Everything You Always Wanted to Know about β3-AR * (* But Were Afraid to Ask)
Abstract
:1. Introduction
2. Gene and Protein
3. Pharmacology of the Receptor
3.1. Agonists
3.2. Antagonists
4. β3-AR as Therapeutic Target
4.1. Urinary System
4.2. Adipose Tissue
4.3. Myocardium
4.4. Brain
4.5. Retina
4.6. Cancer Therapy
5. Polymorphisms
6. Conclusions
Funding
Acknowledgments
Conflicts of Interest
References
- Nergardh, A.; Boreus, L.O.; Naglo, A.S. Characterization of the adrenergic beta-receptor in the urinary bladder of man and cat. Acta Pharmacol. Toxicol. 1977, 40, 14–21. [Google Scholar] [CrossRef]
- Emorine, L.J.; Marullo, S.; Briend-Sutren, M.M.; Patey, G.; Tate, K.; Delavier-Klutchko, C.; Strosberg, A.D. Molecular characterization of the human beta 3-adrenergic receptor. Science 1989, 245, 1118–1121. [Google Scholar] [CrossRef]
- Procino, G.; Carmosino, M.; Milano, S.; Dal Monte, M.; Schena, G.; Mastrodonato, M.; Gerbino, A.; Bagnoli, P.; Svelto, M. Beta3 adrenergic receptor in the kidney may be a new player in sympathetic regulation of renal function. Kidney Int. 2016, 90, 555–567. [Google Scholar] [CrossRef]
- Decara, J.; Rivera, P.; Arrabal, S.; Vargas, A.; Serrano, A.; Pavon, F.J.; Dieguez, C.; Nogueiras, R.; Rodriguez de Fonseca, F.; Suarez, J. Cooperative role of the glucagon-like peptide-1 receptor and β3-adrenergic-mediated signalling on fat mass reduction through the downregulation of pka/akt/ampk signalling in the adipose tissue and muscle of rats. Acta Physiol. 2018, 222, e13008. [Google Scholar] [CrossRef]
- Banfi, S.; Gusarova, V.; Gromada, J.; Cohen, J.C.; Hobbs, H.H. Increased thermogenesis by a noncanonical pathway in angptl3/8-deficient mice. Proc. Natl. Acad. Sci. USA 2018, 115, E1249–E1258. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Hartung, J.E.; Bortsov, A.V.; Kim, S.; O’Buckley, S.C.; Kozlowski, J.; Nackley, A.G. Sustained stimulation of β2- and β3-adrenergic receptors leads to persistent functional pain and neuroinflammation. Brain Behav. Immun. 2018, 73, 520–532. [Google Scholar] [CrossRef] [PubMed]
- Chapple, C.R.; Cardozo, L.; Nitti, V.W.; Siddiqui, E.; Michel, M.C. Mirabegron in overactive bladder: A review of efficacy, safety, and tolerability. Neurourol. Urodyn. 2014, 33, 17–30. [Google Scholar] [CrossRef] [PubMed]
- Balligand, J.L. Cardiac β3-adrenergic receptors in the clinical arena: The end of the beginning. Eur. J. Heart Fail. 2017, 19, 576–578. [Google Scholar] [CrossRef]
- Baskin, A.S.; Linderman, J.D.; Brychta, R.J.; McGehee, S.; Anflick-Chames, E.; Cero, C.; Johnson, J.W.; O’Mara, A.E.; Fletcher, L.A.; Leitner, B.P.; et al. Regulation of human adipose tissue activation, gallbladder size, and bile acid metabolism by a β3-adrenergic receptor agonist. Diabetes 2018, 67, 2113–2125. [Google Scholar] [CrossRef]
- Jesus, I.C.; Alle, L.F.; Munhoz, E.C.; Silva, L.R.D.; Lopes, W.A.; Tureck, L.V.; Purim, K.S.M.; Titski, A.C.K.; Leite, N. Trp64Arg polymorphism of the ADRB3 gene associated with maximal fat oxidation and LDL-C levels in non-obese adolescents. J. Pediatr. 2018, 94, 425–431. [Google Scholar] [CrossRef]
- Sakane, N.; Sato, J.; Tsushita, K.; Tsujii, S.; Kotani, K.; Tominaga, M.; Kawazu, S.; Sato, Y.; Usui, T.; Kamae, I.; et al. Effects of lifestyle intervention on weight and metabolic parameters in patients with impaired glucose tolerance related to β-3 adrenergic receptor gene polymorphism Trp64Arg(C/T): Results from the japan diabetes prevention program. J. Diabetes Investig. 2016, 7, 338–342. [Google Scholar] [CrossRef]
- Granneman, J.G.; Lahners, K.N.; Chaudhry, A. Molecular cloning and expression of the rat β3-adrenergic receptor. Mol. Pharmacol. 1991, 40, 895–899. [Google Scholar]
- Nahmias, C.; Blin, N.; Elalouf, J.M.; Mattei, M.G.; Strosberg, A.D.; Emorine, L.J. Molecular characterization of the mouse β3-adrenergic receptor: Relationship with the atypical receptor of adipocytes. EMBO J. 1991, 10, 3721–3727. [Google Scholar] [CrossRef] [PubMed]
- Forrest, R.H.; Hickford, J.G. Rapid communication: Nucleotide sequences of the bovine, caprine, and ovine beta3-adrenergic receptor genes. J. Anim. Sci. 2000, 78, 1397–1398. [Google Scholar] [CrossRef]
- Sasaki, N.; Uchida, E.; Niiyama, M.; Yoshida, T.; Saito, M. Anti-obesity effects of selective agonists to the β3-adrenergic receptor in dogs. I. The presence of canine β3-adrenergic receptor and in vivo lipomobilization by its agonists. J. Vet. Med. Sci. 1998, 60, 459–463. [Google Scholar] [CrossRef]
- Jockers, R.; Da Silva, A.; Strosberg, A.D.; Bouvier, M.; Marullo, S. New molecular and structural determinants involved in β2-adrenergic receptor desensitization and sequestration. Delineation using chimeric β3/β2-adrenergic receptors. J. Biol. Chem. 1996, 271, 9355–9362. [Google Scholar] [CrossRef] [PubMed]
- Nantel, F.; Bonin, H.; Emorine, L.J.; Zilberfarb, V.; Strosberg, A.D.; Bouvier, M.; Marullo, S. The human β3-adrenergic receptor is resistant to short term agonist-promoted desensitization. Mol. Pharmacol. 1993, 43, 548–555. [Google Scholar]
- Granneman, J.G.; Lahners, K.N.; Chaudhry, A. Characterization of the human β3-adrenergic receptor gene. Mol. Pharmacol. 1993, 44, 264–270. [Google Scholar] [PubMed]
- Van Spronsen, A.; Nahmias, C.; Krief, S.; Briend-Sutren, M.M.; Strosberg, A.D.; Emorine, L.J. The promoter and intron/exon structure of the human and mouse β3-adrenergic-receptor genes. Eur. J. Biochem. 1993, 213, 1117–1124. [Google Scholar] [CrossRef] [PubMed]
- Evans, B.A.; Papaioannou, M.; Hamilton, S.; Summers, R.J. Alternative splicing generates two isoforms of the β3-adrenoceptor which are differentially expressed in mouse tissues. Br. J. Pharmacol. 1999, 127, 1525–1531. [Google Scholar] [CrossRef] [Green Version]
- Hutchinson, D.S.; Bengtsson, T.; Evans, B.A.; Summers, R.J. Mouse β3a- and β3b-adrenoceptors expressed in chinese hamster ovary cells display identical pharmacology but utilize distinct signalling pathways. Br. J. Pharmacol. 2002, 135, 1903–1914. [Google Scholar] [CrossRef] [PubMed]
- Milano, S.; Gerbino, A.; Schena, G.; Carmosino, M.; Svelto, M.; Procino, G. Human β3-adrenoreceptor is resistant to agonist-induced desensitization in renal epithelial cells. Cell Physiol. Biochem. 2018, 48, 847–862. [Google Scholar] [CrossRef] [PubMed]
- Okeke, K.; Angers, S.; Bouvier, M.; Michel, M.C. Agonist-induced desensitisation ofβ3-adrenoceptors: Where, when and how? Br. J. Pharmacol. 2019. [Google Scholar] [CrossRef] [PubMed]
- Lefkowitz, R.J. G protein-coupled receptors. III. New roles for receptor kinases and β-arrestins in receptor signaling and desensitization. J. Biol. Chem. 1998, 273, 18677–18680. [Google Scholar] [CrossRef] [PubMed]
- Michel-Reher, M.B.; Michel, M.C. Agonist-induced desensitization of human β3-adrenoceptors expressed in human embryonic kidney cells. Naunyn Schmiedebergs Arch. Pharmacol. 2013, 386, 843–851. [Google Scholar] [CrossRef]
- Strosberg, A.D.; Pietri-Rouxel, F. Function and regulation of the β3-adrenoceptor. Trends Pharmacol. Sci. 1996, 17, 373–381. [Google Scholar] [CrossRef]
- Moffett, S.; Mouillac, B.; Bonin, H.; Bouvier, M. Altered phosphorylation and desensitization patterns of a human β2-adrenergic receptor lacking the palmitoylated Cys341. EMBO J. 1993, 12, 349–356. [Google Scholar] [CrossRef]
- Baker, J.G. The selectivity of beta-adrenoceptor antagonists at the human β1, β2 and β3 adrenoceptors. Br. J. Pharmacol. 2005, 144, 317–322. [Google Scholar] [CrossRef]
- Dessy, C.; Balligand, J.L. Beta3-adrenergic receptors in cardiac and vascular tissues emerging concepts and therapeutic perspectives. Adv. Pharmacol. 2010, 59, 135–163. [Google Scholar]
- Emorine, L.; Blin, N.; Strosberg, A.D. The human β3-adrenoceptor: The search for a physiological function. Trends Pharmacol. Sci. 1994, 15, 3–7. [Google Scholar] [CrossRef]
- Hoffmann, C.; Leitz, M.R.; Oberdorf-Maass, S.; Lohse, M.J.; Klotz, K.N. Comparative pharmacology of human beta-adrenergic receptor subtypes—Characterization of stably transfected receptors in cho cells. Naunyn Schmiedebergs Arch. Pharmacol. 2004, 369, 151–159. [Google Scholar] [CrossRef]
- Rozec, B.; Gauthier, C. Beta3-adrenoceptors in the cardiovascular system: Putative roles in human pathologies. Pharmacol. Ther. 2006, 111, 652–673. [Google Scholar] [CrossRef]
- Strosberg, A.D. Structure and function of the β3-adrenergic receptor. Annu. Rev. Pharmacol. Toxicol. 1997, 37, 421–450. [Google Scholar] [CrossRef]
- Perrone, M.G.; Bleve, L.; Santandrea, E.; Vitale, P.; Niso, M.; Scilimati, A. The tertiary amine nitrogen atom of piperazine sulfonamides as a novel determinant of potent and selective β3-adrenoceptor agonists. ChemMedChem 2009, 4, 2080–2097. [Google Scholar] [CrossRef]
- Perrone, M.G.; Scilimati, A. B3-adrenoceptor agonists and (antagonists as) inverse agonists: History, perspective, constitutive activity, and stereospecific binding. Methods Enzymol. 2010, 484, 197–230. [Google Scholar]
- Cernecka, H.; Sand, C.; Michel, M.C. The odd sibling: Features of β3-adrenoceptor pharmacology. Mol. Pharmacol. 2014, 86, 479–484. [Google Scholar] [CrossRef]
- Vrydag, W.; Michel, M.C. Tools to study β3-adrenoceptors. Naunyn Schmiedebergs Arch. Pharmacol. 2007, 374, 385–398. [Google Scholar] [CrossRef] [Green Version]
- Takasu, T.; Ukai, M.; Sato, S.; Matsui, T.; Nagase, I.; Maruyama, T.; Sasamata, M.; Miyata, K.; Uchida, H.; Yamaguchi, O. Effect of (r)-2-(2-aminothiazol-4-yl)-4′-{2-[(2-hydroxy-2-phenylethyl)amino]ethyl} acetanilide (ym178), a novel selective beta3-adrenoceptor agonist, on bladder function. J. Pharmacol. Exp. Ther. 2007, 321, 642–647. [Google Scholar] [CrossRef]
- Bianchetti, A.; Manara, L. In vitro inhibition of intestinal motility by phenylethanolaminotetralines: Evidence of atypical beta-adrenoceptors in rat colon. Br. J. Pharmacol. 1990, 100, 831–839. [Google Scholar] [CrossRef]
- Sanofi. Available online: http://www.sanofi.com/ (accessed on 14 February 2019).
- Simiand, J.; Keane, P.E.; Guitard, J.; Langlois, X.; Gonalons, N.; Martin, P.; Bianchetti, A.; Le Fur, G.; Soubrie, P. Antidepressant profile in rodents of sr 58611a, a new selective agonist for atypical beta-adrenoceptors. Eur. J. Pharmacol. 1992, 219, 193–201. [Google Scholar] [CrossRef]
- Hicks, A.; McCafferty, G.P.; Riedel, E.; Aiyar, N.; Pullen, M.; Evans, C.; Luce, T.D.; Coatney, R.W.; Rivera, G.C.; Westfall, T.D.; et al. Gw427353 (solabegron), a novel, selective beta3-adrenergic receptor agonist, evokes bladder relaxation and increases micturition reflex threshold in the dog. J. Pharmacol. Exp. Ther. 2007, 323, 202–209. [Google Scholar] [CrossRef] [PubMed]
- Ohlstein, E.H.; von Keitz, A.; Michel, M.C. A multicenter, double-blind, randomized, placebo-controlled trial of the β3-adrenoceptor agonist solabegron for overactive bladder. Eur. Urol. 2012, 62, 834–840. [Google Scholar] [CrossRef] [PubMed]
- Igawa, Y.; Schneider, T.; Yamazaki, Y.; Tatemichi, S.; Homma, Y.; Nishizawa, O.; Michel, M.C. Functional investigation of beta-adrenoceptors in human isolated detrusor focusing on the novel selective β3-adrenoceptor agonist kuc-7322. Naunyn Schmiedebergs Arch. Pharmacol. 2012, 385, 759–767. [Google Scholar] [CrossRef] [PubMed]
- Michel, M.C.; Korstanje, C. Beta3-adrenoceptor agonists for overactive bladder syndrome: Role of translational pharmacology in a repositioning clinical drug development project. Pharmacol. Ther. 2016, 159, 66–82. [Google Scholar] [CrossRef]
- Keam, S.J. Vibegron: First global approval. Drugs 2018, 78, 1835–1839. [Google Scholar] [CrossRef] [PubMed]
- Yoshida, M.; Takeda, M.; Gotoh, M.; Nagai, S.; Kurose, T. Vibegron, a novel potent and selective β3-adrenoreceptor agonist, for the treatment of patients with overactive bladder: A randomized, double-blind, placebo-controlled phase 3 study. Eur. Urol. 2018, 73, 783–790. [Google Scholar] [CrossRef] [PubMed]
- Blin, N.; Nahmias, C.; Drumare, M.F.; Strosberg, A.D. Mediation of most atypical effects by species homologues of the beta 3-adrenoceptor. Br. J. Pharmacol. 1994, 112, 911–919. [Google Scholar] [CrossRef]
- D’Agostino, G.; Condino, A.M.; Calvi, P. Involvement of β3-adrenoceptors in the inhibitory control of cholinergic activity in human bladder: Direct evidence by [(3)h]-acetylcholine release experiments in the isolated detrusor. Eur. J. Pharmacol. 2015, 758, 115–122. [Google Scholar] [CrossRef] [PubMed]
- Mitidieri, E.; Tramontano, T.; Gurgone, D.; Imbimbo, C.; Mirone, V.; Fusco, F.; Cirino, G.; d’Emmanuele di Villa Bianca, R.; Sorrentino, R. Beta3 adrenergic receptor activation relaxes human corpus cavernosum and penile artery through a hydrogen sulfide/cgmp-dependent mechanism. Pharmacol. Res. 2017, 124, 100–104. [Google Scholar] [CrossRef]
- Baker, J.G. The selectivity of β-adrenoceptor agonists at human β1-, β2- and β3-adrenoceptors. Br. J. Pharmacol. 2010, 160, 1048–1061. [Google Scholar] [CrossRef] [Green Version]
- Leblais, V.; Pourageaud, F.; Ivorra, M.D.; Marthan, R.; Muller, B. Comparison of the alpha-adrenoceptor-mediated effects of β3-adrenoceptor ligands in rat pulmonary artery. Naunyn Schmiedebergs Arch. Pharmacol. 2005, 371, 535–539. [Google Scholar] [CrossRef]
- Pott, C.; Brixius, K.; Bundkirchen, A.; Bolck, B.; Bloch, W.; Steinritz, D.; Mehlhorn, U.; Schwinger, R.H. The preferential β3-adrenoceptor agonist brl 37344 increases force via β1-/β2-adrenoceptors and induces endothelial nitric oxide synthase via beta3-adrenoceptors in human atrial myocardium. Br. J. Pharmacol. 2003, 138, 521–529. [Google Scholar] [CrossRef]
- Wuest, M.; Eichhorn, B.; Grimm, M.O.; Wirth, M.P.; Ravens, U.; Kaumann, A.J. Catecholamines relax detrusor through β2-adrenoceptors in mouse and β3-adrenoceptors in man. J. Pharmacol. Exp. Ther. 2009, 328, 213–222. [Google Scholar] [CrossRef]
- Hutchinson, D.S.; Chernogubova, E.; Sato, M.; Summers, R.J.; Bengtsson, T. Agonist effects of zinterol at the mouse and human β3-adrenoceptor. Naunyn Schmiedebergs Arch. Pharmacol. 2006, 373, 158–168. [Google Scholar] [CrossRef] [PubMed]
- Puzzo, D.; Raiteri, R.; Castaldo, C.; Capasso, R.; Pagano, E.; Tedesco, M.; Gulisano, W.; Drozd, L.; Lippiello, P.; Palmeri, A.; et al. Cl316,243, a β3-adrenergic receptor agonist, induces muscle hypertrophy and increased strength. Sci. Rep. 2016, 5, 37504. [Google Scholar] [CrossRef] [PubMed]
- Lynch, G.S.; Ryall, J.G. Role of beta-adrenoceptor signaling in skeletal muscle: Implications for muscle wasting and disease. Physiol. Rev. 2008, 88, 729–767. [Google Scholar] [CrossRef]
- Karimi Galougahi, K.; Liu, C.C.; Garcia, A.; Gentile, C.; Fry, N.A.; Hamilton, E.J.; Hawkins, C.L.; Figtree, G.A. B3 adrenergic stimulation restores nitric oxide/redox balance and enhances endothelial function in hyperglycemia. J. Am. Heart Assoc. 2016, 5, e002824. [Google Scholar] [CrossRef]
- Biers, S.M.; Reynard, J.M.; Brading, A.F. The effects of a new selective β3-adrenoceptor agonist (GW427353) on spontaneous activity and detrusor relaxation in human bladder. BJU Int. 2006, 98, 1310–1314. [Google Scholar] [CrossRef]
- Rodriguez, M.; Carillon, C.; Coquerel, A.; Le Fur, G.; Ferrara, P.; Caput, D.; Shire, D. Evidence for the presence of β3-adrenergic receptor mrna in the human brain. Brain Res. Mol. Brain Res. 1995, 29, 369–375. [Google Scholar] [CrossRef]
- Summers, R.J.; Papaioannou, M.; Harris, S.; Evans, B.A. Expression of β3-adrenoceptor mrna in rat brain. Br. J. Pharmacol. 1995, 116, 2547–2548. [Google Scholar] [CrossRef] [PubMed]
- Consoli, D.; Leggio, G.M.; Mazzola, C.; Micale, V.; Drago, F. Behavioral effects of the β3 adrenoceptor agonist sr58611a: Is it the putative prototype of a new class of antidepressant/anxiolytic drugs? Eur. J. Pharmacol. 2007, 573, 139–147. [Google Scholar] [CrossRef]
- Overstreet, D.H.; Stemmelin, J.; Griebel, G. Confirmation of antidepressant potential of the selective β3 adrenoceptor agonist amibegron in an animal model of depression. Pharmacol. Biochem. Behav. 2008, 89, 623–626. [Google Scholar] [CrossRef]
- Stemmelin, J.; Cohen, C.; Terranova, J.P.; Lopez-Grancha, M.; Pichat, P.; Bergis, O.; Decobert, M.; Santucci, V.; Francon, D.; Alonso, R.; et al. Stimulation of the β3-adrenoceptor as a novel treatment strategy for anxiety and depressive disorders. Neuropsychopharmacology 2008, 33, 574–587. [Google Scholar] [CrossRef]
- Stemmelin, J.; Cohen, C.; Yalcin, I.; Keane, P.; Griebel, G. Implication of β3-adrenoceptors in the antidepressant-like effects of amibegron using adrb3 knockout mice in the chronic mild stress. Behav. Brain Res. 2010, 206, 310–312. [Google Scholar] [CrossRef]
- Jin, J.; Miao, C.; Wang, Z.; Zhang, W.; Zhang, X.; Xie, X.; Lu, W. Design and synthesis of aryloxypropanolamine as β3-adrenergic receptor antagonist in cancer and lipolysis. Eur. J. Med. Chem. 2018, 150, 757–770. [Google Scholar] [CrossRef]
- Wada, Y.; Nakano, S.; Morimoto, A.; Kasahara, K.I.; Hayashi, T.; Takada, Y.; Suzuki, H.; Niwa-Sakai, M.; Ohashi, S.; Mori, M.; et al. Discovery of novel indazole derivatives as orally available β3-adrenergic receptor agonists lacking off-target-based cardiovascular side effects. J. Med. Chem. 2017, 60, 3252–3265. [Google Scholar] [CrossRef]
- Tewatia, P.; Malik, B.K.; Sahi, S. Identification of novel β3-adrenoceptor agonists using energetic analysis, structure based pharmacophores and virtual screening. Comb. Chem. High. Throughput Screen 2012, 15, 623–640. [Google Scholar] [CrossRef]
- Manara, L.; Croci, T.; Landi, M. β3-adrenoceptors and intestinal motility. Fundam. Clin. Pharmacol. 1995, 9, 332–342. [Google Scholar] [CrossRef]
- Nisoli, E.; Tonello, C.; Landi, M.; Carruba, M.O. Functional studies of the first selective β3-adrenergic receptor antagonist sr 59230a in rat brown adipocytes. Mol. Pharmacol. 1996, 49, 7–14. [Google Scholar]
- Casini, G.; Dal Monte, M.; Fornaciari, I.; Filippi, L.; Bagnoli, P. The β-adrenergic system as a possible new target for pharmacologic treatment of neovascular retinal diseases. Prog. Retin Eye Res. 2014, 42, 103–129. [Google Scholar] [CrossRef]
- Wang, B.; Xu, M.; Li, W.; Li, X.; Zheng, Q.; Niu, X. Aerobic exercise protects against pressure overload-induced cardiac dysfunction and hypertrophy via β3-AR-nNOS-NO activation. PLoS ONE 2017, 12, e0179648. [Google Scholar] [CrossRef]
- Wu, M.J.; Shin, D.H.; Kim, M.Y.; Park, C.G.; Kim, Y.D.; Lee, J.; Park, I.K.; Choi, S.; So, I.; Park, J.S.; et al. Functional effects of β3-adrenoceptor on pacemaker activity in interstitial cells of cajal from the mouse colon. Eur. J. Pharmacol. 2015, 754, 32–40. [Google Scholar] [CrossRef] [PubMed]
- Niclauss, N.; Michel-Reher, M.B.; Alewijnse, A.E.; Michel, M.C. Comparison of three radioligands for the labelling of human β-adrenoceptor subtypes. Naunyn Schmiedebergs Arch. Pharmacol. 2006, 374, 99–105. [Google Scholar] [CrossRef] [PubMed]
- Sato, M.; Horinouchi, T.; Hutchinson, D.S.; Evans, B.A.; Summers, R.J. Ligand-directed signaling at the β3-adrenoceptor produced by 3-(2-ethylphenoxy)-1-[(1,s)-1,2,3,4-tetrahydronapth-1-ylamino]-2s-2-propanol oxalate (sr59230a) relative to receptor agonists. Mol. Pharmacol. 2007, 72, 1359–1368. [Google Scholar] [CrossRef]
- Hutchinson, D.S.; Sato, M.; Evans, B.A.; Christopoulos, A.; Summers, R.J. Evidence for pleiotropic signaling at the mouse β3-adrenoceptor revealed by sr59230a [3-(2-ethylphenoxy)-1-[(1,s)-1,2,3,4-tetrahydronapth-1-ylamino]-2s-2-propanol oxalate]. J. Pharmacol. Exp. Ther. 2005, 312, 1064–1074. [Google Scholar] [CrossRef] [PubMed]
- Candelore, M.R.; Deng, L.; Tota, L.; Guan, X.M.; Amend, A.; Liu, Y.; Newbold, R.; Cascieri, M.A.; Weber, A.E. Potent and selective human β3-adrenergic receptor antagonists. J. Pharmacol. Exp. Ther. 1999, 290, 649–655. [Google Scholar]
- Van Wieringen, J.P.; Michel-Reher, M.B.; Hatanaka, T.; Ueshima, K.; Michel, M.C. The new radioligand [(3)h]-l 748,337 differentially labels human and rat β3-adrenoceptors. Eur. J. Pharmacol. 2013, 720, 124–130. [Google Scholar] [CrossRef] [PubMed]
- Sato, M.; Hutchinson, D.S.; Evans, B.A.; Summers, R.J. The β3-adrenoceptor agonist 4-[[(Hexylamino) carbonyl] amino]-N-[4-[2-[[(2S)-2-hydroxy-3-(4-hydroxyphenoxy) propyl] amino] ethyl]-phenyl]-benzenesulfonamide (L755507) and antagonist (S)-N-[4-[2-[[3-[3-(acetamidomethyl) phenoxy]-2-hydroxypropyl] amino]-ethyl] phenyl] benzenesulfonamide (L748337) activate different signaling pathways in Chinese hamster ovary-K1 cells stably expressing the human β3-adrenoceptor. Mol. Pharmacol. 2008, 74, 1417–1428. [Google Scholar] [PubMed]
- Uhlen, M.; Fagerberg, L.; Hallstrom, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, A.; Kampf, C.; Sjostedt, E.; Asplund, A.; et al. Proteomics. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef] [PubMed]
- Michel, M.C.; Vrydag, W. α1-, α2- and β-adrenoceptors in the urinary bladder, urethra and prostate. Br. J. Pharmacol. 2006, 147 (Suppl. 2), S88–S119. [Google Scholar] [CrossRef] [Green Version]
- Shen, H.; Chen, Z.; Mokhtar, A.D.; Bi, X.; Wu, G.; Gong, S.; Huang, C.; Li, S.; Du, S. Expression of beta-adrenergic receptor subtypes in human normal and dilated ureter. Int. Urol. Nephrol. 2017, 49, 1771–1778. [Google Scholar] [CrossRef] [PubMed]
- Limberg, B.J.; Andersson, K.E.; Aura Kullmann, F.; Burmer, G.; de Groat, W.C.; Rosenbaum, J.S. β-adrenergic receptor subtype expression in myocyte and non-myocyte cells in human female bladder. Cell Tissue Res. 2010, 342, 295–306. [Google Scholar] [CrossRef] [PubMed]
- Calmasini, F.B.; Candido, T.Z.; Alexandre, E.C.; D’Ancona, C.A.; Silva, D.; de Oliveira, M.A.; De Nucci, G.; Antunes, E.; Monica, F.Z. The β-3 adrenoceptor agonist, mirabegron relaxes isolated prostate from human and rabbit: New therapeutic indication? Prostate 2015, 75, 440–447. [Google Scholar] [CrossRef] [PubMed]
- Otsuka, A.; Shinbo, H.; Matsumoto, R.; Kurita, Y.; Ozono, S. Expression and functional role of β-adrenoceptors in the human urinary bladder urothelium. Naunyn Schmiedebergs Arch. Pharmacol. 2008, 377, 473–481. [Google Scholar] [CrossRef] [PubMed]
- Yamaguchi, O. β3-adrenoceptors in human detrusor muscle. Urology 2002, 59, 25–29. [Google Scholar] [CrossRef]
- Larsen, J.J. Alpha and beta-adrenoceptors in the detrusor muscle and bladder base of the pig and β-adrenoceptors in the detrusor muscle of man. Br. J. Pharmacol. 1979, 65, 215–222. [Google Scholar] [CrossRef]
- Igawa, Y.; Yamazaki, Y.; Takeda, H.; Hayakawa, K.; Akahane, M.; Ajisawa, Y.; Yoneyama, T.; Nishizawa, O.; Andersson, K.E. Functional and molecular biological evidence for a possible β3-adrenoceptor in the human detrusor muscle. Br. J. Pharmacol. 1999, 126, 819–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peirce, V.; Carobbio, S.; Vidal-Puig, A. The different shades of fat. Nature 2014, 510, 76–83. [Google Scholar] [CrossRef]
- Tagaya, E.; Tamaoki, J.; Takemura, H.; Isono, K.; Nagai, A. Atypical adrenoceptor-mediated relaxation of canine pulmonary artery through a cyclic adenosine monophosphate-dependent pathway. Lung 1999, 177, 321–332. [Google Scholar] [CrossRef]
- Tanaka, Y.; Horinouchi, T.; Koike, K. New insights into beta-adrenoceptors in smooth muscle: Distribution of receptor subtypes and molecular mechanisms triggering muscle relaxation. Clin. Exp. Pharmacol. Physiol. 2005, 32, 503–514. [Google Scholar] [CrossRef]
- Uchida, H.; Shishido, K.; Nomiya, M.; Yamaguchi, O. Involvement of cyclic amp-dependent and -independent mechanisms in the relaxation of rat detrusor muscle via β-adrenoceptors. Eur. J. Pharmacol. 2005, 518, 195–202. [Google Scholar] [CrossRef]
- Maki, T.; Kajioka, S.; Itsumi, M.; Kareman, E.; Lee, K.; Shiota, M.; Eto, M. Mirabegron induces relaxant effects via camp signaling-dependent and -independent pathways in detrusor smooth muscle. Low. Urin. Tract Symptoms 2019. [Google Scholar] [CrossRef]
- Frazier, E.P.; Mathy, M.J.; Peters, S.L.; Michel, M.C. Does cyclic amp mediate rat urinary bladder relaxation by isoproterenol? J. Pharmacol. Exp. Ther. 2005, 313, 260–267. [Google Scholar] [CrossRef]
- Patel, H.J.; Giembycz, M.A.; Keeling, J.E.; Barnes, P.J.; Belvisi, M.G. Inhibition of cholinergic neurotransmission in guinea pig trachea by ns1619, a putative activator of large-conductance, calcium-activated potassium channels. J. Pharmacol. Exp. Ther. 1998, 286, 952–958. [Google Scholar]
- Coelho, A.; Antunes-Lopes, T.; Gillespie, J.; Cruz, F. β-3 adrenergic receptor is expressed in acetylcholine-containing nerve fibers of the human urinary bladder: An immunohistochemical study. Neurourol. Urodyn. 2017, 36, 1972–1980. [Google Scholar] [CrossRef]
- Kaufmann, J.; Martinka, P.; Moede, O.; Sendeski, M.; Steege, A.; Fahling, M.; Hultstrom, M.; Gaestel, M.; Moraes-Silva, I.C.; Nikitina, T.; et al. Noradrenaline enhances angiotensin ii responses via p38 mapk activation after hypoxia/re-oxygenation in renal interlobar arteries. Acta Physiol. 2015, 213, 920–932. [Google Scholar] [CrossRef]
- Rojek, A.; Nielsen, J.; Brooks, H.L.; Gong, H.; Kim, Y.H.; Kwon, T.H.; Frokiaer, J.; Nielsen, S. Altered expression of selected genes in kidney of rats with lithium-induced ndi. Am. J. Physiol. Renal. Physiol. 2005, 288, F1276–F1289. [Google Scholar] [CrossRef] [PubMed]
- Rains, S.L.; Amaya, C.N.; Bryan, B.A. β-adrenergic receptors are expressed across diverse cancers. Oncoscience 2017, 4, 95–105. [Google Scholar]
- Chen, S.F.; Lee, C.L.; Kuo, H.C. Changes in sensory proteins in the bladder urothelium of patients with chronic kidney disease and end-stage renal disease. Low. Urin. Tract Symptoms 2018. [Google Scholar] [CrossRef]
- Michel, M.C.; Gravas, S. Safety and tolerability of β3-adrenoceptor agonists in the treatment of overactive bladder syndrome-insig.ht from transcriptosome and experimental studies. Expert Opin Drug Saf. 2016, 15, 647–657. [Google Scholar]
- Arch, J.R.; Ainsworth, A.T.; Cawthorne, M.A.; Piercy, V.; Sennitt, M.V.; Thody, V.E.; Wilson, C.; Wilson, S. Atypical β-adrenoceptor on brown adipocytes as target for anti-obesity drugs. Nature 1984, 309, 163–165. [Google Scholar] [CrossRef] [PubMed]
- Krief, S.; Lonnqvist, F.; Raimbault, S.; Baude, B.; Van Spronsen, A.; Arner, P.; Strosberg, A.D.; Ricquier, D.; Emorine, L.J. Tissue distribution of β3-adrenergic receptor mrna in man. J. Clin. Investig. 1993, 91, 344–349. [Google Scholar] [CrossRef]
- Chamberlain, P.D.; Jennings, K.H.; Paul, F.; Cordell, J.; Berry, A.; Holmes, S.D.; Park, J.; Chambers, J.; Sennitt, M.V.; Stock, M.J.; et al. The tissue distribution of the human β3-adrenoceptor studied using a monoclonal antibody: Direct evidence of the β3-adrenoceptor in human adipose tissue, atrium and skeletal muscle. Int. J. Obes. Relat. Metab. Disord 1999, 23, 1057–1065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Matteis, R.; Arch, J.R.; Petroni, M.L.; Ferrari, D.; Cinti, S.; Stock, M.J. Immunohistochemical identification of the β3-adrenoceptor in intact human adipocytes and ventricular myocardium: Effect of obesity and treatment with ephedrine and caffeine. Int. J. Obes. Relat. Metab. Disord 2002, 26, 1442–1450. [Google Scholar] [CrossRef]
- Cypess, A.M.; Lehman, S.; Williams, G.; Tal, I.; Rodman, D.; Goldfine, A.B.; Kuo, F.C.; Palmer, E.L.; Tseng, Y.H.; Doria, A.; et al. Identification and importance of brown adipose tissue in adult humans. N. Engl. J. Med. 2009, 360, 1509–1517. [Google Scholar] [CrossRef]
- Hibi, M.; Oishi, S.; Matsushita, M.; Yoneshiro, T.; Yamaguchi, T.; Usui, C.; Yasunaga, K.; Katsuragi, Y.; Kubota, K.; Tanaka, S.; et al. Brown adipose tissue is involved in diet-induced thermogenesis and whole-body fat utilization in healthy humans. Int. J. Obes. 2016, 40, 1655–1661. [Google Scholar] [CrossRef] [Green Version]
- Sennitt, M.V.; Kaumann, A.J.; Molenaar, P.; Beeley, L.J.; Young, P.W.; Kelly, J.; Chapman, H.; Henson, S.M.; Berge, J.M.; Dean, D.K.; et al. The contribution of classical (β1/2-) and atypical beta-adrenoceptors to the stimulation of human white adipocyte lipolysis and right atrial appendage contraction by novel β3-adrenoceptor agonists of differing selectivities. J. Pharmacol. Exp. Ther. 1998, 285, 1084–1095. [Google Scholar]
- Tavernier, G.; Barbe, P.; Galitzky, J.; Berlan, M.; Caput, D.; Lafontan, M.; Langin, D. Expression of β3-adrenoceptors with low lipolytic action in human subcutaneous white adipocytes. J. Lipid Res. 1996, 37, 87–97. [Google Scholar]
- Robidoux, J.; Kumar, N.; Daniel, K.W.; Moukdar, F.; Cyr, M.; Medvedev, A.V.; Collins, S. Maximal β3-adrenergic regulation of lipolysis involves src and epidermal growth factor receptor-dependent ERK1/2 activation. J. Biol. Chem. 2006, 281, 37794–37802. [Google Scholar] [CrossRef]
- Soeder, K.J.; Snedden, S.K.; Cao, W.; Della Rocca, G.J.; Daniel, K.W.; Luttrell, L.M.; Collins, S. The β3-adrenergic receptor activates mitogen-activated protein kinase in adipocytes through a Gi-dependent mechanism. J. Biol. Chem. 1999, 274, 12017–12022. [Google Scholar] [CrossRef]
- Hodis, J.; Vaclavikova, R.; Farghali, H. β-3 agonist-induced lipolysis and nitric oxide production: Relationship to ppargamma agonist/antagonist and amp kinase modulation. Gen. Physiol. Biophys. 2011, 30, 90–99. [Google Scholar] [CrossRef]
- Barbatelli, G.; Murano, I.; Madsen, L.; Hao, Q.; Jimenez, M.; Kristiansen, K.; Giacobino, J.P.; De Matteis, R.; Cinti, S. The emergence of cold-induced brown adipocytes in mouse white fat depots is determined predominantly by white to brown adipocyte transdifferentiation. Am. J. Physiol. Endocrinol. Metab. 2010, 298, E1244–E1253. [Google Scholar] [CrossRef]
- Jiang, Y.; Berry, D.C.; Graff, J.M. Distinct cellular and molecular mechanisms for β3 adrenergic receptor-induced beige adipocyte formation. Elife 2017, 6, e30329. [Google Scholar] [CrossRef]
- De Jong, J.M.A.; Wouters, R.T.F.; Boulet, N.; Cannon, B.; Nedergaard, J.; Petrovic, N. The β3-adrenergic receptor is dispensable for browning of adipose tissues. Am. J. Physiol. Endocrinol. Metab. 2017, 312, E508–E518. [Google Scholar] [CrossRef]
- Susulic, V.S.; Frederich, R.C.; Lawitts, J.; Tozzo, E.; Kahn, B.B.; Harper, M.E.; Himms-Hagen, J.; Flier, J.S.; Lowell, B.B. Targeted disruption of the β3-adrenergic receptor gene. J. Biol. Chem. 1995, 270, 29483–29492. [Google Scholar] [CrossRef]
- Mattsson, C.L.; Csikasz, R.I.; Chernogubova, E.; Yamamoto, D.L.; Hogberg, H.T.; Amri, E.Z.; Hutchinson, D.S.; Bengtsson, T. β1-Adrenergic receptors increase UCP1 in human MADS brown adipocytes and rescue cold-acclimated β3-adrenergic receptor-knockout mice via nonshivering thermogenesis. Am. J. Physiol. Endocrinol. Metab. 2011, 301, E1108–E1118. [Google Scholar] [CrossRef]
- Preite, N.Z.; Nascimento, B.P.; Muller, C.R.; Americo, A.L.; Higa, T.S.; Evangelista, F.S.; Lancellotti, C.L.; Henriques, F.S.; Batista, M.L., Jr.; Bianco, A.C.; et al. Disruption of β3 adrenergic receptor increases susceptibility to dio in mouse. J. Endocrinol. 2016, 231, 259–269. [Google Scholar] [CrossRef]
- Hong, S.; Song, W.; Zushin, P.H.; Liu, B.; Jedrychowski, M.P.; Mina, A.I.; Deng, Z.; Cabarkapa, D.; Hall, J.A.; Palmer, C.J.; et al. Phosphorylation of β-3 adrenergic receptor at serine 247 by erk map kinase drives lipolysis in obese adipocytes. Mol. Metab. 2018, 12, 25–38. [Google Scholar] [CrossRef]
- Clookey, S.L.; Welly, R.J.; Shay, D.; Woodford, M.L.; Fritsche, K.L.; Rector, R.S.; Padilla, J.; Lubahn, D.B.; Vieira-Potter, V.J. β3 adrenergic receptor activation rescues metabolic dysfunction in female estrogen receptor alpha-null mice. Front. Physiol. 2019, 10, 9. [Google Scholar] [CrossRef]
- Xiao, C.; Goldgof, M.; Gavrilova, O.; Reitman, M.L. Anti-obesity and metabolic efficacy of the β3-adrenergic agonist, CL316243, in mice at thermoneutrality compared to 22 °C. Obesity 2015, 23, 1450–1459. [Google Scholar] [CrossRef]
- Bhadada, S.V.; Patel, B.M.; Mehta, A.A.; Goyal, R.K. β3 receptors: Role in cardiometabolic disorders. Ther. Adv. Endocrinol. Metab. 2011, 2, 65–79. [Google Scholar] [CrossRef]
- Oana, F.; Takeda, H.; Matsuzawa, A.; Akahane, S.; Isaji, M.; Akahane, M. Adiponectin receptor 2 expression in liver and insulin resistance in DB/DB mice given a β3-adrenoceptor agonist. Eur. J. Pharmacol. 2005, 518, 71–76. [Google Scholar] [CrossRef]
- Larsen, T.M.; Toubro, S.; van Baak, M.A.; Gottesdiener, K.M.; Larson, P.; Saris, W.H.; Astrup, A. Effect of a 28-d treatment with L-796568, a novel β3-adrenergic receptor agonist, on energy expenditure and body composition in obese men. Am. J. Clin. Nutr. 2002, 76, 780–788. [Google Scholar] [CrossRef]
- Van Baak, M.A.; Hul, G.B.; Toubro, S.; Astrup, A.; Gottesdiener, K.M.; DeSmet, M.; Saris, W.H. Acute effect of L-796568, a novel β3-adrenergic receptor agonist, on energy expenditure in obese men. Clin. Pharmacol. Ther. 2002, 71, 272–279. [Google Scholar] [CrossRef]
- Michel, M.C.; Ochodnicky, P.; Summers, R.J. Tissue functions mediated by β3-adrenoceptors-findings and challenges. Naunyn Schmiedebergs Arch. Pharmacol. 2010, 382, 103–108. [Google Scholar] [CrossRef]
- Arch, J.R. Challenges in β3-adrenoceptor agonist drug development. Ther. Adv. Endocrinol. Metab. 2011, 2, 59–64. [Google Scholar] [CrossRef] [Green Version]
- Negreș, S.; Chiriță, C.; Arsene, A.L.; Margină, D.; Moroșan, E.; Zbârcea, C.E. New potential β-3 adrenergic agonists with β-phenylethylamine structure, synthesized for the treatment of dyslipidemia and obesity. In Adiposity: Epidemiology and Treatment Modalities; IntechOpen: London, UK, 2017. [Google Scholar]
- Gauthier, C.; Tavernier, G.; Charpentier, F.; Langin, D.; Le Marec, H. Functional β3-adrenoceptor in the human heart. J. Clin. Investig. 1996, 98, 556–562. [Google Scholar] [CrossRef]
- Berkowitz, D.E.; Nardone, N.A.; Smiley, R.M.; Price, D.T.; Kreutter, D.K.; Fremeau, R.T.; Schwinn, D.A. Distribution of β3-adrenoceptor mrna in human tissues. Eur. J. Pharmacol. 1995, 289, 223–228. [Google Scholar] [CrossRef]
- Gauthier, C.; Tavernier, G.; Trochu, J.N.; Leblais, V.; Laurent, K.; Langin, D.; Escande, D.; Le Marec, H. Interspecies differences in the cardiac negative inotropic effects of β3-adrenoceptor agonists. J. Pharmacol. Exp. Ther. 1999, 290, 687–693. [Google Scholar]
- Tavernier, G.; Toumaniantz, G.; Erfanian, M.; Heymann, M.F.; Laurent, K.; Langin, D.; Gauthier, C. β3-adrenergic stimulation produces a decrease of cardiac contractility ex vivo in mice overexpressing the human β3-adrenergic receptor. Cardiovasc. Res. 2003, 59, 288–296. [Google Scholar] [CrossRef]
- Gauthier, C.; Leblais, V.; Kobzik, L.; Trochu, J.N.; Khandoudi, N.; Bril, A.; Balligand, J.L.; Le Marec, H. The negative inotropic effect of β3-adrenoceptor stimulation is mediated by activation of a nitric oxide synthase pathway in human ventricle. J. Clin. Invest. 1998, 102, 1377–1384. [Google Scholar] [CrossRef]
- Han, X.; Kobzik, L.; Balligand, J.L.; Kelly, R.A.; Smith, T.W. Nitric oxide synthase (nos3)-mediated cholinergic modulation of Ca2+ current in adult rabbit atrioventricular nodal cells. Circ. Res. 1996, 78, 998–1008. [Google Scholar] [CrossRef] [PubMed]
- Kou, R.; Michel, T. Epinephrine regulation of the endothelial nitric-oxide synthase roles of rac1 and β3-adrenergic receptors in endothelial no signaling. J. Biol. Chem. 2007, 282, 32719–32729. [Google Scholar] [CrossRef] [PubMed]
- Salie, R.; Alsalhin, A.K.H.; Marais, E.; Lochner, A. Cardioprotective effects of β3-adrenergic receptor (β3-ar) pre-, per-, and post-treatment in ischemia-reperfusion. In Cardiovascular Drugs and Therapy; Springer: Berlin, Germany, 2019. [Google Scholar]
- Idigo, W.O.; Reilly, S.; Zhang, M.H.; Zhang, Y.H.; Jayaram, R.; Carnicer, R.; Crabtree, M.J.; Balligand, J.L.; Casadei, B. Regulation of endothelial nitric-oxide synthase (NOS) S-glutathionylation by neuronal NOS: Evidence of a functional interaction between myocardial constitutive NOS isoforms. J. Biol. Chem. 2012, 287, 43665–43673. [Google Scholar] [CrossRef] [PubMed]
- Watts, V.L.; Sepulveda, F.M.; Cingolani, O.H.; Ho, A.S.; Niu, X.; Kim, R.; Miller, K.L.; Vandegaer, K.; Bedja, D.; Gabrielson, K.L.; et al. Anti-hypertrophic and anti-oxidant effect of β3-adrenergic stimulation in myocytes requires differential neuronal nos phosphorylation. J. Mol. Cell Cardiol. 2013, 62, 8–17. [Google Scholar] [CrossRef] [PubMed]
- Gauthier, C.; Langin, D.; Balligand, J.L. Beta3-adrenoceptors in the cardiovascular system. Trends Pharmacol. Sci. 2000, 21, 426–431. [Google Scholar] [CrossRef]
- Angelone, T.; Filice, E.; Quintieri, A.M.; Imbrogno, S.; Recchia, A.; Pulera, E.; Mannarino, C.; Pellegrino, D.; Cerra, M.C. β3-adrenoceptors modulate left ventricular relaxation in the rat heart via the no-cgmp-pkg pathway. Acta Physiol. 2008, 193, 229–239. [Google Scholar] [CrossRef]
- Skeberdis, V.A.; Gendviliene, V.; Zablockaite, D.; Treinys, R.; Macianskiene, R.; Bogdelis, A.; Jurevicius, J.; Fischmeister, R. β3-adrenergic receptor activation increases human atrial tissue contractility and stimulates the L-type Ca2+ current. J. Clin. Investig. 2008, 118, 3219–3227. [Google Scholar] [CrossRef]
- Christ, T.; Molenaar, P.; Klenowski, P.M.; Ravens, U.; Kaumann, A.J. Human atrial β(1L)-adrenoceptor but not β3-adrenoceptor activation increases force and Ca2+ current at physiological temperature. Br. J. Pharmacol. 2011, 162, 823–839. [Google Scholar] [CrossRef] [PubMed]
- Treinys, R.; Zablockaite, D.; Gendviliene, V.; Jurevicius, J.; Skeberdis, V.A. β3-adrenergic regulation of L-type Ca2+ current and force of contraction in human ventricle. J. Membr. Biol. 2014, 247, 309–318. [Google Scholar] [CrossRef] [PubMed]
- Arioglu-Inan, E.; Kayki-Mutlu, G.; Michel, M.C. Cardiac β3-adrenoceptors—A role in human pathophysiology? Br. J. Pharmacol. 2019. [Google Scholar] [CrossRef]
- Mo, W.; Michel, M.C.; Lee, X.W.; Kaumann, A.J.; Molenaar, P. The β3-adrenoceptor agonist mirabegron increases human atrial force through β1-adrenoceptors: An indirect mechanism? Br. J. Pharmacol. 2017, 174, 2706–2715. [Google Scholar] [CrossRef] [PubMed]
- Balligand, J.L. Cardiac salvage by tweaking with β-3-adrenergic receptors. Cardiovasc. Res. 2016, 111, 128–133. [Google Scholar] [CrossRef]
- Hermida, N.; Michel, L.; Esfahani, H.; Dubois-Deruy, E.; Hammond, J.; Bouzin, C.; Markl, A.; Colin, H.; Steenbergen, A.V.; De Meester, C.; et al. Cardiac myocyte β3-adrenergic receptors prevent myocardial fibrosis by modulating oxidant stress-dependent paracrine signaling. Eur. Heart J. 2018, 39, 888–898. [Google Scholar] [CrossRef]
- Dincer, U.D.; Bidasee, K.R.; Guner, S.; Tay, A.; Ozcelikay, A.T.; Altan, V.M. The effect of diabetes on expression of β1-, β2-, and β3-adrenoreceptors in rat hearts. Diabetes 2001, 50, 455–461. [Google Scholar] [CrossRef]
- Moniotte, S.; Kobzik, L.; Feron, O.; Trochu, J.N.; Gauthier, C.; Balligand, J.L. Upregulation of β3-adrenoceptors and altered contractile response to inotropic amines in human failing myocardium. Circulation 2001, 103, 1649–1655. [Google Scholar] [CrossRef] [PubMed]
- Moniotte, S.; Belge, C.; Sekkali, B.; Massion, P.B.; Rozec, B.; Dessy, C.; Balligand, J.L. Sepsis is associated with an upregulation of functional β3 adrenoceptors in the myocardium. Eur. J. Heart Fail. 2007, 9, 1163–1171. [Google Scholar] [CrossRef] [Green Version]
- Aragon, J.P.; Condit, M.E.; Bhushan, S.; Predmore, B.L.; Patel, S.S.; Grinsfelder, D.B.; Gundewar, S.; Jha, S.; Calvert, J.W.; Barouch, L.A.; et al. β3-adrenoreceptor stimulation ameliorates myocardial ischemia-reperfusion injury via endothelial nitric oxide synthase and neuronal nitric oxide synthase activation. J. Am. Coll. Cardiol. 2011, 58, 2683–2691. [Google Scholar] [CrossRef]
- Cannavo, A.; Koch, W.J. Targeting β3-adrenergic receptors in the heart: Selective agonism and beta-blockade. J. Cardiovasc. Pharmacol. 2017, 69, 71–78. [Google Scholar] [PubMed]
- Belge, C.; Hammond, J.; Dubois-Deruy, E.; Manoury, B.; Hamelet, J.; Beauloye, C.; Markl, A.; Pouleur, A.C.; Bertrand, L.; Esfahani, H.; et al. Enhanced expression of β3-adrenoceptors in cardiac myocytes attenuates neurohormone-induced hypertrophic remodeling through nitric oxide synthase. Circulation 2014, 129, 451–462. [Google Scholar] [CrossRef]
- Cannavo, A.; Liccardo, D.; Lymperopoulos, A.; Gambino, G.; D’Amico, M.L.; Rengo, F.; Koch, W.J.; Leosco, D.; Ferrara, N.; Rengo, G. Beta adrenergic receptor kinase c-terminal peptide gene-therapy improves β2-adrenergic receptor-dependent neoangiogenesis after hindlimb ischemia. J. Pharmacol. Exp. Ther. 2016, 356, 503–513. [Google Scholar] [CrossRef] [PubMed]
- Sorrentino, S.A.; Doerries, C.; Manes, C.; Speer, T.; Dessy, C.; Lobysheva, I.; Mohmand, W.; Akbar, R.; Bahlmann, F.; Besler, C.; et al. Nebivolol exerts beneficial effects on endothelial function, early endothelial progenitor cells, myocardial neovascularization, and left ventricular dysfunction early after myocardial infarction beyond conventional β1-blockade. J. Am. Coll. Cardiol. 2011, 57, 601–611. [Google Scholar] [CrossRef] [PubMed]
- Nisoli, E.; Tonello, C.; Benarese, M.; Carruba, M.O. Rat Frontal Cortex β1-Adrenoceptors Are Activated by the β3-Adrenoceptor Agonists SR 58611A and SR 58878A but Not by BRL 37344 or ICI 215,001. J. Neurochem. 1995, 65, 1580–1587. [Google Scholar] [CrossRef] [PubMed]
- Ressler, K.J.; Nemeroff, C.B. Role of serotonergic and noradrenergic systems in the pathophysiology of depression and anxiety disorders. Depress. Anxiety 2000, 12 (Suppl. 1), 2–19. [Google Scholar] [CrossRef]
- Claustre, Y.; Leonetti, M.; Santucci, V.; Bougault, I.; Desvignes, C.; Rouquier, L.; Aubin, N.; Keane, P.; Busch, S.; Chen, Y.; et al. Effects of the β3-adrenoceptor (Adrb3) agonist SR58611A (amibegron) on serotonergic and noradrenergic transmission in the rodent: relevance to its antidepressant/anxiolytic-like profile. Neuroscience 2008, 156, 353–364. [Google Scholar] [CrossRef]
- Tamburella, A.; Micale, V.; Leggio, G.M.; Drago, F. The β3 adrenoceptor agonist, amibegron (SR58611A) counteracts stress-induced behavioral and neurochemical changes. Eur. Neuropsychopharmacol. 2010, 20, 704–713. [Google Scholar] [CrossRef]
- Tanyeri, P.; Buyukokuroglu, M.E.; Mutlu, O.; Ulak, G.; Akar, F.Y.; Celikyurt, I.K.; Erden, B.F. Evidence that the anxiolytic-like effects of the β3 receptor agonist amibegron involve serotoninergic receptor activity. Pharmacol. Biochem. Behav. 2013, 110, 27–32. [Google Scholar] [CrossRef]
- Jhaveri, D.J.; Mackay, E.W.; Hamlin, A.S.; Marathe, S.V.; Nandam, L.S.; Vaidya, V.A.; Bartlett, P.F. Norepinephrine directly activates adult hippocampal precursors via β3-adrenergic receptors. J. Neurosci. 2010, 30, 2795–2806. [Google Scholar] [CrossRef]
- Church, T.W.; Brown, J.T.; Marrion, N.V. β3-adrenergic receptor-dependent modulation of the medium afterhyperpolarization in rat hippocampal ca1 pyramidal neurons. J. Neurophysiol. 2019, 121, 773–784. [Google Scholar] [CrossRef]
- Gibbs, M.E.; Hutchinson, D.S.; Summers, R.J. Role of beta-adrenoceptors in memory consolidation: β3-adrenoceptors act on glucose uptake and β2-adrenoceptors on glycogenolysis. Neuropsychopharmacology 2008, 33, 2384–2397. [Google Scholar] [CrossRef]
- Souza-Braga, P.; Lorena, F.B.; Nascimento, B.P.P.; Marcelino, C.P.; Ravache, T.T.; Ricci, E.; Bernardi, M.M.; Ribeiro, M.O. Adrenergic receptor β3 is involved in the memory consolidation process in mice. Braz. J. Med. Biol. Res. 2018, 51, e7564. [Google Scholar] [CrossRef]
- Kanno, T.; Yaguchi, T.; Nishizaki, T. Noradrenaline stimulates atp release from drg neurons by targeting β3 adrenoceptors as a factor of neuropathic pain. J. Cell Physiol. 2010, 224, 345–351. [Google Scholar] [CrossRef]
- Valdes, A.M.; Abhishek, A.; Muir, K.; Zhang, W.; Maciewicz, R.A.; Doherty, M. Association of β-blocker use with less prevalent joint pain and lower opioid requirement in people with osteoarthritis. Arthritis Care Res. 2017, 69, 1076–1081. [Google Scholar] [CrossRef]
- Mori, A.; Miwa, T.; Sakamoto, K.; Nakahara, T.; Ishii, K. Pharmacological evidence for the presence of functional β3-adrenoceptors in rat retinal blood vessels. Naunyn Schmiedebergs Arch. Pharmacol. 2010, 382, 119–126. [Google Scholar] [CrossRef]
- Steinle, J.J.; Booz, G.W.; Meininger, C.J.; Day, J.N.; Granger, H.J. β3-adrenergic receptors regulate retinal endothelial cell migration and proliferation. J. Biol. Chem. 2003, 278, 20681–20686. [Google Scholar] [CrossRef]
- Steinle, J.J.; Zamora, D.O.; Rosenbaum, J.T.; Granger, H.J. β3-adrenergic receptors mediate choroidal endothelial cell invasion, proliferation, and cell elongation. Exp. Eye Res. 2005, 80, 83–91. [Google Scholar] [CrossRef] [PubMed]
- Ristori, C.; Filippi, L.; Dal Monte, M.; Martini, D.; Cammalleri, M.; Fortunato, P.; la Marca, G.; Fiorini, P.; Bagnoli, P. Role of the adrenergic system in a mouse model of oxygen-induced retinopathy: Antiangiogenic effects of β-adrenoreceptor blockade. Invest. Ophthalmol. Vis. Sci. 2011, 52, 155–170. [Google Scholar] [CrossRef]
- Dal Monte, M.; Filippi, L.; Bagnoli, P. β3-adrenergic receptors modulate vascular endothelial growth factor release in response to hypoxia through the nitric oxide pathway in mouse retinal explants. Naunyn Schmiedebergs Arch. Pharmacol. 2013, 386, 269–278. [Google Scholar] [CrossRef]
- Steinle, J.J. Sympathetic neurotransmission modulates expression of inflammatory markers in the rat retina. Exp. Eye Res. 2007, 84, 118–125. [Google Scholar] [CrossRef]
- Chisholm, K.M.; Chang, K.W.; Truong, M.T.; Kwok, S.; West, R.B.; Heerema-McKenney, A.E. Beta-adrenergic receptor expression in vascular tumors. Mod. Pathol. 2012, 25, 1446–1451. [Google Scholar] [CrossRef]
- Lamkin, D.M.; Sloan, E.K.; Patel, A.J.; Chiang, B.S.; Pimentel, M.A.; Ma, J.C.; Arevalo, J.M.; Morizono, K.; Cole, S.W. Chronic stress enhances progression of acute lymphoblastic leukemia via beta-adrenergic signaling. Brain Behav. Immun. 2012, 26, 635–641. [Google Scholar] [CrossRef] [PubMed]
- Perrone, M.G.; Notarnicola, M.; Caruso, M.G.; Tutino, V.; Scilimati, A. Upregulation of beta3-adrenergic receptor mrna in human colon cancer: A preliminary study. Oncology 2008, 75, 224–229. [Google Scholar] [CrossRef] [PubMed]
- Calvani, M.; Pelon, F.; Comito, G.; Taddei, M.L.; Moretti, S.; Innocenti, S.; Nassini, R.; Gerlini, G.; Borgognoni, L.; Bambi, F.; et al. Norepinephrine promotes tumor microenvironment reactivity through β3-adrenoreceptors during melanoma progression. Oncotarget 2015, 6, 4615–4632. [Google Scholar] [CrossRef]
- Montoya, A.; Amaya, C.N.; Belmont, A.; Diab, N.; Trevino, R.; Villanueva, G.; Rains, S.; Sanchez, L.A.; Badri, N.; Otoukesh, S.; et al. Use of non-selective beta-blockers is associated with decreased tumor proliferative indices in early stage breast cancer. Oncotarget 2017, 8, 6446–6460. [Google Scholar] [CrossRef] [PubMed]
- Daly, C.J.; McGrath, J.C. Previously unsuspected widespread cellular and tissue distribution of β-adrenoceptors and its relevance to drug action. Trends Pharmacol. Sci. 2011, 32, 219–226. [Google Scholar] [CrossRef] [PubMed]
- Dal Monte, M.; Casini, G.; Filippi, L.; Nicchia, G.P.; Svelto, M.; Bagnoli, P. Functional involvement of β3-adrenergic receptors in melanoma growth and vascularization. J. Mol. Med. 2013, 91, 1407–1419. [Google Scholar] [CrossRef] [PubMed]
- Calvani, M.; Cavallini, L.; Tondo, A.; Spinelli, V.; Ricci, L.; Pasha, A.; Bruno, G.; Buonvicino, D.; Bigagli, E.; Vignoli, M.; et al. β3-adrenoreceptors control mitochondrial dormancy in melanoma and embryonic stem cells. Oxid. Med. Cell Longev. 2018, 2018, 6816508. [Google Scholar] [CrossRef] [PubMed]
- Calvani, M.; Bruno, G.; Dal Monte, M.; Nassini, R.; Fontani, F.; Casini, A.; Cavallini, L.; Becatti, M.; Bianchini, F.; De Logu, F.; et al. β3-adrenoceptor as a potential immuno-suppressor agent in melanoma. Br. J. Pharmacol. 2019. [Google Scholar] [CrossRef]
- Dal Monte, M.; Calvani, M.; Cammalleri, M.; Favre, C.; Filippi, L.; Bagnoli, P. Beta-adrenoceptors as drug targets in melanoma: Novel preclinical evidence for a role of β3-adrenoceptors. Br. J. Pharmacol. 2018. [Google Scholar] [CrossRef]
- Leineweber, K.; Buscher, R.; Bruck, H.; Brodde, O.E. Beta-adrenoceptor polymorphisms. Naunyn Schmiedebergs Arch. Pharmacol. 2004, 369, 1–22. [Google Scholar] [CrossRef]
- Clement, K.; Vaisse, C.; Manning, B.S.; Basdevant, A.; Guy-Grand, B.; Ruiz, J.; Silver, K.D.; Shuldiner, A.R.; Froguel, P.; Strosberg, A.D. Genetic variation in the β3-adrenergic receptor and an increased capacity to gain weight in patients with morbid obesity. N. Engl. J. Med. 1995, 333, 352–354. [Google Scholar] [CrossRef]
- Walston, J.; Silver, K.; Bogardus, C.; Knowler, W.C.; Celi, F.S.; Austin, S.; Manning, B.; Strosberg, A.D.; Stern, M.P.; Raben, N.; et al. Time of onset of non-insulin-dependent diabetes mellitus and genetic variation in the β3-adrenergic-receptor gene. N. Engl. J. Med. 1995, 333, 343–347. [Google Scholar] [CrossRef]
- Hoffstedt, J.; Poirier, O.; Thorne, A.; Lonnqvist, F.; Herrmann, S.M.; Cambien, F.; Arner, P. Polymorphism of the human β3-adrenoceptor gene forms a well-conserved haplotype that is associated with moderate obesity and altered receptor function. Diabetes 1999, 48, 203–205. [Google Scholar] [CrossRef] [PubMed]
- Ryuk, J.A.; Zhang, X.; Ko, B.S.; Daily, J.W.; Park, S. Association of β3-adrenergic receptor rs4994 polymorphisms with the risk of type 2 diabetes: A systematic review and meta-analysis. Diabetes Res. Clin. Pract. 2017, 129, 86–96. [Google Scholar] [CrossRef] [PubMed]
- Kurokawa, N.; Young, E.H.; Oka, Y.; Satoh, H.; Wareham, N.J.; Sandhu, M.S.; Loos, R.J. The ADRB3 Trp64Arg variant and BMI: a meta-analysis of 44 833 individuals. Int J. Obes. 2008, 32, 1240–1249. [Google Scholar] [CrossRef] [Green Version]
- Szendrei, B.; Gonzalez-Lamuno, D.; Amigo, T.; Wang, G.; Pitsiladis, Y.; Benito, P.J.; Gomez-Candela, C.; Calderon, F.J.; Cupeiro, R.; Group, P.S. Influence of ADRB2 Gln27Glu and ADRB3 Trp64Arg polymorphisms on body weight and body composition changes after a controlled weight-loss intervention. Appl. Physiol. Nutr. Metab. 2016, 41, 307–314. [Google Scholar] [CrossRef] [Green Version]
- Daghestani, M.; Daghestani, M.; Daghistani, M.; Eldali, A.; Hassan, Z.K.; Elamin, M.H.; Warsy, A. ADRB3 polymorphism rs4994 (Trp64Arg) associates significantly with bodyweight elevation and dyslipidaemias in Saudis but not rs1801253 (Arg389Gly) polymorphism in ARDB1. Lipids Health Dis. 2018, 17, 58. [Google Scholar] [CrossRef]
- Qu, H.C.; Zhang, W.; Liu, Y.L.; Wang, P. Association between polymorphism of β3-adrenoceptor gene and overactive bladder: A meta-analysis. Genet. Mol. Res. 2015, 14, 2495–2501. [Google Scholar] [CrossRef]
- Allison, D.B.; Heo, M.; Faith, M.S.; Pietrobelli, A. Meta-analysis of the association of the Trp64Arg polymorphism in the β3 adrenergic receptor with body mass index. Int. J. Obes. Relat. Metab. Disord 1998, 22, 559–566. [Google Scholar] [CrossRef] [Green Version]
- Rawson, E.S.; Nolan, A.; Silver, K.; Shuldiner, A.R.; Poehlman, E.T. No effect of the Trp64Arg β3-adrenoceptor gene variant on weight loss, body composition, or energy expenditure in obese, caucasian postmenopausal women. Metabolism 2002, 51, 801–805. [Google Scholar] [CrossRef]
- Teitsma, C.A.; de la Rosette, J.J.; Michel, M.C. Are polymorphisms of the β3-adrenoceptor gene associated with an altered bladder function? Neurourol. Urodyn. 2013, 32, 276–280. [Google Scholar] [CrossRef]
- Honda, K.; Nomiya, M.; Shishido, K.; Yoshimura, Y.; Yamaguchi, O. Mutation of β3-adrenoceptor gene: A genetic marker for overactive bladder. In Neurourology and Urodynamics; John Wiley & Sons: Hoboken, NJ, USA, 2006. [Google Scholar]
- Fatima, T.; Altaf, S.; Phipps-Green, A.; Topless, R.; Flynn, T.J.; Stamp, L.K.; Dalbeth, N.; Merriman, T.R. Association analysis of the β-3 adrenergic receptor Trp64Arg (RS4994) polymorphism with urate and gout. Rheumatol. Int. 2016, 36, 255–261. [Google Scholar] [CrossRef] [PubMed]
- Babol, K.; Przybylowska, K.; Lukaszek, M.; Pertynski, T.; Blasiak, J. An association between the Trp64Arg polymorphism in the β3-adrenergic receptor gene and endometrial cancer and obesity. J. Exp. Clin. Cancer Res. 2004, 23, 669–674. [Google Scholar] [PubMed]
- Huang, X.E.; Hamajima, N.; Saito, T.; Matsuo, K.; Mizutani, M.; Iwata, H.; Iwase, T.; Miura, S.; Mizuno, T.; Tokudome, S.; et al. Possible association of β2- and β3-adrenergic receptor gene polymorphisms with susceptibility to breast cancer. Breast Cancer Res. 2001, 3, 264–269. [Google Scholar] [CrossRef] [PubMed]
- Tan, W.; Gao, M.; Liu, N.; Zhang, G.; Xu, T.; Cui, W. Body mass index and risk of gallbladder cancer: Systematic review and meta-analysis of observational studies. Nutrients 2015, 7, 8321–8334. [Google Scholar] [CrossRef] [PubMed]
- Bardou, M.; Loustalot, C.; Cortijo, J.; Simon, B.; Naline, E.; Dumas, M.; Esteve, S.; Croci, T.; Chalon, P.; Frydman, R.; et al. Functional, biochemical and molecular biological evidence for a possible β3-adrenoceptor in human near-term myometrium. Br. J. Pharmacol. 2000, 130, 1960–1966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rouget, C.; Bardou, M.; Breuiller-Fouche, M.; Loustalot, C.; Qi, H.; Naline, E.; Croci, T.; Cabrol, D.; Advenier, C.; Leroy, M.J. β3-adrenoceptor is the predominant beta-adrenoceptor subtype in human myometrium and its expression is up-regulated in pregnancy. J. Clin. Endocrinol. Metab. 2005, 90, 1644–1650. [Google Scholar] [CrossRef]
- Hadi, T.; Douhard, R.; Dias, A.M.M.; Wendremaire, M.; Pezze, M.; Bardou, M.; Sagot, P.; Garrido, C.; Lirussi, F. β3 adrenergic receptor stimulation in human macrophages inhibits nadphoxidase activity and induces catalase expression via ppargamma activation. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 1769–1784. [Google Scholar] [CrossRef] [PubMed]
Compound | Manufacturer/Sponsor | Therapeutic Indications | Status | References |
---|---|---|---|---|
Mirabegron (YM178) | Astellas Pharma | Overactive bladder syndrome | FDA-approved | [38] |
Amibegron (SR58611A) | Sanofi | Antidepressant, antianxiolytic | Phase II and III (2005–2008) then discontinued 1 | [39,40,41]; Clinicaltrials.gov NCT00252330 |
Solabegron (GW427353) | Glaxosmithkline | Overactive bladder syndrome | Phase II (ongoing) | [42,43]; Clinicaltrials.gov NCT03475706 |
Ritobegron (KUC-7483) | Kissei Pharmaceuticals Co. Ltd. | Overactive bladder syndrome | Phase III Discontinued 1 | [42,43,44,45]; Clinicaltrials.gov NCT01003405 |
Vibegron 2 | Kyorin Pharmaceutical Co., Ltd. and Kissei Pharmaceutical Co., Ltd. | Overactive bladder syndrome | Approved (Japan) | [46,47] |
Tissue | Protein | mRNA | Function | Downstream Signaling (Mediators) | Potential Therapeutic Indications | Agonists Currently Developing | Status | |
---|---|---|---|---|---|---|---|---|
Urinary System | Bladder | Yes | Yes | Bladder relaxation | Gs (cAMP/PKA) | Treatment of Overactive bladder syndrome | Mirabegron Solabegron Vibegron | Approved Phase II Phase III |
Kidney | Yes | Yes | Water and solute reabsorption | Gs (cAMP/PKA) | - | - | - | |
Central Nervous System | Brain | No | Yes | Increase of trp and 5-HT levels | - | Antidepressants | Amibegron (SR58611A) | Discontinued |
Retina | Yes | Yes | Endothelial cell growth, migration, and elongation | MAPK cascade, metalloproteinases 2/9, PKG | - | - | - | |
Adipose tissue | Yes | Yes | Lipolysis, thermogenesis | Gs (cAMP/PKA) and Gi (NO) | Anti-obesity, anti-diabetic | Beta-phenylethylamine | Pre-clinical | |
Myocardium | Yes | Yes | Lowering/Increasing of cardiac contractility | Gi (NO)/ Gs (cAMP/PKA) | Treatment of heart failure | Mirabegron (repurposing study) | Phase II | |
Myometrium | Yes | Yes | Relaxation of myometrial contractions | cAMP | Management of preterm labor | - | - |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Schena, G.; Caplan, M.J. Everything You Always Wanted to Know about β3-AR * (* But Were Afraid to Ask). Cells 2019, 8, 357. https://doi.org/10.3390/cells8040357
Schena G, Caplan MJ. Everything You Always Wanted to Know about β3-AR * (* But Were Afraid to Ask). Cells. 2019; 8(4):357. https://doi.org/10.3390/cells8040357
Chicago/Turabian StyleSchena, Giorgia, and Michael J. Caplan. 2019. "Everything You Always Wanted to Know about β3-AR * (* But Were Afraid to Ask)" Cells 8, no. 4: 357. https://doi.org/10.3390/cells8040357
APA StyleSchena, G., & Caplan, M. J. (2019). Everything You Always Wanted to Know about β3-AR * (* But Were Afraid to Ask). Cells, 8(4), 357. https://doi.org/10.3390/cells8040357