Medium-chain fatty acids as ligands for orphan G protein-coupled receptor GPR84. J. Biol. Chem. 281, 34457-34464.   DOI   ScienceOn
15 Wang, L., Radu, C. G., Yang, L. V., Bentolila, L. A., Riedinger, M. and Witte, O. N. (2005) Lysophosphatidylcholine-induced surface redistribution regulates signaling of the murine G protein-coupled receptor G2A. Mol. Biol. Cell 16, 2234-2247.   DOI   ScienceOn
16 Kostenis, E. (2004) A glance at G-protein-coupled receptors for lipid mediators: a growing receptor family with remarkably diverse ligands. Pharmacol. Ther. 102, 243-257.   DOI   ScienceOn
17 Kenakin, T. P. (2001) Quantitation in receptor pharmacology. Receptors Channels 7, 371-385.
18 Kitamura, H., Makide, K., Shuto, A., Ikubo, M., Inoue, A., Suzuki, K., Sato, Y., Nakamura, S., Otani, Y., Ohwada, T. and Aoki, J. (2012) GPR34 is a receptor for lysophosphatidylserine with a fatty acid at the sn-2 position. J. Biochem. 151, 511-518.   DOI   ScienceOn
19 Kohno, M., Hasegawa, H., Inoue, A., Muraoka, M., Miyazaki, T., Oka, K. and Yasukawa, M. (2006) Identification of N-arachidonylglycine as the endogenous ligand for orphan G-protein-coupled receptor GPR18. Biochem. Biophys. Res. Commun. 347, 827-832.   DOI   ScienceOn
20 Lauckner, J. E., Jensen, J. B., Chen, H. Y., Lu, H. C., Hille, B. and Mackie, K. (2008) GPR55 is a cannabinoid receptor that increases intracellular calcium and inhibits M current. Proc. Natl. Acad Sci. U. S.A. 105, 2699-2704.   DOI   ScienceOn
21 Lerner, M. R. (1994) Tools for investigating functional interactions between ligands and G-protein-coupled receptors. Trends Neurosci. 17, 142-146.   DOI   ScienceOn
22 Liebscher, I., Muller, U., Teupser, D., Engemaier, E., Engel, K. M., Ritscher, L., Thor, D., Sangkuhl, K., Ricken, A., Wurm, A., Piehler, D., Schmutzler, S., Fuhrmann, H., Albert, F. W., Reichenbach, A., Thiery, J., Schoneberg, T. and Schulz, A. (2011) Altered immune response in mice deficient for the G protein-coupled receptor GPR34. J. Biol. Chem. 286, 2101-2110.   DOI   ScienceOn
23 Liu, C., Yang, X. V., Wu, J., Kuei, C., Mani, N. S., Zhang, L., Yu, J., Sutton, S. W., Qin, N., Banie, H., Karlsson, L., Sun, S. and Lovenberg, T. W. (2011) Oxysterols direct B-cell migration through EBI2. Nature 475, 519-523.   DOI   ScienceOn
24 Yanagida, K., Kurikawa, Y., Shimizu, T. and Ishii, S. (2013) Current progress in non-Edg family LPA receptor research. Biochim. Biophys. Acta 1831, 33-41.   DOI   ScienceOn
25 Watanabe, M., Houten, S. M., Mataki, C., Christoffolete, M. A., Kim, B. W., Sato, H., Messaddeq, N., Harney, J. W., Ezaki, O., Kodama, T., Schoonjans, K., Bianco, A. C. and Auwerx, J. (2006) Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 439, 484-489.   DOI   ScienceOn
26 Yamashita, A., Oka, S., Tanikawa, T., Hayashi, Y., Nemoto-Sasaki, Y. and Sugiura, T. (2013) The actions and metabolism of lysophosphatidylinositol, an endogenous agonist for GPR55. Prostaglandins Other Lipid Mediat. [Epub ahead of print]
27 Yan, J. J., Jung, J. S., Lee, J. E., Lee, J., Huh, S. O., Kim, H. S., Jung, K. C., Cho, J. Y., Nam, J. S., Suh, H. W., Kim, Y. H. and Song, D. K. (2004) Therapeutic effects of lysophosphatidylcholine in experimental sepsis. Nat. Med. 10, 161-167.   DOI   ScienceOn
28 Yang, C. R., Wei, Y., Qi, S. T., Chen, L., Zhang, Q. H., Ma, J. Y., Luo, Y. B., Wang, Y. P., Hou, Y., Schatten, H., Liu, Z. H. and Sun, Q. Y. (2012) The G protein coupled receptor 3 is involved in cAMP and cGMP signaling and maintenance of meiotic arrest in porcine oocytes. PloS one 7, e38807.   DOI
29 Yin, H., Chu, A., Li, W., Wang, B., Shelton, F., Otero, F., Nguyen, D. G., Caldwell, J. S. and Chen, Y. A. (2009) Lipid G protein-coupled receptor ligand identifi cation using beta-arrestin PathHunter assay. J. Biol. Chem. 284, 12328-12338.   DOI   ScienceOn
30 Yoshida, M., Miyazato, M. and Kangawa, K. (2012) Orphan GPCRs and methods for identifying their ligands. Methods Enzymol. 514, 33-44.   DOI   ScienceOn
31 Zhang, B. L., Li, Y., Ding, J. H., Dong, F. L., Hou, Y. J., Jiang, B. C., Shi, F. X. and Xu, Y. X. (2012) Sphingosine 1-phosphate acts as an activator for the porcine Gpr3 of constitutively active G proteincoupled receptors. Journal of Zhejiang University. J. Zhejang Univ. Sci. B 13, 555-566.   DOI
32 Bratton, D. L. and Henson, P. M. (2008) Apoptotic cell recognition: will the real phosphatidylserine receptor(s) please stand up? Curr. Biol. 18, R76-79.   DOI   ScienceOn
33 Arterburn, J. B., Oprea, T. I., Prossnitz, E. R., Edwards, B. S. and Sklar, L. A. (2009) Discovery of selective probes and antagonists for G-protein-coupled receptors FPR/FPRL1 and GPR30. Curr. Top. Med. Chem. 9, 1227-1236.   DOI   ScienceOn
34 Baker, D., Pryce, G., Davies, W. L. and Hiley, C. R. (2006) In silico patent searching reveals a new cannabinoid receptor. Trends Pharmacol. Sci. 27, 1-4.   DOI   ScienceOn
35 Benned-Jensen, T. and Rosenkilde, M. M. (2010) Distinct expression and ligand-binding profi les of two constitutively active GPR17 splice variants. Br. J. Pharmacol. 159, 1092-1105.   DOI   ScienceOn
36 Bresnick, J. N., Skynner, H. A., Chapman, K. L., Jack, A. D., Zamiara, E., Negulescu, P., Beaumont, K., Patel, S. and McAllister, G. (2003) Identifi cation of signal transduction pathways used by orphan g protein-coupled receptors. Assay Drug Dev. Technol. 1, 239-249.   DOI   ScienceOn
37 Maruyama, T., Tanaka, K., Suzuki, J., Miyoshi, H., Harada, N., Nakamura, T., Miyamoto, Y., Kanatani, A. and Tamai, Y. (2006) Targeted disruption of G protein-coupled bile acid receptor 1 (Gpbar1/M-Bar) in mice. J. Endocrinol. 191, 197-205.   DOI   ScienceOn
38 Lu, V. B., Puhl, H. L., 3rd and Ikeda, S. R. (2013) N-Arachidonyl glycine does not activate G protein-coupled receptor 18 signaling via canonical pathways. Mol. Pharmacol. 83, 267-282.   DOI
39 Maekawa, A., Balestrieri, B., Austen, K. F. and Kanaoka, Y. (2009) GPR17 is a negative regulator of the cysteinyl leukotriene 1 receptor response to leukotriene D4. Proc. Natl. Acad Sci. U.S.A. 106, 11685-11690.   DOI   ScienceOn
40 Maruyama, T., Miyamoto, Y., Nakamura, T., Tamai, Y., Okada, H., Sugiyama, E., Itadani, H. and Tanaka, K. (2002) Identification of membrane-type receptor for bile acids (M-BAR) Biochem. Biophys. Res. Commun. 298, 714-719.   DOI   ScienceOn
41 McHugh, D., Hu, S. S., Rimmerman, N., Juknat, A., Vogel, Z., Walker, J. M. and Bradshaw, H. B. (2010) N-arachidonoyl glycine, an abundant endogenous lipid, potently drives directed cellular migration through GPR18, the putative abnormal cannabidiol receptor. BMC Neurosci. 11, 44.   DOI   ScienceOn
42 McHugh, D., Page, J., Dunn, E. and Bradshaw, H. B. (2012) Delta(9) -Tetrahydrocannabinol and N-arachidonyl glycine are full agonists at GPR18 receptors and induce migration in human endometrial HEC-1B cells. Br. J. Pharmacol. 165, 2414-2424.   DOI   ScienceOn
43 Oka, S., Toshida, T., Maruyama, K., Nakajima, K., Yamashita, A. and Sugiura, T. (2009) 2-Arachidonoyl-sn-glycero-3-phosphoinositol: a possible natural ligand for GPR55. J. Biochem. 145, 13-20.
44 Castelino, F. V., Seiders, J., Bain, G., Brooks, S. F., King, C. D., Swaney, J. S., Lorrain, D. S., Chun, J., Luster, A. D. and Tager, A. M. (2011) Amelioration of dermal fibrosis by genetic deletion or pharmacologic antagonism of lysophosphatidic acid receptor 1 in a mouse model of scleroderma. Arthritis Rheum. 63, 1405-1415.   DOI   ScienceOn
45 Zhang, R. and Xie, X. (2012) Tools for GPCR drug discovery. Acta Pharmacol. Sin. 33, 372-384.   DOI
46 Zhao, P. and Abood, M. E. (2013) GPR55 and GPR35 and their relationship to cannabinoid and lysophospholipid receptors. Life Sci. 92, 453-457.   DOI   ScienceOn
47 Zhu, T., Gobeil, F., Vazquez-Tello, A., Leduc, M., Rihakova, L., Bossolasco, M., Bkaily, G., Peri, K., Varma, D. R., Orvoine, R. and Chemtob, S. (2006) Intracrine signaling through lipid mediators and their cognate nuclear G-protein-coupled receptors: a paradigm based on PGE2, PAF, and LPA1 receptors. Can. J. Physiol. Pharmacol. 84, 377-391.   DOI   ScienceOn
48 Brink, C., Dahlen, S. E., Drazen, J., Evans, J. F., Hay, D. W., Nicosia, S., Serhan, C. N., Shimizu, T. and Yokomizo, T. (2003) International Union of Pharmacology XXXVII. Nomenclature for leukotriene and lipoxin receptors. Pharmacol. Rev. 55, 195-227.   DOI   ScienceOn
49 Brink, C., Dahlen, S. E., Drazen, J., Evans, J. F., Hay, D. W., Rovati, G. E., Serhan, C. N., Shimizu, T. and Yokomizo, T. (2004) International Union of Pharmacology XLIV. Nomenclature for the oxoeicosanoid receptor. Pharmacol. Rev. 56, 149-157.   DOI   ScienceOn
50 Choi, J. W. and Chun, J. (2013) Lysophospholipids and their receptors in the central nervous system. Biochim. Biophys. Acta 1831, 20-32.   DOI   ScienceOn
51 Chu, Z. L., Carroll, C., Chen, R., Alfonso, J., Gutierrez, V., He, H., Lucman, A., Xing, C., Sebring, K., Zhou, J., Wagner, B., Unett, D., Jones, R. M., Behan, D. P. and Leonard, J. (2010) N-oleoyldopamine enhances glucose homeostasis through the activation of GPR119. Mol. Endocrinol. 24, 161-170.   DOI   ScienceOn
52 Chun, J., Hla, T., Lynch, K. R., Spiegel, S. and Moolenaar, W. H. (2010) International Union of Basic and Clinical Pharmacology. LXXVIII. Lysophospholipid receptor nomenclature. Pharmacol. Rev. 62, 579-587.   DOI   ScienceOn
53 Ciana, P., Fumagalli, M., Trincavelli, M. L., Verderio, C., Rosa, P., Lecca, D., Ferrario, S., Parravicini, C., Capra, V., Gelosa, P., Guerrini, U., Belcredito, S., Cimino, M., Sironi, L., Tremoli, E., Rovati, G. E., Martini, C. and Abbracchio, M. P. (2006) The orphan receptor GPR17 identifi ed as a new dual uracil nucleotides/cysteinyl-leukotrienes receptor. EMBO J. 25, 4615-4627.   DOI   ScienceOn
54 Neetoo-Isseljee, Z., MacKenzie, A. E., Southern, C., Jerman, J., Mc-Iver, E. G., Harries, N., Taylor, D. L. and Milligan, G. (2013) Highthroughput identifi cation and characterization of novel, speciesselective GPR35 agonists. J. Pharmacol. Exp. Ther. 344, 568-578.   DOI
55 McIntyre, T. M., Pontsler, A. V., Silva, A. R., St Hilaire, A., Xu, Y., Hinshaw, J. C., Zimmerman, G. A., Hama, K., Aoki, J., Arai, H. and Prestwich, G. D. (2003) Identification of an intracellular receptor for lysophosphatidic acid (LPA): LPA is a transcellular PPARgamma agonist. Proc. Natl. Acad Sci. U.S.A. 100, 131-136.   DOI   ScienceOn
56 Meyer zu Heringdorf, D. and Jakobs, K. H. (2007) Lysophospholipid receptors: signalling, pharmacology and regulation by lysophospholipid metabolism. Biochim. Biophys. Acta 1768, 923-940.   DOI   ScienceOn
57 Murakami, M., Shiraishi, A., Tabata, K. and Fujita, N. (2008) Identifi cation of the orphan GPCR, P2Y(10) receptor as the sphingosine-1-phosphate and lysophosphatidic acid receptor. Biochem. Biophys. Res. Commun. 371, 707-712.   DOI   ScienceOn
58 Niedernberg, A., Tunaru, S., Blaukat, A., Ardati, A. and Kostenis, E. (2003) Sphingosine 1-phosphate and dioleoylphosphatidic acid are low affinity agonists for the orphan receptor GPR63. Cell. Signal. 15, 435-446.   DOI   ScienceOn
59 Obinata, H., Hattori, T., Nakane, S., Tatei, K. and Izumi, T. (2005) Identification of 9-hydroxyoctadecadienoic acid and other oxidized free fatty acids as ligands of the G protein-coupled receptor G2A. J. Biol. Chem. 280, 40676-40683.   DOI   ScienceOn
60 Obinata, H. and Hla, T. (2012) Fine-tuning S1P therapeutics. Chem. Biol. 19, 1080-1082.   DOI   ScienceOn
61 Obinata, H. and Izumi, T. (2009) G2A as a receptor for oxidized free fatty acids. Prostaglandins Other Lipid Mediat. 89, 66-72.   DOI   ScienceOn
62 Das, S., Owen, K. A., Ly, K. T., Park, D., Black, S. G., Wilson, J. M., Sifri, C. D., Ravichandran, K. S., Ernst, P. B. and Casanova, J. E. (2011) Brain angiogenesis inhibitor 1 (BAI1) is a pattern recognition receptor that mediates macrophage binding and engulfment of Gram-negative bacteria. Proc. Natl. Acad Sci. U.S.A. 108, 2136-2141.   DOI   ScienceOn
63 Coppi, E., Maraula, G., Fumagalli, M., Failli, P., Cellai, L., Bonfanti, E., Mazzoni, L., Coppini, R., Abbracchio, M. P., Pedata, F. and Pugliese, A. M. (2013) UDP-glucose enhances outward K(+) currents necessary for cell differentiation and stimulates cell migration by activating the GPR17 receptor in oligodendrocyte precursors. Glia 61, 1155-1171.   DOI   ScienceOn
64 Cork, S. M. and Van Meir, E. G. (2011) Emerging roles for the BAI1 protein family in the regulation of phagocytosis, synaptogenesis, neurovasculature, and tumor development. J. Mol. Med. (Berl) 89, 743-752.   DOI   ScienceOn
65 Cyster, J. G. and Schwab, S. R. (2012) Sphingosine-1-phosphate and lymphocyte egress from lymphoid organs. Annu. Rev. Immunol. 30, 69-94.   DOI   ScienceOn
66 Davenport, A. P., Alexander, S. P., Sharman, J. L., Pawson, A. J., Benson, H. E., Monaghan, A. E., Liew, W. C., Mpamhanga, C. P., Bonner, T. I., Neubig, R. R., Pin, J. P., Spedding, M. and Harmar, A. J. (2013) International Union of Basic and Clinical Pharmacology. LXXXVIII. G protein-coupled receptor list: recommendations for new pairings with cognate ligands. Pharmacol. Rev. 65, 967-986.   DOI   ScienceOn
67 DiLuigi, A., Weitzman, V. N., Pace, M. C., Siano, L. J., Maier, D. and Mehlmann, L. M. (2008a) Meiotic arrest in human oocytes is maintained by a Gs signaling pathway. Biol. Reprod. 78, 667-672.   DOI   ScienceOn
68 DiLuigi, A. J., Maier, D. B. and Benadiva, C. A. (2008b) Ruptured ectopic pregnancy with contralateral adnexal torsion after spontaneous conception. Fertil. Steril. 90, e1-3.   DOI   ScienceOn
69 Ohishi, T. and Yoshida, S. (2012) The therapeutic potential of GPR119 agonists for type 2 diabetes. Expert Opin. Investig. Drugs 21, 321-328.   DOI   ScienceOn
70 Oh, D., Y., Yoon, J. M., Moon, M. J., Hwang, J. I., Choe, H., Lee, J. Y., Kim, J. I., Kim, S., Rhim, H., O'Dell, D. K., Walker, J. M., Na, H. S., Lee, M. G., Kwon, H. B., Kim, K. and Seong, J. Y. (2008) Identification of farnesyl pyrophosphate and N-arachidonylglycine as endogenous ligands for GPR92. J. Biol. Chem. 283, 21054-21064.   DOI   ScienceOn
71 Oka, S., Nakajima, K., Yamashita, A., Kishimoto, S. and Sugiura, T. (2007) Identification of GPR55 as a lysophosphatidylinositol receptor. Biochem. Biophys. Res. Commun. 362, 928-934.   DOI   ScienceOn
72 Oka, S., Ota, R., Shima, M., Yamashita, A. and Sugiura, T. (2010) GPR35 is a novel lysophosphatidic acid receptor. Biochem. Biophys. Res. Commun. 395, 232-237.   DOI   ScienceOn
73 Okajima, F. (2013) Regulation of inflammation by extracellular acidification and proton-sensing GPCRs. Cell. Signal. 25, 2263-2271.   DOI   ScienceOn
74 Overton, H. A., Babbs, A. J., Doel, S. M., Fyfe, M. C., Gardner, L. S., Griffi n, G., Jackson, H. C., Procter, M. J., Rasamison, C. M., Tang-Christensen, M., Widdowson, P. S., Williams, G. M. and Reynet, C. (2006) Deorphanization of a G protein-coupled receptor for oleoylethanolamide and its use in the discovery of small-molecule hypophagic agents. Cell Metab. 3, 167-175.   DOI   ScienceOn
75 Overton, H. A., Fyfe, M. C. and Reynet, C. (2008) GPR119, a novel G protein-coupled receptor target for the treatment of type 2 diabetes and obesity. Br. J. Pharmacol. 153 Suppl 1, S76-81.
76 Padmanabhan, S., Myers, A. G. and Prasad, B. M. (2009) Constitutively active GPR6 is located in the intracellular compartments. FEBS Lett. 583, 107-112.   DOI   ScienceOn
77 Fiorucci, S., Mencarelli, A., Palladino, G. and Cipriani, S. (2009) Bileacid-activated receptors: targeting TGR5 and farnesoid-X-receptor in lipid and glucose disorders. Trends Pharmacol. Sci. 30, 570-580.   DOI   ScienceOn
78 Dixon, R. A., Kobilka, B. K., Strader, D. J., Benovic, J. L., Dohlman, H. G., Frielle, T., Bolanowski, M. A., Bennett, C. D., Rands, E., Diehl, R. E. and et al. (1986) Cloning of the gene and cDNA for mammalian beta-adrenergic receptor and homology with rhodopsin. Nature 321, 75-79.   DOI   ScienceOn
79 Duman, J. G., Tzeng, C. P., Tu, Y. K., Munjal, T., Schwechter, B., Ho, T. S. and Tolias, K. F. (2013) The adhesion-GPCR BAI1 regulates synaptogenesis by controlling the recruitment of the Par3/Tiam1 polarity complex to synaptic sites. J. Neurosci. 33, 6964-6978.   DOI   ScienceOn
80 Eggerickx, D., Denef, J. F., Labbe, O., Hayashi, Y., Refetoff, S., Vassart, G., Parmentier, M. and Libert, F. (1995) Molecular cloning of an orphan G-protein-coupled receptor that constitutively activates adenylate cyclase. Biochem. J. 309 (Pt 3), 837-843.   DOI
81 Franke, H., Parravicini, C., Lecca, D., Zanier, E. R., Heine, C., Bremicker, K., Fumagalli, M., Rosa, P., Longhi, L., Stocchetti, N., De Simoni, M. G., Weber, M. and Abbracchio, M. P. (2013) Changes of the GPR17 receptor, a new target for neurorepair, in neurons and glial cells in patients with traumatic brain injury. Purinergic Signal. 9, 451-462.   DOI   ScienceOn
82 Frasch, S. C. and Bratton, D. L. (2012) Emerging roles for lysophosphatidylserine in resolution of infl ammation. Prog. Lipid Res. 51, 199-207.   DOI   ScienceOn
83 Frasch, S. C., Fernandez-Boyanapalli, R. F., Berry, K. A., Murphy, R. C., Leslie, C. C., Nick, J. A., Henson, P. M. and Bratton, D. L. (2013) Neutrophils regulate tissue Neutrophilia in inflammation via the oxidant-modified lipid lysophosphatidylserine. J. Biol. Chem. 288, 4583-4593.   DOI
84 Pineiro, R. and Falasca, M. (2012) Lysophosphatidylinositol signalling: new wine from an old bottle. Biochim. Biophys. Acta 1821, 694-705.   DOI   ScienceOn
85 Park, D., Tosello-Trampont, A. C., Elliott, M. R., Lu, M., Haney, L. B., Ma, Z., Klibanov, A. L., Mandell, J. W. and Ravichandran, K. S. (2007) BAI1 is an engulfment receptor for apoptotic cells upstream of the ELMO/Dock180/Rac module. Nature 450, 430-434.   DOI   ScienceOn
86 Parks, B. W., Gambill, G. P., Lusis, A. J. and Kabarowski, J. H. (2005) Loss of G2A promotes macrophage accumulation in atherosclerotic lesions of low density lipoprotein receptor-deficient mice. J. Lipid Res. 46, 1405-1415.   DOI   ScienceOn
87 Pertwee, R. G., Howlett, A. C., Abood, M. E., Alexander, S. P., Di Marzo, V., Elphick, M. R., Greasley, P. J., Hansen, H. S., Kunos, G., Mackie, K., Mechoulam, R. and Ross, R. A. (2010) International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid receptors and their ligands: beyond CB(1) and CB(2). Pharmacol. Rev. 62, 588-631.   DOI   ScienceOn
88 Powell, W. S. and Rokach, J. (2013) The eosinophil chemoattractant 5-oxo-ETE and the OXE receptor. Prog. Lipid Res. 52, 651-665.   DOI   ScienceOn
89 Prossnitz, E. R. and Barton, M. (2009) Signaling, physiological functions and clinical relevance of the G protein-coupled estrogen receptor GPER. Prostaglandins Other Lipid Mediat. 89, 89-97.   DOI   ScienceOn
90 Prossnitz, E. R. and Barton, M. (2011) The G-protein-coupled estrogen receptor GPER in health and disease. Nat. Rev. Endocrinol. 7, 715-726.   DOI   ScienceOn
91 Qi, A. D., Harden, T. K. and Nicholas, R. A. (2013) Is GPR17 a P2Y/ leukotriene receptor? Examination of uracil nucleotides, nucleotide sugars, and cysteinyl leukotrienes as agonists of GPR17. J. Pharmacol. Exp. Ther. 347, 38-46.   DOI
92 Goetzl, E. J. (2007) Diverse pathways for nuclear signaling by G protein-coupled receptors and their ligands. FASEB J. 21, 638-642.   DOI   ScienceOn
93 Frasch, S. C., Zemski-Berry, K., Murphy, R. C., Borregaard, N., Henson, P. M. and Bratton, D. L. (2007) Lysophospholipids of different classes mobilize neutrophil secretory vesicles and induce redundant signaling through G2A. J. Immunol. 178, 6540-6548.   DOI
94 Fredriksson, R., Lagerstrom, M. C., Lundin, L. G. and Schioth, H. B. (2003) The G-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol. Pharmacol. 63, 1256-1272.   DOI   ScienceOn
95 Freudzon, L., Norris, R. P., Hand, A. R., Tanaka, S., Saeki, Y., Jones, T. L., Rasenick, M. M., Berlot, C. H., Mehlmann, L. M. and Jaffe, L. A. (2005) Regulation of meiotic prophase arrest in mouse oocytes by GPR3, a constitutive activator of the Gs G protein. J. Cell Biol. 171, 255-265.   DOI   ScienceOn
96 Gore, V., Patel, P., Chang, C. T., Sivendran, S., Kang, N., Ouedraogo, Y. P., Gravel, S., Powell, W. S. and Rokach, J. (2013) 5-Oxo-ETE receptor antagonists. J. Med. Chem. 56, 3725-3732.   DOI   ScienceOn
97 Grant, G. E., Rokach, J. and Powell, W. S. (2009) 5-Oxo-ETE and the OXE receptor. Prostaglandins Other Lipid Mediat. 89, 98-104.   DOI   ScienceOn
98 Grzelczyk, A. and Gendaszewska-Darmach, E. (2013) Novel bioactive glycerol-based lysophospholipids: new data--new insight into their function. Biochimie 95, 667-679.   DOI   ScienceOn
99 Guo, Y., Zhang, W., Giroux, C., Cai, Y., Ekambaram, P., Dilly, A. K., Hsu, A., Zhou, S., Maddipati, K. R., Liu, J., Joshi, S., Tucker, S. C., Lee, M. J. and Honn, K. V. (2011) Identifi cation of the orphan G protein-coupled receptor GPR31 as a receptor for 12-(S)-hydroxyeicosatetraenoic acid. J. Biol. Chem. 286, 33832-33840.   DOI   ScienceOn
100 Qin, Y., Verdegaal, E. M., Siderius, M., Bebelman, J. P., Smit, M. J., Leurs, R., Willemze, R., Tensen, C. P. and Osanto, S. (2011) Quantitative expression profiling of G-protein-coupled receptors (GPCRs) in metastatic melanoma: the constitutively active orphan GPCR GPR18 as novel drug target. Pigment Cell Melanoma Res. 24, 207-218.   DOI   ScienceOn
101 Quancard, J., Bollbuck, B., Janser, P., Angst, D., Berst, F., Buehlmayer, P., Streiff, M., Beerli, C., Brinkmann, V., Guerini, D., Smith, P. A., Seabrook, T. J., Traebert, M., Seuwen, K., Hersperger, R., Bruns, C., Bassilana, F. and Bigaud, M. (2012) A potent and selective S1P(1) antagonist with effi cacy in experimental autoimmune encephalomyelitis. Chem. Biol. 19, 1142-1151.   DOI   ScienceOn
102 Radu, C. G., Nijagal, A., McLaughlin, J., Wang, L. and Witte, O. N. (2005) Differential proton sensitivity of related G protein-coupled receptors T cell death-associated gene 8 and G2A expressed in immune cells. Proc. Natl. Acad. Sci. U.S.A. 102, 1632-1637.   DOI   ScienceOn
103 Rajagopal, S., Rajagopal, K. and Lefkowitz, R. J. (2010) Teaching old receptors new tricks: biasing seven-transmembrane receptors. Nat. Rev. Drug Discov. 9, 373-386.   DOI   ScienceOn
104 Ren, H., Orozco, I. J., Su, Y., Suyama, S., Gutierrez-Juarez, R., Horvath, T. L., Wardlaw, S. L., Plum, L., Arancio, O. and Accili, D. (2012) FoxO1 target Gpr17 activates AgRP neurons to regulate food intake. Cell 149, 1314-1326.   DOI   ScienceOn
105 Ritscher, L., Engemaier, E., Staubert, C., Liebscher, I., Schmidt, P., Hermsdorf, T., Rompler, H., Schulz, A. and Schoneberg, T. (2012) The ligand specificity of the G-protein-coupled receptor GPR34. Biochem. J. 443, 841-850.   DOI   ScienceOn
106 Rosenkilde, M. M., Benned-Jensen, T., Andersen, H., Holst, P. J., Kledal, T. N., Luttichau, H. R., Larsen, J. K., Christensen, J. P. and Schwartz, T. W. (2006) Molecular pharmacological phenotyping of EBI2. An orphan seven-transmembrane receptor with constitutive activity. J. Biol. Chem. 281, 13199-13208.   DOI   ScienceOn
107 Hinckley, M., Vaccari, S., Horner, K., Chen, R. and Conti, M. (2005) The G-protein-coupled receptors GPR3 and GPR12 are involved in cAMP signaling and maintenance of meiotic arrest in rodent oocytes. Dev. Biol. 287, 249-261.   DOI   ScienceOn
108 Hannedouche, S., Zhang, J., Yi, T., Shen, W., Nguyen, D., Pereira, J. P., Guerini, D., Baumgarten, B. U., Roggo, S., Wen, B., Knochenmuss, R., Noel, S., Gessier, F., Kelly, L. M., Vanek, M., Laurent, S., Preuss, I., Miault, C., Christen, I., Karuna, R., Li, W., Koo, D. I., Suply, T., Schmedt, C., Peters, E. C., Falchetto, R., Katopodis, A., Spanka, C., Roy, M. O., Detheux, M., Chen, Y. A., Schultz, P. G., Cho, C. Y., Seuwen, K., Cyster, J. G. and Sailer, A. W. (2011) Oxysterols direct immune cell migration via EBI2. Nature 475, 524-527.   DOI   ScienceOn
109 Hansen, K. B., Rosenkilde, M. M., Knop, F. K., Wellner, N., Diep, T. A., Rehfeld, J. F., Andersen, U. B., Holst, J. J. and Hansen, H. S. (2011) 2-Oleoyl glycerol is a GPR119 agonist and signals GLP-1 release in humans. J. Clin. Endocrinol. Metab. 96, E1409-1417.   DOI
110 Hara, T., Kimura, I., Inoue, D., Ichimura, A. and Hirasawa, A. (2013) Free fatty acid receptors and their role in regulation of energy metabolism. Rev. Physiol. Biochem. Pharmacol. 164, 77-116.   DOI   ScienceOn
111 Hochreiter-Hufford, A. E., Lee, C. S., Kinchen, J. M., Sokolowski, J. D., Arandjelovic, S., Call, J. A., Klibanov, A. L., Yan, Z., Mandell, J. W. and Ravichandran, K. S. (2013) Phosphatidylserine receptor BAI1 and apoptotic cells as new promoters of myoblast fusion. Nature 497, 263-267.   DOI   ScienceOn
112 Hosoi, T., Koguchi, Y., Sugikawa, E., Chikada, A., Ogawa, K., Tsuda, N., Suto, N., Tsunoda, S., Taniguchi, T. and Ohnuki, T. (2002) Identification of a novel human eicosanoid receptor coupled to G(i/o) J. Biol. Chem. 277, 31459-31465.   DOI   ScienceOn
113 Howard, A. D., McAllister, G., Feighner, S. D., Liu, Q., Nargund, R. P., Van der Ploeg, L. H. and Patchett, A. A. (2001) Orphan G-proteincoupled receptors and natural ligand discovery. Trends Pharmacol. Sci. 22, 132-140.
114 Serhan, C. N., Hong, S., Gronert, K., Colgan, S. P., Devchand, P. R., Mirick, G. and Moussignac, R. L. (2002) Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J. Exp. Med. 196, 1025-1037.   DOI   ScienceOn
115 Ruiz-Medina, J., Ledent, C. and Valverde, O. (2011) GPR3 orphan receptor is involved in neuropathic pain after peripheral nerve injury and regulates morphine-induced antinociception. Neuropharmacology 61, 43-50.   DOI   ScienceOn
116 Ryberg, E., Larsson, N., Sjogren, S., Hjorth, S., Hermansson, N. O., Leonova, J., Elebring, T., Nilsson, K., Drmota, T. and Greasley, P. J. (2007) The orphan receptor GPR55 is a novel cannabinoid receptor. Br. J. Pharmacol. 152, 1092-1101.
117 Serhan, C. N., Chiang, N. and Van Dyke, T. E. (2008) Resolving infl ammation: dual anti-inflammatory and pro-resolution lipid mediators. Nat. Rev. Immunol. 8, 349-361.   DOI   ScienceOn
118 Serhan, C. N., Krishnamoorthy, S., Recchiuti, A. and Chiang, N. (2011) Novel anti-inflammatory--pro-resolving mediators and their receptors. Curr. Top. Med. Chem. 11, 629-647.   DOI
119 Shah, U. and Kowalski, T. J. (2010) GPR119 agonists for the potential treatment of type 2 diabetes and related metabolic disorders. Vitam. Horm. 84, 415-448.   DOI   ScienceOn
120 Sharman, J. L., Mpamhanga, C. P., Spedding, M., Germain, P., Staels, B., Dacquet, C., Laudet, V. and Harmar, A. J. (2011) IUPHAR-DB: new receptors and tools for easy searching and visualization of pharmacological data. Nucleic Acids Res. 39, D534-538.   DOI
121 Shukla, A. K., Xiao, K. and Lefkowitz, R. J. (2011) Emerging paradigms of beta-arrestin-dependent seven transmembrane receptor signaling. Trends Biochem. Sci. 36, 457-469.   DOI   ScienceOn
122 Im, D. S. (2004) Discovery of new G protein-coupled receptors for lipid mediators. J. Lipid Res. 45, 410-418.   DOI
123 Ignatov, A., Lintzel, J., Hermans-Borgmeyer, I., Kreienkamp, H. J., Joost, P., Thomsen, S., Methner, A. and Schaller, H. C. (2003) Role of the G-protein-coupled receptor GPR12 as high-affinity receptor for sphingosylphosphorylcholine and its expression and function in brain development. J. Neurosci. 23, 907-914.
124 Im, D. S. (2002) Orphan G protein-coupled receptors and beyond. Jpn. J. Pharmacol. 90, 101-106.   DOI   ScienceOn
125 Im, D. S. (2003) Linking Chinese medicine and G-protein-coupled receptors. Trends Pharmacol. Sci. 24, 2-4.   DOI   ScienceOn
126 Im, D. S. (2005) Two ligands for a GPCR, proton vs lysolipid. Acta Pharmacol. Sin. 26, 1435-1441.   DOI   ScienceOn
127 Im, D. S. (2009) New intercellular lipid mediators and their GPCRs: an update. Prostaglandins Other Lipid Mediat. 89, 53-56.   DOI   ScienceOn
128 Inoue, A., Ishiguro, J., Kitamura, H., Arima, N., Okutani, M., Shuto, A., Higashiyama, S., Ohwada, T., Arai, H., Makide, K. and Aoki, J. (2012) TGFalpha shedding assay: an accurate and versatile method for detecting GPCR activation. Nat. Methods 9, 1021-1029.   DOI   ScienceOn
129 Irannejad, R., Tomshine, J. C., Tomshine, J. R., Chevalier, M., Mahoney, J. P., Steyaert, J., Rasmussen, S. G., Sunahara, R. K., El-Samad, H., Huang, B. and von Zastrow, M. (2013) Conformational biosensors reveal GPCR signalling from endosomes. Nature 495, 534-538.   DOI   ScienceOn
130 Iwashita, M., Makide, K., Nonomura, T., Misumi, Y., Otani, Y., Ishida, M., Taguchi, R., Tsujimoto, M., Aoki, J., Arai, H. and Ohwada, T. (2009) Synthesis and evaluation of lysophosphatidylserine analogues as inducers of mast cell degranulation. Potent activities of lysophosphatidylthreonine and its 2-deoxy derivative. J. Med. Chem. 52, 5837-5863.   DOI   ScienceOn
131 Sugo, T., Tachimoto, H., Chikatsu, T., Murakami, Y., Kikukawa, Y., Sato, S., Kikuchi, K., Nagi, T., Harada, M., Ogi, K., Ebisawa, M. and Mori, M. (2006) Identification of a lysophosphatidylserine receptor on mast cells. Biochem. Biophys. Res. Commun. 341, 1078-1087.   DOI   ScienceOn
132 Soga, T., Ohishi, T., Matsui, T., Saito, T., Matsumoto, M., Takasaki, J., Matsumoto, S., Kamohara, M., Hiyama, H., Yoshida, S., Momose, K., Ueda, Y., Matsushime, H., Kobori, M. and Furuichi, K. (2005) Lysophosphatidylcholine enhances glucose-dependent insulin secretion via an orphan G-protein-coupled receptor. Biochem. Biophys. Res. Commun. 326, 744-751.   DOI   ScienceOn
133 Southern, C., Cook, J. M., Neetoo-Isseljee, Z., Taylor, D. L., Kettleborough, C. A., Merritt, A., Bassoni, D. L., Raab, W. J., Quinn, E., Wehrman, T. S., Davenport, A. P., Brown, A. J., Green, A., Wigglesworth, M. J. and Rees, S. (2013) Screening beta-arrestin recruitment for the identifi cation of natural ligands for orphan G-proteincoupled receptors. J. Biomol. Screen. 18, 599-609.   DOI   ScienceOn
134 Spann, N. J. and Glass, C. K. (2013) Sterols and oxysterols in immune cell function. Nat. Immunol. 14, 893-900.   DOI   ScienceOn
135 Suzuki, M., Takaishi, S., Nagasaki, M., Onozawa, Y., Iino, I., Maeda, H., Komai, T. and Oda, T. (2013) Medium-chain fatty acid-sensing receptor, GPR84, is a proinflammatory receptor. J. Biol. Chem. 288, 10684-10691.   DOI   ScienceOn
136 Swaney, J. S., Chapman, C., Correa, L. D., Stebbins, K. J., Broadhead, A. R., Bain, G., Santini, A. M., Darlington, J., King, C. D., Baccei, C. S., Lee, C., Parr, T. A., Roppe, J. R., Seiders, T. J., Ziff, J., Prasit, P., Hutchinson, J. H., Evans, J. F. and Lorrain, D. S. (2011) Pharmacokinetic and pharmacodynamic characterization of an oral lysophosphatidic acid type 1 receptor-selective antagonist. J. Pharmacol. Exp. Ther. 336, 693-700.   DOI   ScienceOn
137 Tabata, K., Baba, K., Shiraishi, A., Ito, M. and Fujita, N. (2007) The orphan GPCR GPR87 was deorphanized and shown to be a lysophosphatidic acid receptor. Biochem. Biophys. Res. Commun. 363, 861-866.   DOI   ScienceOn
138 Tanaka, S., Shaikh, I. M., Chiocca, E. A. and Saeki, Y. (2009) The Gslinked receptor GPR3 inhibits the proliferation of cerebellar granule cells during postnatal development. PloS one 4, e5922.   DOI   ScienceOn
139 Talukdar, S., Olefsky, J. M. and Osborn, O. (2011) Targeting GPR120 and other fatty acid-sensing GPCRs ameliorates insulin resistance and inflammatory diseases. Trends Pharmacol. Sci. 32, 543-550.   DOI   ScienceOn
140 Tanaka, S., Ishii, K., Kasai, K., Yoon, S. O. and Saeki, Y. (2007) Neural expression of G protein-coupled receptors GPR3, GPR6, and GPR12 up-regulates cyclic AMP levels and promotes neurite outgrowth. J. Biol. Chem. 282, 10506-10515.   DOI   ScienceOn
141 Thathiah, A., Spittaels, K., Hoffmann, M., Staes, M., Cohen, A., Horre, K., Vanbrabant, M., Coun, F., Baekelandt, V., Delacourte, A., Fischer, D. F., Pollet, D., De Strooper, B. and Merchiers, P. (2009) The orphan G protein-coupled receptor 3 modulates amyloid-beta peptide generation in neurons. Science 323, 946-951.   DOI   ScienceOn
142 Thomas, C., Pellicciari, R., Pruzanski, M., Auwerx, J. and Schoonjans, K. (2008) Targeting bile-acid signalling for metabolic diseases. Nat. Rev. Drug Discov. 7, 678-693.   DOI   ScienceOn
143 Tian, Z., Wang, Y., Zhang, N., Guo, Y. Y., Feng, B., Liu, S. B. and Zhao, M. G. (2013) Estrogen receptor GPR30 exerts anxiolytic effects by maintaining the balance between GABAergic and glutamatergic transmission in the basolateral amygdala of ovariectomized mice after stress. Psychoneuroendocrinology 38, 2218-2233.   DOI   ScienceOn
144 Tomura, H., Mogi, C., Sato, K. and Okajima, F. (2005) Proton-sensing and lysolipid-sensitive G-protein-coupled receptors: a novel type of multi-functional receptors. Cell. Signal. 17, 1466-1476.   DOI   ScienceOn
145 Tourino, C., Valjent, E., Ruiz-Medina, J., Herve, D., Ledent, C. and Valverde, O. (2012) The orphan receptor GPR3 modulates the early phases of cocaine reinforcement. Br. J. Pharmacol. 167, 892-904.   DOI   ScienceOn