Human Luteinizing Hormone and Chorionic Gonadotropin Display Biased Agonism at the LH and LH/CG Receptors

Laura Riccetti, Romain Yvinec, Danièle Klett, Nathalie Gallay, Yves Combarnous, Eric Reiter, Manuela Simoni, Livio Casarini, Mohammed Akli Ayoub, Laura Riccetti, Romain Yvinec, Danièle Klett, Nathalie Gallay, Yves Combarnous, Eric Reiter, Manuela Simoni, Livio Casarini, Mohammed Akli Ayoub

Abstract

Human luteinizing hormone (LH) and chorionic gonadotropin (hCG) have been considered biologically equivalent because of their structural similarities and their binding to the same receptor; the LH/CGR. However, accumulating evidence suggest that LH/CGR differentially responds to the two hormones triggering differential intracellular signaling and steroidogenesis. The mechanistic basis of such differential responses remains mostly unknown. Here, we compared the abilities of recombinant rhLH and rhCG to elicit cAMP, β-arrestin 2 activation, and steroidogenesis in HEK293 cells and mouse Leydig tumor cells (mLTC-1). For this, BRET and FRET technologies were used allowing quantitative analyses of hormone activities in real-time and in living cells. Our data indicate that rhLH and rhCG differentially promote cell responses mediated by LH/CGR revealing interesting divergences in their potencies, efficacies and kinetics: rhCG was more potent than rhLH in both HEK293 and mLTC-1 cells. Interestingly, partial effects of rhLH were found on β-arrestin recruitment and on progesterone production compared to rhCG. Such a link was further supported by knockdown experiments. These pharmacological differences demonstrate that rhLH and rhCG act as natural biased agonists. The discovery of novel mechanisms associated with gonadotropin-specific action may ultimately help improve and personalize assisted reproduction technologies.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
LH/CGR-mediated cAMP production. cAMP responses induced by rhCG and rhLH in HEK293 cells transiently co-expressing hLH/CGR and CAMYEL sensor (A,B) and in mLTC-1 cells transiently expressing CAMYEL sensor alone (C) or not (D). Cells were stimulated 30 minutes with increasing doses (A,C and D) or immediately with the indicated doses (B) of rhCG and rhLH, before cAMP production was measured by BRET (A,B and C) or HTRF (D). Data are means ± SEM of 3–8 independent experiments. The kinetic curves in panel B are representative of 3 experiments performed in triplicate.
Figure 2
Figure 2
LH/CGR-promoted β-arrestin 2 recruitment and activation in HEK293 cells. HEK293 cells transiently co-expressing either hLH/CGR-Rluc8 and yPET-β-arrestin 2 (A and B) or wildtype hLHR/CGR and Rluc8-β-arrestin 2-RGFP (C) were stimulated 30 minutes with increasing doses (A and D) or immediately with the indicated doses (B) or 0.25 µM (C) of rhCG and rhLH before BRET was measured. Data are means ± SEM of 3–5 independent experiments. The kinetic curves in panel B are representative of 3 experiments performed in triplicate.
Figure 3
Figure 3
LH/CGR-promoted integrated responses in HEK293 and mLTC-1 cells. (A) HEK293 transiently co-transfected with plasmids coding for LH/CGR and pSOM-Luc reporter gene were stimulated 6 hours with increasing doses of rhCG and rhLH before luciferase activity was measured (A). mLTC-1 cells transiently transfected with plasmid coding pSOM-Luc reporter gene were stimulated 6 hours with increasing doses of rhCG and rhLH before luciferase activity was measured (B). For LHR-mediated steroid production, mLTC-1 cells were stimulated 3 hours with increasing doses of rhCG and rhLH and the supernatant levels of progesterone (C) and testosterone (D) were measured by ELISA or HTRF, respectively. Data are means ± SEM of 3–5 independent experiments performed in single or duplicate.
Figure 4
Figure 4
Implication of β-arrestins in LH/CGR-promoted steroid production in mLTC-1 cells. mLTC-1 cells were transiently transfected with either control, β-arrestin 1 or β-arrestin 2 siRNAs. Then, cells were stimulated 3 hours with increasing doses of rhCG (A,C and E) and rhLH (B,D and F) before progesterone (A and B), testosterone (C and D) and cAMP (E and F) productions were measured. The maximal rhCG-mediated responses obtained at 10 nM with control siRNA was taken as 100%. Data are means ± SEM of 3 independent experiments performed in duplicate.

References

    1. Arey BJ, Lopez FJ. Are circulating gonadotropin isoforms naturally occurring biased agonists? Basic and therapeutic implications. Rev Endocr Metab Disord. 2011;12:275–288. doi: 10.1007/s11154-011-9188-y.
    1. Bousfield GR, Dias JA. Synthesis and secretion of gonadotropins including structure-function correlates. Rev Endocr Metab Disord. 2011;12:289–302. doi: 10.1007/s11154-011-9191-3.
    1. Leão RB, Esteves SC. Gonadotropin therapy in assisted reproduction: an evolutionary perspective from biologics to biotech. Clinics (Sao Paulo) 2014;69:279–293. doi: 10.6061/clinics/2014(04)10.
    1. Ascoli M, Fanelli F, Segaloff DL. The lutropin/choriogonadotropin receptor, a 2002 perspective. Endocrine reviews. 2002;23:141–174. doi: 10.1210/edrv.23.2.0462.
    1. Choi J, Smitz J. Luteinizing hormone and human chorionic gonadotropin: distinguishing unique physiologic roles. Gynecol Endocrinol. 2014;30:174–181. doi: 10.3109/09513590.2013.859670.
    1. Gudermann T, Birnbaumer M, Birnbaumer L. Evidence for dual coupling of the murine luteinizing hormone receptor to adenylyl cyclase and phosphoinositide breakdown and Ca2+ mobilization. Studies with the cloned murine luteinizing hormone receptor expressed in L cells. J Biol Chem. 1992;267:4479–4488.
    1. Gudermann T, Nichols C, Levy FO, Birnbaumer M, Birnbaumer L. Ca2+ mobilization by the LH receptor expressed in Xenopus oocytes independent of 3′,5′-cyclic adenosine monophosphate formation: evidence for parallel activation of two signaling pathways. Mol Endocrinol. 1992;6:272–278.
    1. Ulloa-Aguirre A, Crepieux P, Poupon A, Maurel MC, Reiter E. Novel pathways in gonadotropin receptor signaling and biased agonism. Rev Endocr Metab Disord. 2011;12:259–274. doi: 10.1007/s11154-011-9176-2.
    1. Nakamura K, Lazari MF, Li S, Korgaonkar C, Ascoli M. Role of the rate of internalization of the agonist-receptor complex on the agonist-induced down-regulation of the lutropin/choriogonadotropin receptor. Molecular endocrinology. 1999;13:1295–1304. doi: 10.1210/mend.13.8.0331.
    1. Ayoub MA, et al. Assessing Gonadotropin Receptor Function by Resonance Energy Transfer-Based Assays. Front Endocrinol (Lausanne) 2015;6:130.
    1. Ayoub MA, et al. Profiling of FSHR negative allosteric modulators on LH/CGR reveals biased antagonism with implications in steroidogenesis. Molecular and cellular endocrinology. 2016;436:10–22. doi: 10.1016/j.mce.2016.07.013.
    1. Lefkowitz RJ, Shenoy SK. Transduction of receptor signals by beta-arrestins. Science. 2005;308:512–517. doi: 10.1126/science.1109237.
    1. Reiter E, Lefkowitz RJ. GRKs and beta-arrestins: roles in receptor silencing, trafficking and signaling. Trends Endocrinol Metab. 2006;17:159–165. doi: 10.1016/j.tem.2006.03.008.
    1. Reiter E, Ahn S, Shukla AK, Lefkowitz RJ. Molecular mechanism of beta-arrestin-biased agonism at seven-transmembrane receptors. Annu Rev Pharmacol Toxicol. 2012;52:179–197. doi: 10.1146/annurev.pharmtox.010909.105800.
    1. Casarini L, Reiter E, Simoni M. beta-arrestins regulate gonadotropin receptor-mediated cell proliferation and apoptosis by controlling different FSHR or LHCGR intracellular signaling in the hGL5 cell line. Molecular and cellular endocrinology. 2016;437:11–21. doi: 10.1016/j.mce.2016.08.005.
    1. Galet C, Ascoli M. Arrestin-3 is essential for the activation of Fyn by the luteinizing hormone receptor (LHR) in MA-10 cells. Cellular signalling. 2008;20:1822–1829. doi: 10.1016/j.cellsig.2008.06.005.
    1. Krishnamurthy H, Galet C, Ascoli M. The association of arrestin-3 with the follitropin receptor depends on receptor activation and phosphorylation. Molecular and cellular endocrinology. 2003;204:127–140. doi: 10.1016/S0303-7207(03)00088-1.
    1. Neill JD, Duck LW, Musgrove LC, Sellers JC. Potential regulatory roles for G protein-coupled receptor kinases and beta-arrestins in gonadotropin-releasing hormone receptor signaling. Endocrinology. 1998;139:1781–1788.
    1. Wehbi V, et al. Partially deglycosylated equine LH preferentially activates beta-arrestin-dependent signaling at the follicle-stimulating hormone receptor. Molecular endocrinology. 2010;24:561–573. doi: 10.1210/me.2009-0347.
    1. Tranchant T, et al. Preferential beta-arrestin signalling at low receptor density revealed by functional characterization of the human FSH receptor A189 V mutation. Molecular and cellular endocrinology. 2011;331:109–118. doi: 10.1016/j.mce.2010.08.016.
    1. Kara E, et al. A phosphorylation cluster of five serine and threonine residues in the C-terminus of the follicle-stimulating hormone receptor is important for desensitization but not for beta-arrestin-mediated ERK activation. Molecular endocrinology. 2006;20:3014–3026. doi: 10.1210/me.2006-0098.
    1. Khan-Sabir, N., Beshay, V. E. & Carr, B. R. In Endotext Vol. chapter 3 (2008).
    1. Huhtaniemi IT, Catt KJ. Differential binding affinities of rat testis luteinizing hormone (LH) receptors for human chorionic gonadotropin, human LH, and ovine LH. Endocrinology. 1981;108:1931–1938. doi: 10.1210/endo-108-5-1931.
    1. Galet C, Ascoli M. The differential binding affinities of the luteinizing hormone (LH)/choriogonadotropin receptor for LH and choriogonadotropin are dictated by different extracellular domain residues. Mol Endocrinol. 2005;19:1263–1276. doi: 10.1210/me.2004-0410.
    1. Grzesik P, et al. Differences in Signal Activation by LH and hCG are Mediated by the LH/CG Receptor’s Extracellular Hinge Region. Front Endocrinol (Lausanne) 2015;6:140.
    1. Grzesik P, et al. Differences between lutropin-mediated and choriogonadotropin-mediated receptor activation. FEBS J. 2014;281:1479–1492. doi: 10.1111/febs.12718.
    1. Choi J, Smitz J. Luteinizing hormone and human chorionic gonadotropin: origins of difference. Molecular and cellular endocrinology. 2014;383:203–213. doi: 10.1016/j.mce.2013.12.009.
    1. Galandrin S, Oligny-Longpre G, Bouvier M. The evasive nature of drug efficacy: implications for drug discovery. Trends Pharmacol Sci. 2007;28:423–430. doi: 10.1016/j.tips.2007.06.005.
    1. Kenakin T. Functional selectivity through protean and biased agonism: who steers the ship? Mol Pharmacol. 2007;72:1393–1401. doi: 10.1124/mol.107.040352.
    1. Landomiel F, et al. Biased signalling in follicle stimulating hormone action. Molecular and cellular endocrinology. 2014;382:452–459. doi: 10.1016/j.mce.2013.09.035.
    1. Gromoll J, Eiholzer U, Nieschlag E, Simoni M. Male hypogonadism caused by homozygous deletion of exon 10 of the luteinizing hormone (LH) receptor: differential action of human chorionic gonadotropin and LH. J Clin Endocrinol Metab. 2000;85:2281–2286. doi: 10.1210/jcem.85.6.6636.
    1. Muller T, Gromoll J, Simoni M. Absence of exon 10 of the human luteinizing hormone (LH) receptor impairs LH, but not human chorionic gonadotropin action. J Clin Endocrinol Metab. 2003;88:2242–2249. doi: 10.1210/jc.2002-021946.
    1. Casarini L, et al. LH and hCG action on the same receptor results in quantitatively and qualitatively different intracellular signalling. PLoS One. 2012;7:e46682. doi: 10.1371/journal.pone.0046682.
    1. Casarini L, et al. Follicle-stimulating hormone potentiates the steroidogenic activity of chorionic gonadotropin and the anti-apoptotic activity of luteinizing hormone in human granulosa-lutein cells in vitro. Molecular and cellular endocrinology. 2016;422:103–114. doi: 10.1016/j.mce.2015.12.008.
    1. Gupta C, et al. Differential response to sustained stimulation by hCG & LH on goat ovarian granulosa cells. The Indian journal of medical research. 2012;135:331–340. doi: 10.4103/0971-5916.93429.
    1. Klett D, et al. Low reversibility of intracellular cAMP accumulation in mouse Leydig tumor cells (MLTC-1) stimulated by human Luteinizing Hormone (hLH) and Chorionic Gonadotropin (hCG) Molecular and cellular endocrinology. 2016;434:144–153. doi: 10.1016/j.mce.2016.06.028.
    1. Black JW, Leff P. Operational models of pharmacological agonism. Proc R Soc Lond B Biol Sci. 1983;220:141–162. doi: 10.1098/rspb.1983.0093.
    1. van der Westhuizen ET, Breton B, Christopoulos A, Bouvier M. Quantification of ligand bias for clinically relevant beta2-adrenergic receptor ligands: implications for drug taxonomy. Molecular Pharmacology. 2014;85:492–509. doi: 10.1124/mol.113.088880.
    1. Kovacs JJ, et al. Beta-arrestin-mediated localization of smoothened to the primary cilium. Science. 2008;320:1777–1781. doi: 10.1126/science.1157983.
    1. Charest, P. G., Terrillon, S. & Bouvier, M. Monitoring agonist-promoted conformational changes of beta-arrestin in living cells by intramolecular BRET. EMBO Rep6, 334–340, 7400373 [pii] doi:10.1038/sj.embor.7400373 (2005).
    1. Lee MH, et al. The conformational signature of beta-arrestin2 predicts its trafficking and signalling functions. Nature. 2016;531:665–668. doi: 10.1038/nature17154.
    1. Kamal, M. et al. Improved donor/acceptor BRET couples for monitoring beta-arrestin recruitment to G protein-coupled receptors. Biotechnol J, doi:10.1002/biot.200900016 (2009).
    1. Troispoux C, et al. Involvement of G protein-coupled receptor kinases and arrestins in desensitization to follicle-stimulating hormone action. Molecular endocrinology. 1999;13:1599–1614. doi: 10.1210/mend.13.9.0342.
    1. Canepa S, et al. Validation d’une méthode immunoenzymatique pour le dosage de la progesterone dans le plasma des ovins et des bovins. Cahiers techniques de l’INRA. 2008;64:19–30.
    1. Rebois RV. Establishment of gonadotropin-responsive murine leydig tumor cell line. The Journal of cell biology. 1982;94:70–76. doi: 10.1083/jcb.94.1.70.
    1. Miller WL, Auchus RJ. The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocrine Reviews. 2011;32:81–151. doi: 10.1210/er.2010-0013.
    1. Riccetti L, et al. Human LH and hCG stimulate differently the early signalling pathways but result in equal testosterone synthesis in mouse Leydig cells in vitro. Reprod Biol Endocrinol. 2017;15:2. doi: 10.1186/s12958-016-0224-3.
    1. Klein Herenbrink C, et al. The role of kinetic context in apparent biased agonism at GPCRs. Nat Commun. 2016;7:10842. doi: 10.1038/ncomms10842.
    1. Shukla AK, et al. Distinct conformational changes in beta-arrestin report biased agonism at seven-transmembrane receptors. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:9988–9993. doi: 10.1073/pnas.0804246105.
    1. Nuber S, et al. beta-Arrestin biosensors reveal a rapid, receptor-dependent activation/deactivation cycle. Nature. 2016;531:661–664. doi: 10.1038/nature17198.
    1. Kenakin T, Christopoulos A. Signalling bias in new drug discovery: detection, quantification and therapeutic impact. Nature reviews. Drug discovery. 2013;12:205–216. doi: 10.1038/nrd3954.
    1. Luttrell LM. Minireview: More than just a hammer: ligand “bias” and pharmaceutical discovery. Molecular endocrinology. 2014;28:281–294. doi: 10.1210/me.2013-1314.

Source: PubMed

Подписаться