1
|
Jayakody T, Budagoda DK, Mendis K, Dilshan WD, Bethmage D, Dissasekara R, Dawe GS. Biased agonism in peptide-GPCRs: A structural perspective. Pharmacol Ther 2025; 269:108806. [PMID: 39889970 DOI: 10.1016/j.pharmthera.2025.108806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/13/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
G protein-coupled receptors (GPCRs) are dynamic membrane receptors that transduce extracellular signals to the cell interior by forming a ligand-receptor-effector (ternary) complex that functions via allosterism. Peptides constitute an important class of ligands that interact with their cognate GPCRs (peptide-GPCRs) to form the ternary complex. "Biased agonism", a therapeutically relevant phenomenon exhibited by GPCRs owing to their allosteric nature, has also been observed in peptide-GPCRs, leading to the development of selective therapeutics with fewer side effects. In this review, we have focused on the structural basis of signalling bias at peptide-GPCRs of classes A and B, and reviewed the therapeutic relevance of bias at peptide-GPCRs, with the hope of contributing to the discovery of novel biased peptide drugs.
Collapse
Affiliation(s)
- Tharindunee Jayakody
- Department of Chemistry, University of Colombo, P.O. Box 1490, Colombo 00300, Sri Lanka
| | | | - Krishan Mendis
- Department of Chemistry, University of Colombo, P.O. Box 1490, Colombo 00300, Sri Lanka
| | | | - Duvindu Bethmage
- Department of Chemistry, University of Colombo, P.O. Box 1490, Colombo 00300, Sri Lanka
| | - Rashmi Dissasekara
- Department of Chemistry, University of Colombo, P.O. Box 1490, Colombo 00300, Sri Lanka; The Graduate School, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Gavin Stewart Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore; Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
2
|
Ulloa-Aguirre A, Zariñán T, Dias JA, Kumar TR, Bousfield GR. Biased signaling by human follicle-stimulating hormone variants. Pharmacol Ther 2025; 268:108821. [PMID: 39961417 DOI: 10.1016/j.pharmthera.2025.108821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/23/2025]
Abstract
Follicle-stimulating hormone (FSH) or follitropin plays a fundamental role in several mammalian species, including humans. This gonadotropin is produced by the anterior pituitary gland and has as its main targets the granulosa cells of the ovary and the Sertoli cells of the testis. Structurally, FSH is composed of two non-convalently linked subunits, the α- and β-subunit, as well as highly heterogenous oligosaccharide structures, which play a key role in determining a number of physiological and biological features of the hormone. Glycosylation in FSH and the other members belonging to the glycoprotein hormone family, is essential for many functions of the gonadotropin, including subunit assembly and stability, secretion, circulatory half-life and biological activity. Carbohydrate heterogeneity in FSH comes in two forms, microheterogeneity, which results from variations in the carbohydrate structural complexity in those oligosaccharides attached to the α- or β-subunit of the hormone and macroheterogeneity, which results from the absence of carbohydrate chain at FSHβ Asn-glycosylation sites. A number of in vitro and in vivo studies have conclusively demonstrated differential, unique and even opposing effects provoked by variations in the carbohydrate structures of FSH, including circulatory survival, binding to and activation of its cognate receptor in the gonads, intracellular signaling, and activation/inhibition of a number of FSH-regulated genes essential for follicle development. Herein, we review the effects of the FSH oligosaccharides on several functions of FSH, and how variations in these structures have been shown to lead to functional selectivity of the hormone.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico..
| | - Teresa Zariñán
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - James A Dias
- Department of Biomedical Sciences, State University of New York at Albany, Albany, NY, USA
| | - T Rajendra Kumar
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - George R Bousfield
- Department of Biological Sciences, Wichita State University, Wichita, KS, USA
| |
Collapse
|
3
|
Kim SH, Byambaragchaa M, Park SH, Park MH, Kang MH, Min KS. The N-Linked Glycosylation Asn191 and Asn199 Sites Are Controlled Differently Between PKA Signal Transduction and pEKR1/2 Activity in Equine Follicle-Stimulating Hormone Receptor. Curr Issues Mol Biol 2025; 47:168. [PMID: 40136422 PMCID: PMC11940890 DOI: 10.3390/cimb47030168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/25/2025] [Accepted: 03/01/2025] [Indexed: 03/27/2025] Open
Abstract
Equine follicle-stimulating hormone receptor (eFSHR) contains four extracellular N-linked glycosylation sites, which play important roles in agonist-induced signal transduction. Glycosylation regulates G protein-coupled receptor mechanisms by influencing folding, ligand binding, signaling, trafficking, and internalization. Here, we examined whether the glycosylated sites in eFSHR are necessary for cyclic adenosine monophosphate (cAMP) signal transduction and the phosphate extracellular signal-regulated kinase 1/2 (pERK1/2) response. We constructed mutants (N191Q, N199Q, N268Q, and N293Q) of the four N-linked glycosylation sites in eFSHR using site-directed mutagenesis. In wild-type (wt) eFSHR, the cAMP response gradually increased dose-dependently, displaying a strong response at the EC50 and Rmax. Two mutants (N191Q and N199Q) considerably decreased the cAMP response. Both EC50 values were approximately 0.46- and 0.44-fold compared to that of the eFSHR-wt, whereas Rmax levels were 0.29- and 0.45-fold compared to eFSHR-wt because of high-ligand treatment. Specifically, the EC50 and Rmax values in the N268Q mutant were increased 1.23- and 1.46-fold, respectively, by eFSHR-wt. pERK1/2 activity in eFSHR-wt cells was rapid, peaked within 5 min, consistently sustained until 15 min, and then sharply decreased. pERK1/2 activity in the N191Q mutant showed a pattern similar to that of the wild type, despite impaired cAMP responsiveness. The N199Q mutant showed low pERK1/2 activity at 5 and 15 min. Interestingly, pERK1/2 activity in the N268Q and N298Q mutants was similar to that of eFSHR-wt at 5 min, but neither mutant showed any signaling at 15 min, despite displaying high cAMP responsiveness. Overall, eFSHR N-linked glycosylation sites can signal to pERK1/2 via PKA and the other signals, dependent on G protein coupling and β-arrestin-dependent recruitment. Our results provide strong evidence for a new paradigm in which cAMP signaling is not activated, yet pERK1/2 cascade remains strongly induced.
Collapse
Affiliation(s)
- Sung-Hoon Kim
- Graduate School of Animal BioScience, Hankyong National University, Anseong 17579, Republic of Korea; (S.-H.K.); (S.H.P.)
| | - Munkhzaya Byambaragchaa
- Carbon-Neutral Resources Research Center, Hankyong National University, Aseong 17579, Republic of Korea;
- Genetic Engineering, Hankyong National University, Anseong 17579, Republic of Korea
| | - Sei Hyen Park
- Graduate School of Animal BioScience, Hankyong National University, Anseong 17579, Republic of Korea; (S.-H.K.); (S.H.P.)
| | | | - Myung-Hwa Kang
- Department of Food Science and Nutrition, Hoseo University, Asan 31499, Republic of Korea;
| | - Kwan-Sik Min
- Graduate School of Animal BioScience, Hankyong National University, Anseong 17579, Republic of Korea; (S.-H.K.); (S.H.P.)
- Carbon-Neutral Resources Research Center, Hankyong National University, Aseong 17579, Republic of Korea;
- Genetic Engineering, Hankyong National University, Anseong 17579, Republic of Korea
- Division of Animal BioScience, School of Animal Life Convergence Sciences, Hankyong National University, Anseong 17579, Republic of Korea
| |
Collapse
|
4
|
Shpakov AO. Hormonal and Allosteric Regulation of the Luteinizing Hormone/Chorionic Gonadotropin Receptor. FRONT BIOSCI-LANDMRK 2024; 29:313. [PMID: 39344322 DOI: 10.31083/j.fbl2909313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 10/01/2024]
Abstract
Luteinizing hormone (LH) and human chorionic gonadotropin (CG), like follicle-stimulating hormone, are the most important regulators of the reproductive system. They exert their effect on the cell through the LH/CG receptor (LHCGR), which belongs to the family of G protein-coupled receptors. Binding to gonadotropin induces the interaction of LHCGR with various types of heterotrimeric G proteins (Gs, Gq/11, Gi) and β-arrestins, which leads to stimulation (Gs) or inhibition (Gi) of cyclic adenosine monophosphate-dependent cascades, activation of the phospholipase pathway (Gq/11), and also to the formation of signalosomes that mediate the stimulation of mitogen-activated protein kinases (β-arrestins). The efficiency and selectivity of activation of intracellular cascades by different gonadotropins varies, which is due to differences in their interaction with the ligand-binding site of LHCGR. Gonadotropin signaling largely depends on the status of N- and O-glycosylation of LH and CG, on the formation of homo- and heterodimeric receptor complexes, on the cell-specific microenvironment of LHCGR and the presence of autoantibodies to it, and allosteric mechanisms are important in the implementation of these influences, which is due to the multiplicity of allosteric sites in different loci of the LHCGR. The development of low-molecular-weight allosteric regulators of LHCGR with different profiles of pharmacological activity, which can be used in medicine for the correction of reproductive disorders and in assisted reproductive technologies, is promising. These and other issues regarding the hormonal and allosteric regulation of LHCGR are summarized and discussed in this review.
Collapse
Affiliation(s)
- Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
5
|
Zhang X, Zhang X, Zhang Z, Shi Y, Wang J, Ru S, Tian H. Bisphenol S causes excessive estrogen synthesis by activating FSHR and the downstream cAMP/PKA signaling pathway. Commun Biol 2024; 7:844. [PMID: 38987655 PMCID: PMC11237073 DOI: 10.1038/s42003-024-06449-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
Estrogen excess in females has been linked to a diverse array of chronic and acute diseases. Emerging research shows that exposure to estrogen-like compounds such as bisphenol S leads to increases in 17β-estradiol levels, but the mechanism of action is unclear. The aim of this study was to reveal the underlying signaling pathway-mediated mechanisms, target site and target molecule of action of bisphenol S causing excessive estrogen synthesis. Human ovarian granulosa cells SVOG were exposed to bisphenol S at environmentally relevant concentrations (1 μg/L, 10 μg/L, and 100 μg/L) for 48 h. The results confirms that bisphenol S accumulates mainly on the cell membrane, binds to follicle stimulating hormone receptor (FSHR) located on the cell membrane, and subsequently activates the downstream cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) signaling pathway, leading to enhanced conversion of testosterone to 17β-estradiol. This study deepens our knowledge of the mechanisms of environmental factors in pathogenesis of hyperestrogenism.
Collapse
Affiliation(s)
- Xiaorong Zhang
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China
- Tai'an Agriculture and Rural Affairs Bureau, 271000, Tai'an, Shandong Province, China
| | - Xinda Zhang
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China
| | - Zhenzhong Zhang
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China
| | - Yijiao Shi
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China
| | - Jun Wang
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China
| | - Hua Tian
- College of Marine Life Sciences, Ocean University of China, 266003, Qingdao, Shandong Province, China.
| |
Collapse
|
6
|
Zhan T, Zhang J, Zhang Y, Zhao Q, Chemerinski A, Douglas NC, Zhang Q, Xiao S. A Dose-Response Study on Functional and Transcriptomic Effects of FSH on Ex Vivo Mouse Folliculogenesis. Endocrinology 2024; 165:bqae054. [PMID: 38735763 PMCID: PMC11129714 DOI: 10.1210/endocr/bqae054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
Follicle-stimulating hormone (FSH) binds to its membrane receptor (FSHR) in granulosa cells to activate various signal transduction pathways and drive the gonadotropin-dependent phase of folliculogenesis. Both FSH insufficiency (due to genetic or nongenetic factors) and FSH excess (as encountered with ovarian stimulation in assisted reproductive technology [ART]) can cause poor female reproductive outcomes, but the underlying molecular mechanisms remain elusive. Herein, we conducted single-follicle and single-oocyte RNA sequencing analysis along with other approaches in an ex vivo mouse folliculogenesis and oogenesis system to investigate the effects of different concentrations of FSH on key follicular events. Our study revealed that a minimum FSH threshold is required for follicle maturation into the high estradiol-secreting preovulatory stage, and such threshold is moderately variable among individual follicles between 5 and 10 mIU/mL. FSH at 5, 10, 20, and 30 mIU/mL induced distinct expression patterns of follicle maturation-related genes, follicular transcriptomics, and follicular cAMP levels. RNA sequencing analysis identified FSH-stimulated activation of G proteins and downstream canonical and novel signaling pathways that may critically regulate follicle maturation, including the cAMP/PKA/CREB, PI3K/AKT/FOXO1, and glycolysis pathways. High FSH at 20 and 30 mIU/mL resulted in noncanonical FSH responses, including premature luteinization, high production of androgen and proinflammatory factors, and reduced expression of energy metabolism-related genes in oocytes. Together, this study improves our understanding of gonadotropin-dependent folliculogenesis and provides crucial insights into how high doses of FSH used in ART may impact follicular health, oocyte quality, pregnancy outcome, and systemic health.
Collapse
Affiliation(s)
- Tingjie Zhan
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, NJ 08854, USA
- Center for Environmental Exposures and Disease, Rutgers University, Piscataway, NJ 08854, USA
| | - Jiyang Zhang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, NJ 08854, USA
- Center for Environmental Exposures and Disease, Rutgers University, Piscataway, NJ 08854, USA
| | - Ying Zhang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, NJ 08854, USA
- Center for Environmental Exposures and Disease, Rutgers University, Piscataway, NJ 08854, USA
| | - Qingshi Zhao
- Department of Obstetrics, Gynecology and Reproductive Health, New Jersey Medical School (NJMS), Rutgers University, Newark, NJ 07103, USA
| | - Anat Chemerinski
- Department of Obstetrics, Gynecology and Reproductive Health, New Jersey Medical School (NJMS), Rutgers University, Newark, NJ 07103, USA
| | - Nataki C Douglas
- Department of Obstetrics, Gynecology and Reproductive Health, New Jersey Medical School (NJMS), Rutgers University, Newark, NJ 07103, USA
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ 07103, USA
| | - Qiang Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Shuo Xiao
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), Rutgers University, Piscataway, NJ 08854, USA
- Center for Environmental Exposures and Disease, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
7
|
Jardón-Valadez E, Ulloa-Aguirre A. Tracking conformational transitions of the gonadotropin hormone receptors in a bilayer of (SDPC) poly-unsaturated lipids from all-atom molecular dynamics simulations. PLoS Comput Biol 2024; 20:e1011415. [PMID: 38206994 PMCID: PMC10807830 DOI: 10.1371/journal.pcbi.1011415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/24/2024] [Accepted: 12/15/2023] [Indexed: 01/13/2024] Open
Abstract
Glycoprotein hormone receptors [thyrotropin (TSHR), luteinizing hormone/chorionic gonadotropin (LHCGR), and follicle stimulating hormone (FSHR) receptors] are rhodopsin-like G protein-coupled receptors. These receptors display common structural features including a prominent extracellular domain with leucine-rich repeats (LRR) stabilized by β-sheets and a long and flexible loop known as the hinge region (HR), and a transmembrane (TM) domain with seven α-helices interconnected by intra- and extracellular loops. Binding of the ligand to the LRR resembles a hand coupling transversally to the α- and β-subunits of the hormone, with the thumb being the HR. The structure of the FSH-FSHR complex suggests an activation mechanism in which Y335 at the HR binds into a pocket between the α- and β-chains of the hormone, leading to an adjustment of the extracellular loops. In this study, we performed molecular dynamics (MD) simulations to identify the conformational changes of the FSHR and LHCGR. We set up a FSHR structure as predicted by AlphaFold (AF-P23945); for the LHCGR structure we took the cryo-electron microscopy structure for the active state (PDB:7FII) as initial coordinates. Specifically, the flexibility of the HR domain and the correlated motions of the LRR and TM domain were analyzed. From the conformational changes of the LRR, TM domain, and HR we explored the conformational landscape by means of MD trajectories in all-atom approximation, including a membrane of polyunsaturated phospholipids. The distances and procedures here defined may be useful to propose reaction coordinates to describe diverse processes, such as the active-to-inactive transition, and to identify intermediaries suited for allosteric regulation and biased binding to cellular transducers in a selective activation strategy.
Collapse
Affiliation(s)
- Eduardo Jardón-Valadez
- Departamento de Recursos de la Tierra, Unidad Lerma, Universidad Autónoma Metropolitana, Lerma de Villada, Estado de México, Mexico
| | - Alfredo Ulloa-Aguirre
- Instituto Nacional de Ciencias Medicas y Nutrición “Salvador Zubiran”. Mexico City, Mexico
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México. Mexico City, Mexico
| |
Collapse
|
8
|
Zehnaker A, Vallet A, Gourdon J, Sarti C, Jugnarain V, Haj Hassan M, Mathias L, Gauthier C, Raynaud P, Boulo T, Beauclair L, Bigot Y, Casarini L, Crépieux P, Poupon A, Piégu B, Jean-Alphonse F, Bruneau G, Reiter É. Combined Multiplexed Phage Display, High-Throughput Sequencing, and Functional Assays as a Platform for Identifying Modulatory VHHs Targeting the FSHR. Int J Mol Sci 2023; 24:15961. [PMID: 37958944 PMCID: PMC10650796 DOI: 10.3390/ijms242115961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Developing modulatory antibodies against G protein-coupled receptors is challenging. In this study, we targeted the follicle-stimulating hormone receptor (FSHR), a significant regulator of reproduction, with variable domains of heavy chain-only antibodies (VHHs). We built two immune VHH libraries and submitted them to multiplexed phage display approaches. We used next-generation sequencing to identify 34 clusters of specifically enriched sequences that were functionally assessed in a primary screen based on a cAMP response element (CRE)-dependent reporter gene assay. In this assay, 23 VHHs displayed negative or positive modulation of FSH-induced responses, suggesting a high success rate of the multiplexed strategy. We then focused on the largest cluster identified (i.e., PRC1) that displayed positive modulation of FSH action. We demonstrated that PRC1 specifically binds to the human FSHR and human FSHR/FSH complex while potentiating FSH-induced cAMP production and Gs recruitment. We conclude that the improved selection strategy reported here is effective for rapidly identifying functionally active VHHs and could be adapted to target other challenging membrane receptors. This study also led to the identification of PRC1, the first potential positive modulator VHH reported for the human FSHR.
Collapse
Affiliation(s)
- Anielka Zehnaker
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Amandine Vallet
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Juliette Gourdon
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Caterina Sarti
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Vinesh Jugnarain
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Maya Haj Hassan
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Laetitia Mathias
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Camille Gauthier
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Pauline Raynaud
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Thomas Boulo
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Linda Beauclair
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Yves Bigot
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Livio Casarini
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Pascale Crépieux
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Inria, Inria Saclay-Ile-de-France, 91120 Palaiseau, France
| | - Anne Poupon
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Inria, Inria Saclay-Ile-de-France, 91120 Palaiseau, France
- MAbSilico, 1 Impasse du Palais, 37000 Tours, France
| | - Benoît Piégu
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Frédéric Jean-Alphonse
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Inria, Inria Saclay-Ile-de-France, 91120 Palaiseau, France
| | - Gilles Bruneau
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
| | - Éric Reiter
- Physiologie de la Reproduction et des Comportements (PRC), Institut National de Recherche Pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Université de Tours, 37380 Nouzilly, France; (A.Z.); (A.V.); (J.G.); (C.S.); (V.J.); (M.H.H.); (L.M.); (C.G.); (P.R.); (T.B.); (L.B.); (Y.B.); (L.C.); (P.C.); (A.P.); (B.P.); (F.J.-A.)
- Inria, Inria Saclay-Ile-de-France, 91120 Palaiseau, France
| |
Collapse
|
9
|
Guner JZ, Monsivais D, Yu H, Stossi F, Johnson HL, Gibbons WE, Matzuk MM, Palmer S. Oral follicle-stimulating hormone receptor agonist affects granulosa cells differently than recombinant human FSH. Fertil Steril 2023; 120:1061-1070. [PMID: 37532169 PMCID: PMC10659100 DOI: 10.1016/j.fertnstert.2023.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/04/2023]
Abstract
OBJECTIVE To determine whether TOP5300, a novel oral follicle-stimulating hormone (FSH) receptor (FSHR) allosteric agonist, elicits a different cellular response than recombinant human FSH (rh-FSH) in human granulosa cells from patients undergoing in vitro fertilization. DESIGN Basic science research with a preclinical allosteric FSHR agonist. SETTING University hospital. PATIENT(S) Patients with infertility at a single academic fertility clinic were recruited under an Institutional Review Board-approved protocol. Primary granulosa cell cultures were established for 41 patients, of whom 8 had normal ovarian reserve (NOR), 17 were of advanced reproductive age (ARA), 12 had a diagnosis of polycystic ovary syndrome (PCOS), and 4 had a combination of diagnoses, such as ARA and PCOS. INTERVENTION(S) Primary granulosa-lutein (GL) cell cultures were treated with rh-FSH, TOP5300, or vehicle. MAIN OUTCOME MEASURE(S) Estradiol (E2) production using enzyme-linked immunosorbent assay, steroid pathway gene expression of StAR and aromatase using quantitative polymerase chain reaction, and FSHR membrane localization using immunofluorescence were measured in human GL cells. RESULT(S) TOP5300 consistently stimulated E2 production among patients with NOR, ARA, and PCOS. Recombinant FSH was the more potent ligand in GL cells from patients with NOR but was ineffective in cells from patients with ARA or PCOS. The lowest level of FSHR plasma membrane localization was seen in patients with ARA, although FSHR localization was more abundant in cells from patients with PCOS; the highest levels were present in cells from patients with NOR. The localization of FSHR was not affected by TOP5300 relative to rh-FSH in any patient group. TOP5300 stimulated greater expression of StAR and CYP19A1 across cells from all patients with NOR, ARA, and PCOS combined, although rh-FSH was unable to stimulate StAR and aromatase (CYP19A1) expression in cells from patients with PCOS. TOP5300-induced expression of StAR and CYP19A1 mRNA among patients with ARA and NOR was consistently lower than that observed in cells from patients with PCOS. CONCLUSION(S) TOP5300 appears to stimulate E2 production and steroidogenic gene expression from GL cells more than rh-FSH in PCOS, relative to patients with ARA and NOR. It does not appear that localization of FSHR at cell membranes is a limiting step for TOP5300 or rh-FSH stimulation of steroidogenic gene expression and E2 production.
Collapse
Affiliation(s)
- Joie Z Guner
- Center for Drug Discovery, Baylor College of Medicine, Houston, Texas; Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, California
| | - Diana Monsivais
- Center for Drug Discovery, Baylor College of Medicine, Houston, Texas; Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas
| | - Henry Yu
- CanWell Pharma, Wellesley, Massachusetts
| | - Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas; Integrated Microscopy Core and GCC Center for Advanced Microscopy and Image Informatics, Baylor College of Medicine, Houston, Texas
| | - Hannah L Johnson
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas; Integrated Microscopy Core and GCC Center for Advanced Microscopy and Image Informatics, Baylor College of Medicine, Houston, Texas
| | - William E Gibbons
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Baylor College of Medicine, and Texas Children's Hospital Family Fertility Center, Houston, Texas
| | - Martin M Matzuk
- Center for Drug Discovery, Baylor College of Medicine, Houston, Texas; Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas
| | - Stephen Palmer
- Center for Drug Discovery, Baylor College of Medicine, Houston, Texas; Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas.
| |
Collapse
|
10
|
Lazzaretti C, Simoni M, Casarini L, Paradiso E. Allosteric modulation of gonadotropin receptors. Front Endocrinol (Lausanne) 2023; 14:1179079. [PMID: 37305033 PMCID: PMC10248450 DOI: 10.3389/fendo.2023.1179079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
Gonadotropins regulate reproductive functions by binding to G protein-coupled receptors (FSHR and LHCGR) expressed in the gonads. They activate multiple, cell-specific signalling pathways, consisting of ligand-dependent intracellular events. Signalling cascades may be modulated by synthetic compounds which bind allosteric sites of FSHR and LHCGR or by membrane receptor interactions. Despite the hormone binding to the orthosteric site, allosteric ligands, and receptor heteromerizations may reshape intracellular signalling pattern. These molecules act as positive, negative, or neutral allosteric modulators, as well as non-competitive or inverse agonist ligands, providing a set of new compounds of a different nature and with unique pharmacological characteristics. Gonadotropin receptor allosteric modulation is gathering increasing interest from the scientific community and may be potentially exploited for clinical purposes. This review summarizes the current knowledge on gonadotropin receptor allosteric modulation and their potential, clinical use.
Collapse
Affiliation(s)
- Clara Lazzaretti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, Modena, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
- Department of Medical Specialties, Azienda Ospedaliero-Universitaria di Modena, Baggiovara Hospital, Modena, Italy
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Elia Paradiso
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, Baggiovara Hospital, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
11
|
Johnson GP, Onabanjo CGA, Hardy K, Butnev VY, Bousfield GR, Jonas KC. Follicle-Stimulating Hormone Glycosylation Variants Distinctly Modulate Pre-antral Follicle Growth and Survival. Endocrinology 2022; 163:6750033. [PMID: 36201606 PMCID: PMC9598563 DOI: 10.1210/endocr/bqac161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Indexed: 11/19/2022]
Abstract
Follicle-stimulating hormone (FSH) is a key endocrine regulator of ovarian function. FSH is secreted as 2 macroglycosylation variants: partially glycosylated FSH (FSH21/18) and fully glycosylated FSH (FSH24). FSH21/18 is more potent than FSH24 at binding to and activating the FSH receptor (R). The ratio of FSH21/18:FSH24 has been shown to change with age, with FSH21/18 predominant at reproductive prime, and FSH24 predominant during perimenopause/menopause. How these FSH glycosylation variants modulate ovarian follicle functions remains largely unknown. The aim of this study was to investigate the effect of FSH glycosylation variants of pre-antral follicle function. Pre-antral follicles were isolated from 3- to 5-week-old C57BL/6 mice and treated ±10 ng/mL FSH21/18, FSH24, a ratio of 80:20 FSH21/18:FSH24 (to mimic reproductive prime), 50:50 FSH21/18:FSH24 (perimenopause), or 20:80 FSH21/18:FSH24 (menopause) for up to 96 hours. FSH21/18 and 80:20 FSH21/18:FSH24 increased follicle growth, in comparison with control, contrasting with FSH24 and 20:80 FSH21/18:FSH24. Survival rates were decreased in follicles treated with FSH24 or 20:80 FSH21/18:FSH24, with follicles undergoing basement membrane rupture and oocyte extrusion, increased Caspase3 gene and protein expression, and decreased markers of cell proliferation in FSH24 or 20:80 FSH21/18:FSH24-treated follicles. Moreover, this correlated with differential regulation of key genes modulating follicular functions. Pharmacological inhibitors of key FSH signal pathways suggests FSH21/18 and FSH24 initiate different FSHR signal pathway activation, which may determine their differential effects on follicle growth and survival. These data suggest that the nature of FSH glycosylation modulates the follicular cellular environment to regulate follicle growth and survival and may underpin the increasing ovarian resistance to FSH observed during aging.
Collapse
Affiliation(s)
- Gillian P Johnson
- Department of Women and Children's Health, School of Life Course and Population Health Sciences, Kings College London, London SE1 1UL, UK
- Institute of Reproductive and Developmental Biology, Imperial College London, London W12 0NN, UK
| | - Caitlan G A Onabanjo
- Department of Women and Children's Health, School of Life Course and Population Health Sciences, Kings College London, London SE1 1UL, UK
| | - Kate Hardy
- Institute of Reproductive and Developmental Biology, Imperial College London, London W12 0NN, UK
| | - Viktor Y Butnev
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260, USA
| | - George R Bousfield
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260, USA
| | - Kim C Jonas
- Correspondence: Kim C. Jonas, MD, PhD, Department of Women and Children's Health, School of Life Course and Population Health Sciences, Kings College London, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
12
|
Nagel M, Moretti R, Paschke R, von Bergen M, Meiler J, Kalkhof S. Integrative model of the FSH receptor reveals the structural role of the flexible hinge region. Structure 2022; 30:1424-1431.e3. [PMID: 35973423 DOI: 10.1016/j.str.2022.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/16/2021] [Accepted: 07/20/2022] [Indexed: 11/16/2022]
Abstract
The follicle-stimulating hormone receptor (FSHR) belongs to the glycoprotein hormone receptors, a subfamily of G-protein-coupled receptors (GPCRs). FSHR is involved in reproductive processes such as gonadal development and maturation. Structurally, the extensive extracellular domain, which contains the hormone-binding site and is linked to the transmembrane domain by the hinge region (HR), is characteristic for these receptors. How this HR is involved in hormone binding and signal transduction is still an open question. We combined in vitro and in situ chemical crosslinking, disulfide pattern analysis, and mutation data with molecular modeling to generate experimentally driven full-length models. These models provide insights into the interface, important side-chain interactions, and activation mechanism. The interface indicates a strong involvement of the connecting loop. A major rearrangement of the HR seems implausible due to the tight arrangement and fixation by disulfide bonds. The models are expected to allow for testable hypotheses about signal transduction and drug development for GPHRs.
Collapse
Affiliation(s)
- Marcus Nagel
- Department for Molecular Systems Biology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany; Center for Structural Biology, Vanderbilt University, Nashville, TN 37212, USA; Division of Endocrinology, Department of Endocrinology and Nephrology, University Clinic Leipzig, Germany
| | - Rocco Moretti
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37212, USA
| | - Ralf Paschke
- Division of Endocrinology, Department of Endocrinology and Nephrology, University Clinic Leipzig, Germany; Department of Medicine, Division of Endocrinology, Departments of Oncology, Pathology, and Biochemistry and Molecular Biology & Arnie Charbonneau Cancer Institute Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Martin von Bergen
- Department for Molecular Systems Biology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany; Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, Leipzig, Germany
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37212, USA; Leipzig University Medical School, Institute for Drug Discovery, 04103 Leipzig, Germany.
| | - Stefan Kalkhof
- Department for Molecular Systems Biology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany; Institute for Bioanalysis, University of Applied Sciences Coburg, Coburg, Germany; Fraunhofer Institute for Cell Therapy and Immunology, Department of Preclinical Development and Validation, 04103 Leipzig, Germany.
| |
Collapse
|
13
|
Combarnous Y, Nguyen TMD. Membrane Hormone Receptors and Their Signaling Pathways as Targets for Endocrine Disruptors. J Xenobiot 2022; 12:64-73. [PMID: 35466213 PMCID: PMC9036253 DOI: 10.3390/jox12020007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 12/19/2022] Open
Abstract
The endocrine disruptors are mostly small organic molecules developed for numerous and very diverse industrial applications. They essentially act through nuclear receptors with small and hydrophobic endogenous ligands. Nevertheless, potential adverse effects through membrane hormone receptors cannot be ruled out, and have indeed been observed. The present paper reviews how orthosteric and allosteric binding sites of the different families of membrane receptors can be targets for man-made hydrophobic molecules (components of plastics, paints, flame retardants, herbicides, pesticides, etc.). We also review potential target proteins for such small hydrophobic molecules downstream of membrane receptors at the level of their intracellular signaling pathways. From the currently available information, although endocrine disruptors primarily affect nuclear receptors’ signaling, membrane receptors for hormones, cytokines, neuro-mediators, and growth factors can be affected as well and deserve attention.
Collapse
Affiliation(s)
- Yves Combarnous
- INRAe, CNRS, Tours University Joint Unit, Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France;
- Correspondence: ; Tel.: +33-(0)24-7427-650
| | - Thi Mong Diep Nguyen
- INRAe, CNRS, Tours University Joint Unit, Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France;
- Faculty of Natural Sciences, Quy Nhon University, Quy Nhon 820000, Vietnam
| |
Collapse
|
14
|
Lundin K, Sepponen K, Väyrynen P, Liu X, Yohannes DA, Survila M, Ghimire B, Känsäkoski J, Katayama S, Partanen J, Vuoristo S, Paloviita P, Rahman N, Raivio T, Luiro K, Huhtaniemi I, Varjosalo M, Tuuri T, Tapanainen JS. OUP accepted manuscript. Mol Hum Reprod 2022; 28:6574364. [PMID: 35471239 PMCID: PMC9308958 DOI: 10.1093/molehr/gaac012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 03/11/2022] [Indexed: 11/14/2022] Open
Affiliation(s)
- K Lundin
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - K Sepponen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - P Väyrynen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - X Liu
- Molecular Systems Biology Research Group, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
- Proteomics Unit, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
| | - D A Yohannes
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Translational Immunology & Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - M Survila
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - B Ghimire
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - J Känsäkoski
- Department of Physiology, University of Helsinki, Helsinki, Finland
| | - S Katayama
- Folkhälsan Research Center, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J Partanen
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - S Vuoristo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - P Paloviita
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - N Rahman
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - T Raivio
- Department of Physiology, University of Helsinki, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- New Children's Hospital, Pediatric Research Center, Helsinki University Hospital, HUH, Helsinki, Finland
| | - K Luiro
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - I Huhtaniemi
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Metabolism, Endocrinology and Reproduction, Faculty of Medicine, Hammersmith Campus, Imperial College London, London, UK
| | - M Varjosalo
- Molecular Systems Biology Research Group, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
- Proteomics Unit, Institute of Biotechnology & HiLIFE, University of Helsinki, Helsinki, Finland
| | - T Tuuri
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - J S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynecology, University Hospital of Oulu, University of Oulu, Medical Research Center Oulu and PEDEGO Research Unit, Oulu, Finland
- Corresponding author. Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, PO Box 140, 00029 Helsinki, Finland. Tel: +358-94711; E-mail:
| |
Collapse
|
15
|
Pharmacological Characterization of Low Molecular Weight Biased Agonists at the Follicle Stimulating Hormone Receptor. Int J Mol Sci 2021; 22:ijms22189850. [PMID: 34576014 PMCID: PMC8469697 DOI: 10.3390/ijms22189850] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 01/14/2023] Open
Abstract
Follicle-stimulating hormone receptor (FSHR) plays a key role in reproduction through the activation of multiple signaling pathways. Low molecular weight (LMW) ligands composed of biased agonist properties are highly valuable tools to decipher complex signaling mechanisms as they allow selective activation of discrete signaling cascades. However, available LMW FSHR ligands have not been fully characterized yet. In this context, we explored the pharmacological diversity of three benzamide and two thiazolidinone derivatives compared to FSH. Concentration/activity curves were generated for Gαs, Gαq, Gαi, β-arrestin 2 recruitment, and cAMP production, using BRET assays in living cells. ERK phosphorylation was analyzed by Western blotting, and CRE-dependent transcription was assessed using a luciferase reporter assay. All assays were done in either wild-type, Gαs or β-arrestin 1/2 CRISPR knockout HEK293 cells. Bias factors were calculated for each pair of read-outs by using the operational model. Our results show that each ligand presented a discrete pharmacological efficacy compared to FSH, ranging from super-agonist for β-arrestin 2 recruitment to pure Gαs bias. Interestingly, LMW ligands generated kinetic profiles distinct from FSH (i.e., faster, slower or transient, depending on the ligand) and correlated with CRE-dependent transcription. In addition, clear system biases were observed in cells depleted of either Gαs or β-arrestin genes. Such LMW properties are useful pharmacological tools to better dissect the multiple signaling pathways activated by FSHR and assess their relative contributions at the cellular and physio-pathological levels.
Collapse
|
16
|
Johnson GP, Jonas KC. Mechanistic insight into how gonadotropin hormone receptor complexes direct signaling†. Biol Reprod 2021; 102:773-783. [PMID: 31882999 DOI: 10.1093/biolre/ioz228] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/09/2019] [Accepted: 12/17/2019] [Indexed: 12/29/2022] Open
Abstract
Gonadotropin hormones and their receptors play a central role in the control of male and female reproduction. In recent years, there has been growing evidence surrounding the complexity of gonadotropin hormone/receptor signaling, with it increasingly apparent that the Gαs/cAMP/PKA pathway is not the sole signaling pathway that confers their biological actions. Here we review recent literature on the different receptor-receptor, receptor-scaffold, and receptor-signaling molecule complexes formed and how these modulate and direct gonadotropin hormone-dependent intracellular signal activation. We will touch upon the more controversial issue of extragonadal expression of FSHR and the differential signal pathways activated in these tissues, and lastly, highlight the open questions surrounding the role these gonadotropin hormone receptor complexes and how this will shape future research directions.
Collapse
Affiliation(s)
| | - Kim Carol Jonas
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| |
Collapse
|
17
|
Novel allosteric ligands of the angiotensin receptor AT1R as autoantibody blockers. Proc Natl Acad Sci U S A 2021; 118:2019126118. [PMID: 34380734 DOI: 10.1073/pnas.2019126118] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
While orthosteric ligands of the angiotensin II (AngII) type 1 receptor (AT1R) are available for clinical and research applications, allosteric ligands are not known for this important G protein-coupled receptor (GPCR). Allosteric ligands are useful tools to modulate receptor pharmacology and subtype selectivity. Here, we report AT1R allosteric ligands for a potential application to block autoimmune antibodies. The epitope of autoantibodies for AT1R is outside the orthosteric pocket in the extracellular loop 2. A molecular dynamics simulation study of AT1R structure reveals the presence of a druggable allosteric pocket encompassing the autoantibody epitope. Small molecule binders were then identified for this pocket using structure-based high-throughput virtual screening. The top 18 hits obtained inhibited the binding of antibody to AT1R and modulated agonist-induced calcium response of AT1R. Two compounds out of 18 studied in detail exerted a negative allosteric modulator effect on the functions of the natural agonist AngII. They blocked antibody-enhanced calcium response and reactive oxygen species production in vascular smooth muscle cells as well as AngII-induced constriction of blood vessels, demonstrating their efficacy in vivo. Our study thus demonstrates the feasibility of discovering inhibitors of the disease-causing autoantibodies for GPCRs. Specifically, for AT1R, we anticipate development of more potent allosteric drug candidates for intervention in autoimmune maladies such as preeclampsia, bilateral adrenal hyperplasia, and the rejection of organ transplants.
Collapse
|
18
|
Hua G, George JW, Clark KL, Jonas KC, Johnson GP, Southekal S, Guda C, Hou X, Blum HR, Eudy J, Butnev VY, Brown AR, Katta S, May JV, Bousfield GR, Davis JS. Hypo-glycosylated hFSH drives ovarian follicular development more efficiently than fully-glycosylated hFSH: enhanced transcription and PI3K and MAPK signaling. Hum Reprod 2021; 36:1891-1906. [PMID: 34059912 PMCID: PMC8213452 DOI: 10.1093/humrep/deab135] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/31/2021] [Indexed: 12/22/2022] Open
Abstract
STUDY QUESTION Does hypo-glycosylated human recombinant FSH (hFSH18/21) have greater in vivo bioactivity that drives follicle development in vivo compared to fully-glycosylated human recombinant FSH (hFSH24)? SUMMARY ANSWER Compared with fully-glycosylated hFSH, hypo-glycosylated hFSH has greater bioactivity, enabling greater follicular health and growth in vivo, with enhanced transcriptional activity, greater activation of receptor tyrosine kinases (RTKs) and elevated phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) and Mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling. WHAT IS KNOWN ALREADY Glycosylation of FSH is necessary for FSH to effectively activate the FSH receptor (FSHR) and promote preantral follicular growth and formation of antral follicles. In vitro studies demonstrate that compared to fully-glycosylated recombinant human FSH, hypo-glycosylated FSH has greater activity in receptor binding studies, and more effectively stimulates the PKA pathway and steroidogenesis in human granulosa cells. STUDY DESIGN, SIZE, DURATION This is a cross-sectional study evaluating the actions of purified recombinant human FSH glycoforms on parameters of follicular development, gene expression and cell signaling in immature postnatal day (PND) 17 female CD-1 mice. To stimulate follicle development in vivo, PND 17 female CD-1 mice (n = 8-10/group) were treated with PBS (150 µl), hFSH18/21 (1 µg/150 µl PBS) or hFSH24 (1 µg/150 µl PBS) by intraperitoneal injection (i.p.) twice daily (8:00 a.m. and 6:00 p.m.) for 2 days. Follicle numbers, serum anti-Müllerian hormone (AMH) and estradiol levels, and follicle health were quantified. PND 17 female CD-1 mice were also treated acutely (2 h) in vivo with PBS, hFSH18/21 (1 µg) or hFSH24 (1 µg) (n = 3-4/group). One ovary from each mouse was processed for RNA sequencing analysis and the other ovary processed for signal transduction analysis. An in vitro ovary culture system was used to confirm the relative signaling pathways. PARTICIPANTS/MATERIALS, SETTING, METHODS The purity of different recombinant hFSH glycoforms was analyzed using an automated western blot system. Follicle numbers were determined by counting serial sections of the mouse ovary. Real-time quantitative RT-PCR, western blot and immunofluorescence staining were used to determine growth and apoptosis markers related with follicle health. RNA sequencing and bioinformatics were used to identify pathways and processes associated with gene expression profiles induced by acute FSH glycoform treatment. Analysis of RTKs was used to determine potential FSH downstream signaling pathways in vivo. Western blot and in vitro ovarian culture system were used to validate the relative signaling pathways. MAIN RESULTS AND THE ROLE OF CHANCE Our present study shows that both hypo- and fully-glycosylated recombinant human FSH can drive follicular growth in vivo. However, hFSH18/21 promoted development of significantly more large antral follicles compared to hFSH24 (P < 0.01). In addition, compared with hFSH24, hFSH18/21 also promoted greater indices of follicular health, as defined by lower BAX/BCL2 ratios and reduced cleaved Caspase 3. Following acute in vivo treatment with FSH glycoforms RNA-sequencing data revealed that both FSH glycoforms rapidly induced ovarian transcription in vivo, but hypo-glycosylated FSH more robustly stimulated Gαs and cAMP-mediated signaling and members of the AP-1 transcription factor complex. Moreover, hFSH18/21 treatment induced significantly greater activation of RTKs, PI3K/AKT and MAPK/ERK signaling compared to hFSH24. FSH-induced indices of follicle growth in vitro were blocked by inhibition of PI3K and MAPK. LARGE SCALE DATA RNA sequencing of mouse ovaries. Data will be shared upon reasonable request to the corresponding author. LIMITATIONS, REASONS FOR CAUTION The observations that hFSH glycoforms have different bioactivities in the present study employing a mouse model of follicle development should be verified in nonhuman primates. The gene expression studies reflect transcriptomes of whole ovaries. WIDER IMPLICATIONS OF THE FINDINGS Commercially prepared recombinant human FSH used for ovarian stimulation in human ART is fully-glycosylated FSH. Our findings that hypo-glycosylated hFSH has greater bioactivity enabling greater follicular health and growth without exaggerated estradiol production in vivo, demonstrate the potential for its development for application in human ART. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by NIH 1P01 AG029531, NIH 1R01 HD 092263, VA I01 BX004272, and the Olson Center for Women's Health. JSD is the recipient of a VA Senior Research Career Scientist Award (1IK6 BX005797). This work was also partially supported by National Natural Science Foundation of China (No. 31872352). The authors declared there are no conflicts of interest.
Collapse
Affiliation(s)
- Guohua Hua
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science & Technology, Huazhong Agricultural University, Wuhan, Hubei, China
- Department of Obstetrics and Gynecology, Olson Center for Women’s Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jitu W George
- Department of Obstetrics and Gynecology, Olson Center for Women’s Health, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE, USA
| | - Kendra L Clark
- Department of Obstetrics and Gynecology, Olson Center for Women’s Health, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE, USA
| | - Kim C Jonas
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College London, Guy’s Campus, London, UK
| | - Gillian P Johnson
- Department of Women and Children’s Health, School of Life Course Sciences, King’s College London, Guy’s Campus, London, UK
| | - Siddesh Southekal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xiaoying Hou
- Department of Obstetrics and Gynecology, Olson Center for Women’s Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Haley R Blum
- Department of Obstetrics and Gynecology, Olson Center for Women’s Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - James Eudy
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Viktor Y Butnev
- Department of Biological Sciences, Wichita State University, Wichita, KS, USA
| | - Alan R Brown
- Department of Biological Sciences, Wichita State University, Wichita, KS, USA
| | - Sahithi Katta
- Department of Biological Sciences, Wichita State University, Wichita, KS, USA
| | - Jeffrey V May
- Department of Biological Sciences, Wichita State University, Wichita, KS, USA
| | - George R Bousfield
- Department of Biological Sciences, Wichita State University, Wichita, KS, USA
| | - John S Davis
- Department of Obstetrics and Gynecology, Olson Center for Women’s Health, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
19
|
Bosch E, Alviggi C, Lispi M, Conforti A, Hanyaloglu AC, Chuderland D, Simoni M, Raine-Fenning N, Crépieux P, Kol S, Rochira V, D'Hooghe T, Humaidan P. Reduced FSH and LH action: implications for medically assisted reproduction. Hum Reprod 2021; 36:1469-1480. [PMID: 33792685 PMCID: PMC8129594 DOI: 10.1093/humrep/deab065] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
Luteinizing hormone (LH) and follicle-stimulating hormone (FSH) play complementary roles in follicle development and ovulation via a complex interaction in the hypothalamus, anterior pituitary gland, reproductive organs, and oocytes. Impairment of the production or action of gonadotropins causes relative or absolute LH and FSH deficiency that compromises gametogenesis and gonadal steroid production, thereby reducing fertility. In women, LH and FSH deficiency is a spectrum of conditions with different functional or organic causes that are characterized by low or normal gonadotropin levels and low oestradiol levels. While the causes and effects of reduced LH and FSH production are very well known, the notion of reduced action has received less attention by researchers. Recent evidence shows that molecular characteristics, signalling as well as ageing, and some polymorphisms negatively affect gonadotropin action. These findings have important clinical implications, in particular for medically assisted reproduction in which diminished action determined by the afore-mentioned factors, combined with reduced endogenous gonadotropin production caused by GnRH analogue protocols, may lead to resistance to gonadotropins and, thus, to an unexpected hypo-response to ovarian stimulation. Indeed, the importance of LH and FSH action has been highlighted by the International Committee for Monitoring Assisted Reproduction Technologies (ICMART) in their definition of hypogonadotropic hypogonadism as gonadal failure associated with reduced gametogenesis and gonadal steroid production due to reduced gonadotropin production or action. The aim of this review is to provide an overview of determinants of reduced FSH and LH action that are associated with a reduced response to ovarian stimulation.
Collapse
Affiliation(s)
| | - C Alviggi
- Department of Neuroscience, Reproductive Science and Odontostomatology, University Federico II, Naples, Italy
| | - M Lispi
- Global Medical Affairs Fertility, Merck KGaA, Darmstadt, Germany.,International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Modena, Italy
| | - A Conforti
- Department of Neuroscience, Reproductive Science and Odontostomatology, University Federico II, Naples, Italy
| | - A C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - D Chuderland
- Global Medical Affairs Fertility, Merck KGaA, Darmstadt, Germany
| | - M Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - N Raine-Fenning
- Department of Child Health, Obstetrics and Gynaecology, School of Medicine, University of Nottingham, Nottingham, UK
| | - P Crépieux
- Physiologie de la Reproduction et des Comportements, UMR INRA 085, CNRS 7247, Université de Tours, Nouzilly, France
| | - S Kol
- IVF Unit, Elisha Hospital, Haifa, Israel
| | - V Rochira
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.,Unit of Endocrinology, Azienda Ospedaliero-Universitaria of Modena, Ospedale Civile di Baggiovara, Modena, Italy
| | - T D'Hooghe
- Global Medical Affairs Fertility, Merck KGaA, Darmstadt, Germany.,Department of Development & Regeneration, University of Leuven (KU Leuven), Leuven, Belgium.,Department of Obstetrics and Gynecology, Yale University, New Haven, CT, USA
| | - P Humaidan
- Fertility Clinic, Skive Regional Hospital, and the Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
20
|
Abstract
Gonadotropins are glycoprotein sex hormones regulating development and reproduction and bind to specific G protein–coupled receptors expressed in the gonads. Their effects on multiple signaling cascades and intracellular events have recently been characterized using novel technological and scientific tools. The impact of allosteric modulators on gonadotropin signaling, the role of sugars linked to the hormone backbone, the detection of endosomal compartments supporting signaling modules, and the dissection of different effects mediated by these molecules are areas that have advanced significantly in the last decade. The classic view providing the exclusive activation of the cAMP/protein kinase A (PKA) and the steroidogenic pathway by these hormones has been expanded with the addition of novel signaling cascades as determined by high-resolution imaging techniques. These new findings provided new potential therapeutic applications. Despite these improvements, unanswered issues of gonadotropin physiology, such as the intrinsic pro-apoptotic potential to these hormones, the existence of receptors assembled as heteromers, and their expression in extragonadal tissues, remain to be studied. Elucidating these issues is a challenge for future research.
Collapse
Affiliation(s)
- Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria di Modena, Via P. Giardini 1355, 41126 Modena, Italy
| |
Collapse
|
21
|
Slosky LM, Caron MG, Barak LS. Biased Allosteric Modulators: New Frontiers in GPCR Drug Discovery. Trends Pharmacol Sci 2021; 42:283-299. [PMID: 33581873 PMCID: PMC9797227 DOI: 10.1016/j.tips.2020.12.005] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/31/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest class of cell surface receptors in the genome and the most successful family of targets of FDA-approved drugs. New frontiers in GPCR drug discovery remain, however, as achieving receptor subtype selectivity and controlling off- and on-target side effects are not always possible with classic agonist and antagonist ligands. These challenges may be overcome by focusing development efforts on allosteric ligands that confer signaling bias. Biased allosteric modulators (BAMs) are an emerging class of GPCR ligands that engage less well-conserved regulatory motifs outside the orthosteric pocket and exert pathway-specific effects on receptor signaling. The unique ways that BAMs texturize receptor signaling present opportunities to fine-tune physiology and develop safer, more selective therapeutics. Here, we provide a conceptual framework for understanding the pharmacology of BAMs, explore their therapeutic potential, and discuss strategies for their discovery.
Collapse
Affiliation(s)
- Lauren M. Slosky
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Marc G. Caron
- Departments of Cell Biology, Neurobiology and Medicine, Duke University, Durham, NC 27710, USA,Correspondence: (L.S.B.); (M.G.C.)
| | - Lawrence S. Barak
- Department of Cell Biology, Duke University, Durham, NC 27710, USA,Correspondence: (L.S.B.); (M.G.C.)
| |
Collapse
|
22
|
Nataraja S, Yu H, Guner J, Palmer S. Discovery and Preclinical Development of Orally Active Small Molecules that Exhibit Highly Selective Follicle Stimulating Hormone Receptor Agonism. Front Pharmacol 2021; 11:602593. [PMID: 33519465 PMCID: PMC7845544 DOI: 10.3389/fphar.2020.602593] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/17/2020] [Indexed: 12/17/2022] Open
Abstract
An orally active follicle stimulating hormone receptor allosteric agonist would provide a preferred treatment for over 16 million infertile women of reproductive age in low complexity methods (ovulation induction-intrauterine insemination) or in high complexity methods (controlled ovarian stimulation-in vitro fertilization). We present two oral follicle stimulating hormone receptor allosteric agonist compounds that have the desired pharmacology, drug metabolism, pharmacokinetics, and safety profile for clinical use. These molecules provide a single agent suitable for ovulation induction-intrauterine insemination or controlled ovarian stimulation-in vitro fertilization that is more convenient for patients and achieves similar preclinical efficacy as rec-hFSH. TOP5668, TOP5300 were evaluated in vitro in Chinese hamster ovary cells transfected with individual glycoprotein receptors measuring cAMP (FSHR, LH/CGR, thyroid stimulating hormone receptor). TOP5668 was found to have solely follicle stimulating hormone receptor allosteric agonist activity while TOP5300 was found to have mixed follicle stimulating hormone receptor allosteric agonist and LHR-AA activity. Both compounds stimulated concentration-dependent increases in estradiol production from cultured rat granulosa cells in the presence or absence of low dose rec-hFSH, while only TOP5300 stimulated testosterone production from rat primary Leydig cells. In pooled human granulosa cells obtained from patients undergoing controlled ovarian stimulation-in vitro fertilization, TOP5300 stimulated 7-fold greater maximal estradiol response than rec-hFSH and TOP5668 was 10-fold more potent than TOP5300. Both TOP5300 and TOP5668 stimulated follicular development in immature rat to the same efficacy as recombinant follicle stimulating hormone. In mice treated with TOP5300, in the presence of low dose of follicle stimulating hormone, there were no differences in oocyte number, fertilization rate, and hatched blastocyst rate in mice with TOP5300 and low dose follicle stimulating hormone vs. reference proteins pregnant mare serum gonadotropin or high dose rec-hFSH. ADME/PK and safety profiles were favorable. In addition, there was no appreciable activity on thyroid hormones by TOP5300 in 14-days toxicological study in rat or dog. The selected lead compound, TOP5300 stimulated a more robust increase in estradiol production from granulosa-lutein cells from women with polycystic ovarian syndrome patient compared to rec-hFSH. Conclusions: Two novel oral FSHR allosteric agonist, TOP5668 and TOP5300, were found to mimic the biological activity of rec hFSH in preclinical studies. Both compounds led to folliculogenesis and superovulation in rat and mice. Specifically, TOP5300 led to a similar number of ovulated oocytes that fertilized and developed into hatched blastocysts in mice when compared to rec-hFSH. The safety profile demonstrated lack of toxicity.
Collapse
Affiliation(s)
| | | | - Joie Guner
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
| | - Stephen Palmer
- TocopheRx, Inc., Groton, MA, United States
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
23
|
Agwuegbo UT, Colley E, Albert AP, Butnev VY, Bousfield GR, Jonas KC. Differential FSH Glycosylation Modulates FSHR Oligomerization and Subsequent cAMP Signaling. Front Endocrinol (Lausanne) 2021; 12:765727. [PMID: 34925235 PMCID: PMC8678890 DOI: 10.3389/fendo.2021.765727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/02/2021] [Indexed: 01/18/2023] Open
Abstract
Follicle-stimulating hormone (FSH) and its target G protein-coupled receptor (FSHR) are essential for reproduction. Recent studies have established that the hypo-glycosylated pituitary FSH glycoform (FSH21/18), is more bioactive in vitro and in vivo than the fully-glycosylated variant (FSH24). FSH21/18 predominates in women of reproductive prime and FSH24 in peri-post-menopausal women, suggesting distinct functional roles of these FSH glycoforms. The aim of this study was to determine if differential FSH glycosylation modulated FSHR oligomerization and resulting impact on cAMP signaling. Using a modified super-resolution imaging technique (PD-PALM) to assess FSHR complexes in HEK293 cells expressing FSHR, we observed time and concentration-dependent modulation of FSHR oligomerization by FSH glycoforms. High eFSH and FSH21/18 concentrations rapidly dissociated FSHR oligomers into monomers, whereas FSH24 displayed slower kinetics. The FSHR β-arrestin biased agonist, truncated eLHβ (Δ121-149) combined with asparagine56-deglycosylated eLHα (dg-eLHt), increased FSHR homomerization. In contrast, low FSH21/18 and FSH24 concentrations promoted FSHR association into oligomers. Dissociation of FSHR oligomers correlated with time points where higher cAMP production was observed. Taken together, these data suggest that FSH glycosylation may modulate the kinetics and amplitude of cAMP production, in part, by forming distinct FSHR complexes, highlighting potential avenues for novel therapeutic targeting of the FSHR to improve IVF outcomes.
Collapse
Affiliation(s)
- Uchechukwu T. Agwuegbo
- School of Life Course and Population Sciences, Department of Women and Children’s Health, Guy’s Campus, King’s College London, London, United Kingdom
| | - Emily Colley
- Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Anthony P. Albert
- Vascular Biology Research Centre, Molecular & Clinical Science Research Centre, St George’s University of London, London, United Kingdom
| | - Viktor Y. Butnev
- Department of Biological Sciences, Wichita State University, Wichita, KS, United States
| | - George R. Bousfield
- Department of Biological Sciences, Wichita State University, Wichita, KS, United States
| | - Kim C. Jonas
- School of Life Course and Population Sciences, Department of Women and Children’s Health, Guy’s Campus, King’s College London, London, United Kingdom
- *Correspondence: Kim C. Jonas,
| |
Collapse
|
24
|
Banerjee AA, Joseph S, Mahale SD. From cell surface to signalling and back: the life of the mammalian FSH receptor. FEBS J 2020; 288:2673-2696. [DOI: 10.1111/febs.15649] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/17/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Antara A. Banerjee
- Division of Structural Biology National Institute for Research in Reproductive Health (Indian Council of Medical Research) Parel India
| | - Shaini Joseph
- Genetic Research Center National Institute for Research in Reproductive Health (Indian Council of Medical Research) Parel India
| | - Smita D. Mahale
- Division of Structural Biology National Institute for Research in Reproductive Health (Indian Council of Medical Research) Parel India
- ICMR Biomedical Informatics Centre National Institute for Research in Reproductive Health (Indian Council of Medical Research) Parel India
| |
Collapse
|
25
|
Clément F, Crépieux P, Yvinec R, Monniaux D. Mathematical modeling approaches of cellular endocrinology within the hypothalamo-pituitary-gonadal axis. Mol Cell Endocrinol 2020; 518:110877. [PMID: 32569857 DOI: 10.1016/j.mce.2020.110877] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 01/26/2023]
Abstract
The reproductive neuroendocrine axis, or hypothalamo-pituitary-gonadal (HPG) axis, is a paragon of complex biological system involving numerous cell types, spread over several anatomical levels communicating through entangled endocrine feedback loops. The HPG axis exhibits remarkable dynamic behaviors on multiple time and space scales, which are an inexhaustible source of studies for mathematical and computational biology. In this review, we will describe a variety of modeling approaches of the HPG axis from a cellular endocrinology viewpoint. We will in particular investigate the questions raised by some of the most striking features of the HPG axis: (i) the pulsatile secretion of hypothalamic and pituitary hormones, and its counterpart, the cell signaling induced by frequency-encoded hormonal signals, and (ii) the dual, gametogenic and glandular function of the gonads, which relies on the tight control of the somatic cell populations ensuring the proper maturation and timely release of the germ cells.
Collapse
Affiliation(s)
- Frédérique Clément
- Inria, Centre de Recherche Inria Saclay-Île-de-France, Palaiseau, France.
| | - Pascale Crépieux
- INRAE, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380, Nouzilly, France; CNRS, UMR7247, F-37380, Nouzilly, France; Université de Tours, F-37041, Tours, France
| | - Romain Yvinec
- INRAE, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380, Nouzilly, France; CNRS, UMR7247, F-37380, Nouzilly, France; Université de Tours, F-37041, Tours, France
| | - Danielle Monniaux
- INRAE, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380, Nouzilly, France; CNRS, UMR7247, F-37380, Nouzilly, France; Université de Tours, F-37041, Tours, France
| |
Collapse
|
26
|
Zariñán T, Butnev VY, Gutiérrez-Sagal R, Maravillas-Montero JL, Martínez-Luis I, Mejía-Domínguez NR, Juárez-Vega G, Bousfield GR, Ulloa-Aguirre A. In Vitro Impact of FSH Glycosylation Variants on FSH Receptor-stimulated Signal Transduction and Functional Selectivity. J Endocr Soc 2020; 4:bvaa019. [PMID: 32342021 PMCID: PMC7175721 DOI: 10.1210/jendso/bvaa019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022] Open
Abstract
FSH exists as different glycoforms that differ in glycosylation of the hormone-specific β-subunit. Tetra-glycosylated FSH (FSH24) and hypo-glycosylated FSH (FSH18/21) are the most abundant glycoforms found in humans. Employing distinct readouts in HEK293 cells expressing the FSH receptor, we compared signaling triggered by human pituitary FSH preparations (FSH18/21 and FSH24) as well as by equine FSH (eFSH), and human recombinant FSH (recFSH), each exhibiting distinct glycosylation patterns. The potency in eliciting cAMP production was greater for eFSH than for FSH18/21, FSH24, and recFSH, whereas in the ERK1/2 activation readout, potency was highest for FSH18/21 followed by eFSH, recFSH, and FSH24. In β-arrestin1/2 CRISPR/Cas9 HEK293-KO cells, FSH18/21 exhibited a preference toward β-arrestin-mediated ERK1/2 activation as revealed by a drastic decrease in pERK during the first 15-minute exposure to this glycoform. Exposure of β-arrestin1/2 KO cells to H89 additionally decreased pERK1/2, albeit to a significantly lower extent in response to FSH18/21. Concurrent silencing of β-arrestin and PKA signaling, incompletely suppressed pERK response to FSH glycoforms, suggesting that pathways other than those dependent on Gs-protein and β-arrestins also contribute to FSH-stimulated pERK1/2. All FSH glycoforms stimulated intracellular Ca2+ (iCa2+) accumulation through both influx from Ca2+ channels and release from intracellular stores; however, iCa2+ in response to FSH18/21 depended more on the latter, suggesting differences in mechanisms through which glycoforms promote iCa2+ accumulation. These data indicate that FSH glycosylation plays an important role in defining not only the intensity but also the functional selectivity for the mechanisms leading to activation of distinct signaling cascades.
Collapse
Affiliation(s)
- Teresa Zariñán
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Viktor Y Butnev
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
| | - Rubén Gutiérrez-Sagal
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José Luis Maravillas-Montero
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Iván Martínez-Luis
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Nancy R Mejía-Domínguez
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Guillermo Juárez-Vega
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - George R Bousfield
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
| | - Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|