1
|
Pienkowski T, Golonko A, Bolkun L, Wawrzak-Pienkowska K, Szczerbinski L, Kretowski A, Ciborowski M, Lewandowski W, Priebe W, Swislocka R. Investigation into biased signaling, glycosylation, and drug vulnerability of acute myeloid leukemia. Pharmacol Ther 2025; 270:108848. [PMID: 40194743 DOI: 10.1016/j.pharmthera.2025.108848] [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: 09/03/2024] [Revised: 01/22/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025]
Abstract
Understanding and harnessing biased signaling offers significant potential for developing novel therapeutic strategies or enhancing existing treatments. By managing biased signaling, it is possible to minimize adverse effects, including toxicity, and to optimize therapeutic outcomes by selectively targeting beneficial pathways. In the context of acute myeloid leukemia (AML), a highly aggressive blood cancer characterized by the rapid proliferation of abnormal myeloid cells in the bone marrow and blood, the dysregulation of these signaling pathways, particularly those involving G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs), significantly contributes to disease progression and therapeutic resistance. Traditional therapies for AML often struggle with resistance and toxicity, leading to poor patient outcomes. However, by exploiting the concept of biased signaling, researchers may be able to design drugs that selectively activate pathways that inhibit cancer cell growth while avoiding those that contribute to resistance or toxicity. Glycosylation, a key post-translational modification (PTM), plays a crucial role in biased signaling by altering receptor conformation and ligand-binding affinity, thereby affecting the outcome of biased signaling. Chemokine receptors like CXCR4, which are often overexpressed and heavily glycosylated in AML, serve as targets for therapeutic intervention. By externally inducing or inhibiting specific PTMs, it may be possible to further refine therapeutic strategies, unlocking new possibilities for developing more effective and less toxic treatments. This review highlights the importance of understanding the dynamic relationship between glycosylation and biased signaling in AML, which is essential for the development of more effective treatments and overcoming drug resistance, ultimately leading to better patient outcomes.
Collapse
Affiliation(s)
- Tomasz Pienkowski
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Aleksandra Golonko
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland; Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Wiejska 45 E, 15-351 Bialystok, Poland; Waclaw Dabrowski Institute of Agricultural and Food Biotechnology State Research Institute, Rakowiecka 36, 02-532 Warsaw, Poland.
| | - Lukasz Bolkun
- Department of Hematology, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Katarzyna Wawrzak-Pienkowska
- Department of Gastroenterology, Hepatology and Internal Diseases, Voivodeship Hospital in Bialystok, 15-278 Bialystok, Poland; Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Lukasz Szczerbinski
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Adam Kretowski
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland; Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Michal Ciborowski
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Wlodzimierz Lewandowski
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Wiejska 45 E, 15-351 Bialystok, Poland
| | - Waldemar Priebe
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1901 East Rd., Houston, TX 77054, USA
| | - Renata Swislocka
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Wiejska 45 E, 15-351 Bialystok, Poland
| |
Collapse
|
2
|
Ji RL, Tao YX. Biased signaling in drug discovery and precision medicine. Pharmacol Ther 2025; 268:108804. [PMID: 39904401 DOI: 10.1016/j.pharmthera.2025.108804] [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: 10/28/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Receptors are crucial for converting chemical and environmental signals into cellular responses, making them prime targets in drug discovery, with about 70% of drugs targeting these receptors. Biased signaling, or functional selectivity, has revolutionized drug development by enabling precise modulation of receptor signaling pathways. This concept is more firmly established in G protein-coupled receptor and has now been applied to other receptor types, including ion channels, receptor tyrosine kinases, and nuclear receptors. Advances in structural biology have further refined our understanding of biased signaling. This targeted approach enhances therapeutic efficacy and potentially reduces side effects. Numerous biased drugs have been developed and approved as therapeutics to treat various diseases, demonstrating their significant therapeutic potential. This review provides a comprehensive overview of biased signaling in drug discovery and disease treatment, highlighting recent advancements and exploring the therapeutic potential of these innovative modulators across various diseases.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
3
|
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
|
4
|
Sathoria P, Chuphal B, Rai U, Roy B. Molecular cloning, characterization and 3D modelling of spotted snakehead fbn1 C-terminal region encoding asprosin and expression analysis of fbn1. Sci Rep 2023; 13:4470. [PMID: 36934166 PMCID: PMC10024713 DOI: 10.1038/s41598-023-31271-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/09/2023] [Indexed: 03/20/2023] Open
Abstract
The FBN1 gene encodes profibrillin protein that is cleaved by the enzyme furin to release fibrillin-1 and a glucogenic hormone, asprosin. Asprosin is implicated in diverse metabolic functions as well as pathological conditions in mammals. However, till date, there are no studies on asprosin in any non-mammalian vertebrate. In this study, we have retrieved the spotted snakehead Channa punctata fbn1 gene (ss fbn1) from the testicular transcriptome data and validated it. The transcript is predicted to encode 2817 amino acid long putative profibrillin protein. Amino acid sequence alignment of deduced ss profibrillin with human profibrillin revealed that the furin cleavage site in profibrillin is well conserved in C. punctata. Further, differential expression of ss fbn1 was observed in various tissues with the highest expression in gonads. Prominent expression of furin was also observed in the gonads suggesting the possibility of proteolytic cleavage of profibrillin protein and secretion of asprosin in C. punctata. In addition, the C-terminal of the fbn1 gene of C. punctata that codes for asprosin protein has been cloned. Using in silico approach, physicochemical properties of the putative ss asprosin were characterized and post-translational changes were predicted. The putative ss asprosin protein sequence is predicted to consist of 142 amino acid residues, with conserved glycosylation sites. Further, the 3D model of ss asprosin was predicted followed by MD (molecular dynamics) simulation for energy minimization. Thus, the current study, for the first time in non-mammalian vertebrates, predicts and characterizes the novel protein asprosin using in silico approach.
Collapse
Affiliation(s)
- Priyanka Sathoria
- Department of Zoology, Maitreyi College, University of Delhi, Chanakyapuri, Delhi, 110021, India
| | - Bhawna Chuphal
- Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Umesh Rai
- University of Jammu, Jammu, Jammu and Kashmir, 180006, India
| | - Brototi Roy
- Department of Zoology, Maitreyi College, University of Delhi, Chanakyapuri, Delhi, 110021, India.
| |
Collapse
|
5
|
Rosenfield RL, Cooke DW, Radovick S. Puberty in the Female and Its Disorders. SPERLING PEDIATRIC ENDOCRINOLOGY 2021:528-626. [DOI: 10.1016/b978-0-323-62520-3.00016-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
6
|
Yang LK, Hou ZS, Tao YX. Biased signaling in naturally occurring mutations of G protein-coupled receptors associated with diverse human diseases. Biochim Biophys Acta Mol Basis Dis 2021; 1867:165973. [PMID: 32949766 PMCID: PMC7722056 DOI: 10.1016/j.bbadis.2020.165973] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
G protein-coupled receptors (GPCRs) play critical roles in transmitting a variety of extracellular signals into the cells and regulate diverse physiological functions. Naturally occurring mutations that result in dysfunctions of GPCRs have been known as the causes of numerous diseases. Significant progresses have been made in elucidating the pathophysiology of diseases caused by mutations. The multiple intracellular signaling pathways, such as G protein-dependent and β-arrestin-dependent signaling, in conjunction with recent advances on biased agonism, have broadened the view on the molecular mechanism of disease pathogenesis. This review aims to briefly discuss biased agonism of GPCRs (biased ligands and biased receptors), summarize the naturally occurring GPCR mutations that cause biased signaling, and propose the potential pathophysiological relevance of biased mutant GPCRs associated with various endocrine diseases.
Collapse
Affiliation(s)
- Li-Kun Yang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Zhi-Shuai Hou
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
7
|
Sayers NS, Anujan P, Yu HN, Palmer SS, Nautiyal J, Franks S, Hanyaloglu AC. Follicle-Stimulating Hormone Induces Lipid Droplets via Gαi/o and β-Arrestin in an Endometrial Cancer Cell Line. Front Endocrinol (Lausanne) 2021; 12:798866. [PMID: 35185785 PMCID: PMC8850301 DOI: 10.3389/fendo.2021.798866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/20/2021] [Indexed: 12/04/2022] Open
Abstract
Follicle-stimulating hormone (FSH) and its G protein-coupled receptor, FSHR, represents a paradigm for receptor signaling systems that activate multiple and complex pathways. Classically, FSHR activates Gαs to increase intracellular levels of cAMP, but its ability to activate other G proteins, and β-arrestin-mediated signaling is well documented in many different cell systems. The pleiotropic signal capacity of FSHR offers a mechanism for how FSH drives multiple and dynamic downstream functions in both gonadal and non-gonadal cell types, including distinct diseases, and how signal bias may be achieved at a pharmacological and cell system-specific manner. In this study, we identify an additional mechanism of FSH-mediated signaling and downstream function in the endometrial adenocarcinoma Ishikawa cell line. While FSH did not induce increases in cAMP levels, this hormone potently activated pertussis toxin sensitive Gαi/o signaling. A selective allosteric FSHR ligand, B3, also activated Gαi/o signaling in these cells, supporting a role for receptor-mediated activation despite the low levels of FSHR mRNA. The low expression levels may attribute to the lack of Gαs/cAMP signaling as increasing FSHR expression resulted in FSH-mediated activation of the Gαs pathway. Unlike prior reports for FSH-mediated Gαs/cAMP signaling, FSH-mediated Gαi/o signaling was not affected by inhibition of dynamin-dependent receptor internalization. While chronic FSH did not alter cell viability, FSH was able to increase lipid droplet size. The β-arrestins are key adaptor proteins known to regulate FSHR signaling. Indeed, a rapid, FSH-dependent increase in interactions between β-arrestin1 and Gαi1 was observed via NanoBiT complementation in Ishikawa cells. Furthermore, both inhibition of Gαi/o signaling and siRNA knockdown of β-arrestin 1/2 significantly reduced FSH-induced lipid droplet accumulation, implying a role for a Gαi/o/β-arrestin complex in FSH functions in this cell type. As FSH/FSHR has been implicated in distinct hormone-dependent cancers, including endometrial cancer, analysis of the cancer genome database from 575 human endometrial adenocarcinoma tumors revealed that a subpopulation of samples expressed FSHR. Overall, this study highlights a novel mechanism for FSHR signal pleiotropy that may be exploited for future personalized therapeutic approaches.
Collapse
Affiliation(s)
- Niamh S. Sayers
- Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Priyanka Anujan
- Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Henry N. Yu
- CanWell Pharma Inc., Wellesley, MA, United States
| | - Stephen S. Palmer
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Jaya Nautiyal
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Stephen Franks
- Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| | - Aylin C. Hanyaloglu
- Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
- *Correspondence: Aylin C. Hanyaloglu,
| |
Collapse
|
8
|
van Gastel J, Leysen H, Boddaert J, Vangenechten L, Luttrell LM, Martin B, Maudsley S. Aging-related modifications to G protein-coupled receptor signaling diversity. Pharmacol Ther 2020; 223:107793. [PMID: 33316288 DOI: 10.1016/j.pharmthera.2020.107793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Aging is a highly complex molecular process, affecting nearly all tissue systems in humans and is the highest risk factor in developing neurodegenerative disorders such as Alzheimer's and Parkinson's disease, cardiovascular disease and Type 2 diabetes mellitus. The intense complexity of the aging process creates an incentive to develop more specific drugs that attenuate or even reverse some of the features of premature aging. As our current pharmacopeia is dominated by therapeutics that target members of the G protein-coupled receptor (GPCR) superfamily it may be prudent to search for effective anti-aging therapeutics in this fertile domain. Since the first demonstration of GPCR-based β-arrestin signaling, it has become clear that an enhanced appreciation of GPCR signaling diversity may facilitate the creation of therapeutics with selective signaling activities. Such 'biased' ligand signaling profiles can be effectively investigated using both standard molecular biological techniques as well as high-dimensionality data analyses. Through a more nuanced appreciation of the quantitative nature across the multiple dimensions of signaling bias that drugs possess, researchers may be able to further refine the efficacy of GPCR modulators to impact the complex aberrations that constitute the aging process. Identifying novel effector profiles could expand the effective pharmacopeia and assist in the design of precision medicines. This review discusses potential non-G protein effectors, and specifically their potential therapeutic suitability in aging and age-related disorders.
Collapse
Affiliation(s)
- Jaana van Gastel
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Hanne Leysen
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Jan Boddaert
- Molecular Pathology Group, Faculty of Medicine and Health Sciences, Laboratory of Cell Biology and Histology, Antwerp, Belgium
| | - Laura Vangenechten
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Louis M Luttrell
- Division of Endocrinology, Diabetes & Medical Genetics, Medical University of South Carolina, USA
| | - Bronwen Martin
- Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
9
|
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
|
10
|
Casarini L, Crépieux P, Reiter E, Lazzaretti C, Paradiso E, Rochira V, Brigante G, Santi D, Simoni M. FSH for the Treatment of Male Infertility. Int J Mol Sci 2020; 21:ijms21072270. [PMID: 32218314 PMCID: PMC7177393 DOI: 10.3390/ijms21072270] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 12/11/2022] Open
Abstract
Follicle-stimulating hormone (FSH) supports spermatogenesis acting via its receptor (FSHR), which activates trophic effects in gonadal Sertoli cells. These pathways are targeted by hormonal drugs used for clinical treatment of infertile men, mainly belonging to sub-groups defined as hypogonadotropic hypogonadism or idiopathic infertility. While, in the first case, fertility may be efficiently restored by specific treatments, such as pulsatile gonadotropin releasing hormone (GnRH) or choriogonadotropin (hCG) alone or in combination with FSH, less is known about the efficacy of FSH in supporting the treatment of male idiopathic infertility. This review focuses on the role of FSH in the clinical approach to male reproduction, addressing the state-of-the-art from the little data available and discussing the pharmacological evidence. New compounds, such as allosteric ligands, dually active, chimeric gonadotropins and immunoglobulins, may represent interesting avenues for future personalized, pharmacological approaches to male infertility.
Collapse
Affiliation(s)
- Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Correspondence: ; Tel.: +39-0593961705; Fax: +39-0593962018
| | - 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), Institut Français du Cheval et de l’Equitation (IFCE), Université de Tours, 37380 Nouzilly, France; (P.C.); (E.R.)
| | - Eric 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), Institut Français du Cheval et de l’Equitation (IFCE), Université de Tours, 37380 Nouzilly, France; (P.C.); (E.R.)
| | - Clara Lazzaretti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Elia Paradiso
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Vincenzo Rochira
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Via P. Giardini 1355, 41126 Modena, Italy
| | - Giulia Brigante
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Via P. Giardini 1355, 41126 Modena, Italy
| | - Daniele Santi
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Via P. Giardini 1355, 41126 Modena, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via P. Giardini 1355, 41126 Modena, Italy; (C.L.); (E.P.); (V.R.); (G.B.); (D.S.); (M.S.)
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- 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), Institut Français du Cheval et de l’Equitation (IFCE), Université de Tours, 37380 Nouzilly, France; (P.C.); (E.R.)
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Via P. Giardini 1355, 41126 Modena, Italy
| |
Collapse
|
11
|
Gao Y, Jing Q, Huang B, Jia Y. Molecular cloning, characterization, and mRNA expression of gonadotropins during larval development in turbot (Scophthalmus maximus). FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:1697-1707. [PMID: 31098916 DOI: 10.1007/s10695-019-00656-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 05/06/2019] [Indexed: 06/09/2023]
Abstract
Gonadotropins (GtHs) play a pivotal role in regulating the reproductive axis and puberty. In this study, full-length sequences coding for common glycoprotein α subunit (CGα) and luteinizing hormone β (LHβ) were isolated from female turbot (Scophthalmus maximus) pituitary by homology cloning and a strategy based on rapid amplification of cDNA end-polymerase chain reaction. Results showed that the two cDNAs consisted of 669 and 660 nucleotides encoding 129 and 139 amino acids, respectively. CGα and LHβ manifested typical characteristics of glycoprotein hormones, high homologies with the corresponding sequences of available teleosts, and high homology with that of Hippoglossus hippoglossus. CGα, FSHβ, and LHβ mRNAs were abundant in the pituitary, but less expressed in extra-pituitary tissues. The cgα, fshβ, and lhβ were detected at 1-day post-hatching (dph) and peaked simultaneously at early-metamorphosis (22 dph). cgα and fshβ mRNA levels were significantly increased at pre-metamorphosis, peaked in early metamorphosis, and then gradually decreased until metamorphosis was completed. Conversely, lhβ mRNA levels gradually decreased at pre-metamorphosis, dramatically peaked at early metamorphosis, and then decreased during metamorphosis. In addition, the mRNA levels of cgα were significantly higher than those of fshβ and lhβ during turbot larval metamorphic development, whereas no significant difference was found between fshβ and lhβ. These results suggested (i) an early activation of the GtHs system after hatching, which was the highest expression at early metamorphosis, and (ii) FSHβ and LHβ were together involved in the establishment of the reproductive axis during larval development in turbot. These findings contribute to further understanding the potential roles of GtHs during fish larval development.
Collapse
Affiliation(s)
- Yunhong Gao
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Qiqi Jing
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Bin Huang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Yudong Jia
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao, 266071, People's Republic of China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
12
|
Bousfield GR, Harvey DJ. Follicle-Stimulating Hormone Glycobiology. Endocrinology 2019; 160:1515-1535. [PMID: 31127275 PMCID: PMC6534497 DOI: 10.1210/en.2019-00001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/16/2019] [Indexed: 01/13/2023]
Abstract
FSH glycosylation varies in two functionally important aspects: microheterogeneity, resulting from oligosaccharide structure variation, and macroheterogeneity, arising from partial FSHβ subunit glycosylation. Although advances in mass spectrometry permit extensive characterization of FSH glycan populations, microheterogeneity remains difficult to illustrate, and comparisons between different studies are challenging because no standard format exists for rendering oligosaccharide structures. FSH microheterogeneity is illustrated using a consistent glycan diagram format to illustrate the large array of structures associated with one hormone. This is extended to commercially available recombinant FSH preparations, which exhibit greatly reduced microheterogeneity at three of four glycosylation sites. Macroheterogeneity is demonstrated by electrophoretic mobility shifts due to the absence of FSHβ glycans that can be assessed by Western blotting of immunopurified FSH. Initially, macroheterogeneity was hoped to matter more than microheterogeneity. However, it now appears that both forms of carbohydrate heterogeneity have to be taken into consideration. FSH glycosylation can reduce its apparent affinity for its cognate receptor by delaying initial interaction with the receptor and limiting access to all of the available binding sites. This is followed by impaired cellular signaling responses that may be related to reduced receptor occupancy or biased signaling. To resolve these alternatives, well-characterized FSH glycoform preparations are necessary.
Collapse
Affiliation(s)
- George R Bousfield
- Department of Biological Sciences, Wichita State University, Wichita, Kansas
- Correspondence: George R. Bousfield, PhD, Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, Kansas 67260. E-mail: ; or David J. Harvey, DSc, Target Discovery Institute, Nuffield Department of Medicine, University of Oxford. Roosevelt Drive, Oxford OX3 7FZ, United Kingdom. E-mail:
| | - David J Harvey
- Target Discovery Institute, Nuffield Department of Medicine, Oxford University, Oxford, United Kingdom
- Correspondence: George R. Bousfield, PhD, Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, Kansas 67260. E-mail: ; or David J. Harvey, DSc, Target Discovery Institute, Nuffield Department of Medicine, University of Oxford. Roosevelt Drive, Oxford OX3 7FZ, United Kingdom. E-mail:
| |
Collapse
|
13
|
Anderson RC, Newton CL, Anderson RA, Millar RP. Gonadotropins and Their Analogs: Current and Potential Clinical Applications. Endocr Rev 2018; 39:911-937. [PMID: 29982442 DOI: 10.1210/er.2018-00052] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022]
Abstract
The gonadotropin receptors LH receptor and FSH receptor play a central role in governing reproductive competency/fertility. Gonadotropin hormone analogs have been used clinically for decades in assisted reproductive therapies and in the treatment of various infertility disorders. Though these treatments are effective, the clinical protocols demand multiple injections, and the hormone preparations can lack uniformity and stability. The past two decades have seen a drive to develop chimeric and modified peptide analogs with more desirable pharmacokinetic profiles, with some displaying clinical efficacy, such as corifollitropin alfa, which is now in clinical use. More recently, low-molecular-weight, orally active molecules with activity at gonadotropin receptors have been developed. Some have excellent characteristics in animals and in human studies but have not reached the market-largely as a result of acquisitions by large pharma. Nonetheless, such molecules have the potential to mitigate risks currently associated with gonadotropin-based fertility treatments, such as ovarian hyperstimulation syndrome and the demands of injection-based therapies. There is also scope for novel use beyond the current remit of gonadotropin analogs in fertility treatments, including application as novel contraceptives; in the treatment of polycystic ovary syndrome; in the restoration of function to inactivating mutations of gonadotropin receptors; in the treatment of ovarian and prostate cancers; and in the prevention of bone loss and weight gain in postmenopausal women. Here we review the properties and clinical application of current gonadotropin preparations and their analogs, as well as the development of novel orally active, small-molecule nonpeptide analogs.
Collapse
Affiliation(s)
- Ross C Anderson
- Centre for Neuroendocrinology, University of Pretoria, Pretoria, South Africa.,Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Claire L Newton
- Centre for Neuroendocrinology, University of Pretoria, Pretoria, South Africa.,Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert P Millar
- Centre for Neuroendocrinology, University of Pretoria, Pretoria, South Africa.,Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
14
|
Ulloa-Aguirre A, Reiter E, Crépieux P. FSH Receptor Signaling: Complexity of Interactions and Signal Diversity. Endocrinology 2018; 159:3020-3035. [PMID: 29982321 DOI: 10.1210/en.2018-00452] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 06/27/2018] [Indexed: 12/20/2022]
Abstract
FSH is synthesized in the pituitary by gonadotrope cells. By binding to and interacting with its cognate receptor [FSH receptor (FSHR)] in the gonads, this gonadotropin plays a key role in the control of gonadal function and reproduction. Upon activation, the FSHR undergoes conformational changes leading to transduction of intracellular signals, including dissociation of G protein complexes into components and activation of several associated interacting partners, which concertedly regulate downstream effectors. The canonical Gs/cAMP/protein kinase A pathway, considered for a long time as the sole effector of FSHR-mediated signaling, is now viewed as one of several mechanisms employed by this receptor to transduce intracellular signals in response to the FSH stimulus. This complex network of signaling pathways allows for a fine-tuning regulation of the gonadotropic stimulus, where activation/inhibition of its multiple components vary depending on the cell context, cell developmental stage, and concentration of associated receptors and corresponding ligands. Activation of these multiple signaling modules eventually converge to the hormone-integrated biological response, including survival, proliferation and differentiation of target cells, synthesis and secretion of paracrine/autocrine regulators, and, at the molecular level, functional selectivity and differential gene expression. In this mini-review, we discuss the complexity of FSHR-mediated intracellular signals activated in response to ligand stimulation. A better understanding of the signaling pathways involved in FSH action might potentially influence the development of new therapeutic strategies for reproductive disorders.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Eric Reiter
- Biology and Bioinformatics of Signaling Systems Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Nouzilly, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Nouzilly, France
- Université François Rabelais, Nouzilly, France
| | - Pascale Crépieux
- Biology and Bioinformatics of Signaling Systems Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Nouzilly, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Nouzilly, France
- Université François Rabelais, Nouzilly, France
| |
Collapse
|
15
|
Abstract
Gonadotropin receptors include the follicle stimulating hormone receptor (FSHR) and the luteinizing hormone/choriogonadotropin receptor (LHCGR), both belong to the G protein-coupled receptor (GPCR) superfamily and are essential to reproduction. FSHR is activated by follicle stimulating hormone (FSH) while LHCGR is activated by either luteinizing hormone (LH) or choriogonadotropin (CG). Upon ligand binding, gonadotropin receptors undergo conformational changes that lead to the activation of the heterotrimeric G protein, resulting in the production of different second messengers. Gonadotropin receptors can also recruit and bind β-arrestins. This particular class of scaffold proteins were initially identified to mediate GPCRs desensitization and recycling, but it is now well established that β-arrestins can also initiate Gs-independent signaling by assembling signaling modules. Furthermore, new advances in structural biology and biophysical techniques have revealed novel activation mechanisms allowing β-arrestins and G proteins to control signaling in time and space. The ability of different ligands to preferentially elicit G- or β-arrestin-mediated signaling is known as functional selectivity or biased signaling. This new concept has switched the view of pharmacology efficacy from monodimensional to multidimensional. Biased signaling offers the possibility to separate therapeutic benefits of a drug from its adverse effects. The proof of concept that gonadotropin receptors can be subjected to biased signaling is now established. The challenge will now be the design of molecules that can specifically activate beneficial signaling pathway at gonadotropin receptors while reducing or abolishing those leading to side effects. Such strategy could for instance lead to improved treatments for infertility.
Collapse
Affiliation(s)
| | - Eric Reiter
- PCR, INRA, CNRS, IFCE, Université de Tours, Nouzilly, France -
| |
Collapse
|
16
|
Nataraja S, Sriraman V, Palmer S. Allosteric Regulation of the Follicle-Stimulating Hormone Receptor. Endocrinology 2018; 159:2704-2716. [PMID: 29800292 DOI: 10.1210/en.2018-00317] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 05/16/2018] [Indexed: 01/08/2023]
Abstract
Follicle-stimulating hormone receptor (FSHR) belongs to the leucine-rich repeat family of the G protein-coupled receptor (LGR), which includes the glycoprotein hormone receptors luteinizing hormone receptor, thyrotropin receptor, and other LGRs 4, 5, 6, and 7. FSH is the key regulator of folliculogenesis in females and spermatogenesis in males. FSH elicits its physiological response through its cognate receptor on the cell surface. Binding of the hormone FSH to its receptor FSHR brings about conformational changes in the receptor that are transduced through the transmembrane domain to the intracellular region, where the downstream effector interaction takes place, leading to activation of the downstream signaling cascade. Identification of small molecules that could activate or antagonize FSHR provided interesting tools to study the signal transduction mechanism of the receptor. However, because of the nature of the ligand-receptor interaction of FSH-FSHR, which contains multiple sites in the extracellular binding domain, most of the small-molecule modulators of FSHR are unable to bind to the orthosteric site of the receptors. Rather they modulate receptor activation through allosteric sites in the transmembrane region. This review will discuss allosteric modulation of FSHR primarily through the discovery of small-molecule modulators, focusing on current data on the status of development and the utility of these as tools to better understand signaling mechanisms.
Collapse
|
17
|
Das N, Kumar TR. Molecular regulation of follicle-stimulating hormone synthesis, secretion and action. J Mol Endocrinol 2018; 60:R131-R155. [PMID: 29437880 PMCID: PMC5851872 DOI: 10.1530/jme-17-0308] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 02/07/2018] [Indexed: 12/11/2022]
Abstract
Follicle-stimulating hormone (FSH) plays fundamental roles in male and female fertility. FSH is a heterodimeric glycoprotein expressed by gonadotrophs in the anterior pituitary. The hormone-specific FSHβ-subunit is non-covalently associated with the common α-subunit that is also present in the luteinizing hormone (LH), another gonadotrophic hormone secreted by gonadotrophs and thyroid-stimulating hormone (TSH) secreted by thyrotrophs. Several decades of research led to the purification, structural characterization and physiological regulation of FSH in a variety of species including humans. With the advent of molecular tools, availability of immortalized gonadotroph cell lines and genetically modified mouse models, our knowledge on molecular mechanisms of FSH regulation has tremendously expanded. Several key players that regulate FSH synthesis, sorting, secretion and action in gonads and extragonadal tissues have been identified in a physiological setting. Novel post-transcriptional and post-translational regulatory mechanisms have also been identified that provide additional layers of regulation mediating FSH homeostasis. Recombinant human FSH analogs hold promise for a variety of clinical applications, whereas blocking antibodies against FSH may prove efficacious for preventing age-dependent bone loss and adiposity. It is anticipated that several exciting new discoveries uncovering all aspects of FSH biology will soon be forthcoming.
Collapse
Affiliation(s)
- Nandana Das
- Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, U.S.A
| | - T. Rajendra Kumar
- Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, U.S.A
- Division of Reproductive Endocrinology and Infertility, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, U.S.A
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, U.S.A
- Author for Correspondence: T. Rajendra Kumar, PhD, Edgar L. and Patricia M. Makowski Professor, Associate Vice-Chair of Research, Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Mail Stop 8613, Research Complex 2, Room # 15-3000B, 12700 E. 19th Avenue, Aurora, CO 80045, USA, Tel: 303-724-8689,
| |
Collapse
|
18
|
Abstract
G protein-coupled receptors (GPCRs) are the largest class of receptors in the human genome and some of the most common drug targets. It is now well established that GPCRs can signal through multiple transducers, including heterotrimeric G proteins, GPCR kinases and β-arrestins. While these signalling pathways can be activated or blocked by 'balanced' agonists or antagonists, they can also be selectively activated in a 'biased' response. Biased responses can be induced by biased ligands, biased receptors or system bias, any of which can result in preferential signalling through G proteins or β-arrestins. At many GPCRs, signalling events mediated by G proteins and β-arrestins have been shown to have distinct biochemical and physiological actions from one another, and an accurate evaluation of biased signalling from pharmacology through physiology is crucial for preclinical drug development. Recent structural studies have provided snapshots of GPCR-transducer complexes, which should aid in the structure-based design of novel biased therapies. Our understanding of GPCRs has evolved from that of two-state, on-and-off switches to that of multistate allosteric microprocessors, in which biased ligands transmit distinct structural information that is processed into distinct biological outputs. The development of biased ligands as therapeutics heralds an era of increased drug efficacy with reduced drug side effects.
Collapse
|
19
|
Follicle-Stimulating Hormone Receptor: Advances and Remaining Challenges. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:1-58. [DOI: 10.1016/bs.ircmb.2018.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
20
|
Andreone L, Ambao V, Pellizzari EH, Loreti N, Cigorraga SB, Campo S. Role of FSH glycan structure in the regulation of Sertoli cell inhibin production. Reproduction 2017; 154:711-721. [PMID: 28855248 DOI: 10.1530/rep-17-0393] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/21/2017] [Accepted: 08/30/2017] [Indexed: 01/16/2023]
Abstract
Variations in follicle-stimulating hormone (FSH) carbohydrate composition and structure are associated with important structural and functional changes in Sertoli cells (SCs) during sexual maturation. The aim of the present study was to investigate the impact of FSH oligosaccharide structure and its interaction with gonadal factors on the regulation of monomeric and dimeric inhibin production at different maturation stages of the SC. Recombinant human FSH (rhFSH) glycosylation variants were isolated according to their sialylation degree (AC and BA) and complexity of oligosaccharides (CO and HY). Native rhFSH stimulated inhibin α-subunit (Pro-αC) but did not show any effect on inhibin B (INHB) production in immature SCs isolated from 8-day-old rats. Activin A stimulated INHB and had a synergistic effect on FSH to stimulate Pro-αC. The less acidic/sialylated rhFSH charge analogues, BA, were the only charge analogue mix that stimulated INHB as well as the most potent stimulus for Pro-αC production. Native rhFSH stimulated both Pro-αC and INHB in SCs at a more advanced maturation stage, isolated from 20-day-old rats. In these cells, all rhFSH glycosylation variants increased INHB and Pro-αC production, even in the presence of growth factors. The BA preparation exerted a more marked stimulatory effect on INHB and Pro-αC than the AC. Glycoforms bearing high mannose and hybrid-type oligosaccharides, HY, stimulated INHB and Pro-αC more effectively than those bearing complex oligosaccharides, CO, even in the presence of gonadal growth factors. These findings demonstrate the modulatory effect of FSH oligosaccharide structure on the regulation of inhibin production in the male gonad.
Collapse
Affiliation(s)
| | - Verónica Ambao
- Centro de Investigaciones Endocrinológicas 'Dr. Cesar Bergadá' (CEDIE-CONICET) Hospital de Niños 'R. Gutiérrez', Buenos Aires, Argentina
| | - Eliana H Pellizzari
- Centro de Investigaciones Endocrinológicas 'Dr. Cesar Bergadá' (CEDIE-CONICET) Hospital de Niños 'R. Gutiérrez', Buenos Aires, Argentina
| | - Nazareth Loreti
- Centro de Investigaciones Endocrinológicas 'Dr. Cesar Bergadá' (CEDIE-CONICET) Hospital de Niños 'R. Gutiérrez', Buenos Aires, Argentina
| | - Selva B Cigorraga
- Centro de Investigaciones Endocrinológicas 'Dr. Cesar Bergadá' (CEDIE-CONICET) Hospital de Niños 'R. Gutiérrez', Buenos Aires, Argentina
| | - Stella Campo
- Centro de Investigaciones Endocrinológicas 'Dr. Cesar Bergadá' (CEDIE-CONICET) Hospital de Niños 'R. Gutiérrez', Buenos Aires, Argentina
| |
Collapse
|
21
|
Ulloa-Aguirre A, Zariñán T. The Follitropin Receptor: Matching Structure and Function. Mol Pharmacol 2016; 90:596-608. [PMID: 27382014 DOI: 10.1124/mol.116.104398] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/28/2016] [Indexed: 02/14/2025] Open
Abstract
Follitropin, or follicle-stimulating hormone (FSH) receptor (FSHR), is a G protein-coupled receptor belonging to the glycoprotein hormone receptor family that plays an essential role in reproduction. Although its primary location is the gonad, the FSHR has also been reported in extragonadal tissues including bone, placenta, endometrium, liver, and blood vessels from a number of malignant tumors. The recently resolved crystal structure of FSH bound to the entire FSHR ectodomain has been instrumental in more clearly defining the role of this domain in ligand binding and receptor activation. Biochemical, biophysical, and structural data also indicate that the FSHR exists as a higher order structure and that it may heterodimerize with its closely related receptor, the luteinizing hormone receptor; this association may have physiologic implications during ovarian follicle maturation given that both receptors may simultaneously coexist in the same cell. FSHR heterodimerization is unique to the ovary because in the testes, gonadotropin receptors are expressed in separate compartments. FSHR self-association appears to be required for receptor coupling to multiple effectors and adaptors, for the activation of multiple signaling pathways and the transduction of asymmetric signaling, and for negative and positive receptor cooperativity. It also provides a mechanism through which the glycosylation variants of FSH may exert distinct and differential effects at the target cell level. Given its importance in regulating activation of distinct signaling pathways, functional selectivity at the FSHR is briefly discussed, as well as the potential implications of this particular functional feature on the design of new pharmacological therapies in reproduction.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Research Support Network, National University of Mexico and National Institutes of Health, Mexico City, Mexico
| | - Teresa Zariñán
- Research Support Network, National University of Mexico and National Institutes of Health, Mexico City, Mexico
| |
Collapse
|
22
|
Affiliation(s)
- Johan Smitz
- Laboratory of Hormonology and Tumormarkers, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Claudio Wolfenson
- Production and Regulatory Affairs, Instituto Massone SA, Buenos Aires, Argentina
| | | | - Jane Ruman
- Department of Reproductive Health, Ferring Pharmaceuticals, Inc, Parsippany, NJ, USA
| |
Collapse
|
23
|
N-linked glycosylation of protease-activated receptor-1 at extracellular loop 2 regulates G-protein signaling bias. Proc Natl Acad Sci U S A 2015; 112:E3600-8. [PMID: 26100877 DOI: 10.1073/pnas.1508838112] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Protease-activated receptor-1 (PAR1) is a G-protein-coupled receptor (GPCR) for the coagulant protease thrombin. Similar to other GPCRs, PAR1 is promiscuous and couples to multiple heterotrimeric G-protein subtypes in the same cell and promotes diverse cellular responses. The molecular mechanism by which activation of a given GPCR with the same ligand permits coupling to multiple G-protein subtypes is unclear. Here, we report that N-linked glycosylation of PAR1 at extracellular loop 2 (ECL2) controls G12/13 versus Gq coupling specificity in response to thrombin stimulation. A PAR1 mutant deficient in glycosylation at ECL2 was more effective at stimulating Gq-mediated phosphoinositide signaling compared with glycosylated wildtype receptor. In contrast, wildtype PAR1 displayed a greater efficacy at G12/13-dependent RhoA activation compared with mutant receptor lacking glycosylation at ECL2. Endogenous PAR1 rendered deficient in glycosylation using tunicamycin, a glycoprotein synthesis inhibitor, also exhibited increased PI signaling and diminished RhoA activation opposite to native receptor. Remarkably, PAR1 wildtype and glycosylation-deficient mutant were equally effective at coupling to Gi and β-arrestin-1. Consistent with preferential G12/13 coupling, thrombin-stimulated PAR1 wildtype strongly induced RhoA-mediated stress fiber formation compared with mutant receptor. In striking contrast, glycosylation-deficient PAR1 was more effective at increasing cellular proliferation, associated with Gq signaling, than wildtype receptor. These studies suggest that N-linked glycosylation at ECL2 contributes to the stabilization of an active PAR1 state that preferentially couples to G12/13 versus Gq and defines a previously unidentified function for N-linked glycosylation of GPCRs in regulating G-protein signaling bias.
Collapse
|
24
|
Asraf H, Amsterdam A, Ben-Menahem D. Modulation of the steroidogenic related activity according to the design of single-chain bovine FSH analogs. Gen Comp Endocrinol 2015; 216:171-81. [PMID: 25863346 DOI: 10.1016/j.ygcen.2015.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/22/2015] [Accepted: 04/05/2015] [Indexed: 10/23/2022]
Abstract
Single-chain (SC) gonadotropins have been genetically engineered to increase the repertoire of analogs for potential use in humans and domestic animals. The major aim of the current study was to examine the steroidogenic related activity of SC FSH analogs carrying structural differences. To address this issue, we designed and expressed three SC bovine FSH analogs in CHO cells: (i) FSHβα in which the tethered subunit domains are linked in tandem; (ii) FSHβCTPα that contains the carboxy terminal peptide (CTP) of the human choriogonadotropin (hCG) β subunit as a spacer, and (iii) FSHβboCTPα in which the linker is derived from a CTP-like sequence (boCTP) decoded from the bovine LHβ DNA. The data suggested that the secretion efficiency of these variants from the transfected cells was unaffected by the presence or absence of the CTP linker, N-glycans were attached to the analogs and the hCGβ-CTP domain in the FSHβCTPα variant was O-glycosylated. In a rat immortalized granulosa cell bioassay the potency of the three variants towards progesterone secretion varied. In immature mice, the analogs increased the ovary weight and induced StAR, Cyp11a (P450scc), Cyp17 (P450c17) and Cyp19 (P450aromatase) transcripts. However, the dose dependence and amplitude of these transcript levels differed in response to FSHβα, FSHβboCTPα and FSHβCTPα. Collectively, these data suggest that the design of the FSH analog can modulate the bioactivity in vitro and in vivo. A systematic analysis of receptor activation with ligands carrying structural differences may identify new regulatory factor/s involved in the pleiotropic FSH activity.
Collapse
Affiliation(s)
- Hila Asraf
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Abraham Amsterdam
- Department of Molecular and Cellular Biology, Weizmann Institute of Science, Rehovot, Israel
| | - David Ben-Menahem
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
25
|
Hunzicker-Dunn M, Mayo K. Gonadotropin Signaling in the Ovary. KNOBIL AND NEILL'S PHYSIOLOGY OF REPRODUCTION 2015:895-945. [DOI: 10.1016/b978-0-12-397175-3.00020-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
26
|
Du X, Zhou J, Guvench O, Sangiorgi FO, Li X, Zhou N, Xu B. Supramolecular assemblies of a conjugate of nucleobase, amino acids, and saccharide act as agonists for proliferation of embryonic stem cells and development of zygotes. Bioconjug Chem 2014; 25:1031-5. [PMID: 24798034 PMCID: PMC4068792 DOI: 10.1021/bc500187m] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Indexed: 12/14/2022]
Abstract
The synthetic challenges in glycobiology and glycochemistry hamper the development of glycobiomaterials for biomedicine. Here we report the use of molecular self-assembly to sidestep the laborious synthesis of complex glycans for promoting the proliferation of murine embryonic stem (mES) cells. Our study shows that the supramolecular assemblies of a small molecule conjugate of nucleobase, amino acids, and saccharide, as a de novo glycoconjugate, promote the proliferation of mES cells and the development of zygotes into blastocysts of mouse. Molecular engineering confirms that each motif (i.e., adenine, Arg-Gly-Asp (RGD) domain, and glucosamine) is indispensable for the observed activity of the conjugate. As the first example of using assemblies of the molecular conjugates of multiple fundamental biological building blocks to control cell behaviors, this work illustrates an unprecedented approach to use supramolecular assemblies as multifunctional mimics of glycoconjugates.
Collapse
Affiliation(s)
- Xuewen Du
- Department of Chemistry, Brandeis
University, 415 South
Street, Waltham, Massachusetts 02454, United States
- Department of Pharmaceutical
Sciences, University of New England College
of Pharmacy, 716 Stevens
Avenue, Portland, Maine 04102, United States
| | - Jie Zhou
- Department of Chemistry, Brandeis
University, 415 South
Street, Waltham, Massachusetts 02454, United States
- Department of Pharmaceutical
Sciences, University of New England College
of Pharmacy, 716 Stevens
Avenue, Portland, Maine 04102, United States
| | - Olgun Guvench
- Department of Chemistry, Brandeis
University, 415 South
Street, Waltham, Massachusetts 02454, United States
- Department of Pharmaceutical
Sciences, University of New England College
of Pharmacy, 716 Stevens
Avenue, Portland, Maine 04102, United States
| | - Frank O. Sangiorgi
- Department of Chemistry, Brandeis
University, 415 South
Street, Waltham, Massachusetts 02454, United States
- Department of Pharmaceutical
Sciences, University of New England College
of Pharmacy, 716 Stevens
Avenue, Portland, Maine 04102, United States
| | - Xinming Li
- Department of Chemistry, Brandeis
University, 415 South
Street, Waltham, Massachusetts 02454, United States
- Department of Pharmaceutical
Sciences, University of New England College
of Pharmacy, 716 Stevens
Avenue, Portland, Maine 04102, United States
| | - Ning Zhou
- Department of Chemistry, Brandeis
University, 415 South
Street, Waltham, Massachusetts 02454, United States
- Department of Pharmaceutical
Sciences, University of New England College
of Pharmacy, 716 Stevens
Avenue, Portland, Maine 04102, United States
| | | |
Collapse
|
27
|
Landomiel F, Gallay N, Jégot G, Tranchant T, Durand G, Bourquard T, Crépieux P, Poupon A, Reiter E. Biased signalling in follicle stimulating hormone action. Mol Cell Endocrinol 2014; 382:452-459. [PMID: 24121199 DOI: 10.1016/j.mce.2013.09.035] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/26/2013] [Accepted: 09/26/2013] [Indexed: 12/22/2022]
Abstract
Follicle-stimulating hormone (FSH) plays a crucial role in the control of reproduction by specifically binding to and activating a membrane receptor (FSHR) that belongs to the G protein-coupled receptor (GPCR) family. Similar to all GPCRs, FSHR activation mechanisms have generally been viewed as a two-state process connecting a unique FSH-bound active receptor to the Gs/cAMP pathway. Over the last decade, paralleling the breakthroughs that were made in the GPCR field, our understanding of FSH actions at the molecular level has dramatically changed. There are numerous facts indicating that the active FSHR is connected to a complex signalling network rather than the sole Gs/cAMP pathway. Consistently, the FSHR probably exists in equilibrium between multiple conformers, a subset of them being stabilized upon ligand binding. Importantly, the nature of the stabilized conformers of the receptor directly depends on the chemical structure of the ligand bound. This implies that it is possible to selectively control the intracellular signalling pathways activated by using biased ligands. Such biased ligands can be of different nature: small chemical molecules, glycosylation variants of the hormone or antibody/hormone complexes. Likewise, mutations or polymorphisms affecting the FSHR can also lead to stabilization of preferential conformers, hence to selective modulation of signalling pathways. These emerging notions offer a new conceptual framework that could potentially lead to the development of more specific drugs while also improving the way FSHR mutants/variants are functionally characterized.
Collapse
Affiliation(s)
- Flavie Landomiel
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Nathalie Gallay
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Gwenhael Jégot
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Thibaud Tranchant
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Guillaume Durand
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Thomas Bourquard
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Pascale Crépieux
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Anne Poupon
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France
| | - Eric Reiter
- BIOS group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France; CNRS, UMR7247, F-37380 Nouzilly, France; Université François Rabelais, F-37041 Tours, France.
| |
Collapse
|
28
|
Ulloa-Aguirre A, Reiter E, Bousfield G, Dias JA, Huhtaniemi I. Constitutive activity in gonadotropin receptors. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 70:37-80. [PMID: 24931192 DOI: 10.1016/b978-0-12-417197-8.00002-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Constitutively active mutants (CAMs) of gonadotropin receptors are, in general, rare conditions. Luteinizing hormone-choriogonadotropin receptor (LHCGR) CAMs provoke the dramatic phenotype of familial gonadotropin-independent isosexual male-limited precocious puberty, whereas in females, there is not yet any identified phenotype. Only one isolated follicle-stimulating hormone receptor (FSHR) CAM (Asp567Gly) has so far been detected in a single male patient, besides other FSHR weak CAMs linked to pregnancy-associated ovarian hyperstimulation syndrome or to impaired desensitization and internalization. Several animal models have been developed for studying enhanced gonadotropin action; in addition to unraveling valuable new information about the possible phenotypes of isolated FSHR and LHCGR CAMs in women, the information obtained from these mouse models has served multiple translational goals, including the development of new diagnostic and therapeutic targets as well as the prediction of phenotypes for mutations not yet identified in humans. Mutagenesis and computational studies have shed important information on the physiopathogenic mechanisms leading to constitutive activity of gonadotropin receptors; a common feature in these receptor CAMs is the release of stabilizing interhelical interactions between transmembrane domains (TMDs) 3 and 6 leading to an increase, with respect to the wild-type receptor, in the solvent accessibility at the cytosolic extension of TMDs 3, 5, and 6, which involves the highly conserved Glu/Asp-Arg-Tyr/Trp sequence. In this chapter, we summarize the structural features, functional consequences, and mechanisms that lead to constitutive activation of gonadotropin receptor CAMs and provide information on pharmacological approaches that might potentially modulate gonadotropin receptor CAM function.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Studium Consortium for Research and Training in Reproductive Sciences (sCORTS), Tours, France; Research Support Network, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán" and Universidad Nacional Autónoma de México, México D.F., Mexico.
| | - Eric Reiter
- Studium Consortium for Research and Training in Reproductive Sciences (sCORTS), Tours, France; BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais, Tours, France
| | - George Bousfield
- Studium Consortium for Research and Training in Reproductive Sciences (sCORTS), Tours, France; Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
| | - James A Dias
- Studium Consortium for Research and Training in Reproductive Sciences (sCORTS), Tours, France; Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, New York, USA
| | - Ilpo Huhtaniemi
- Studium Consortium for Research and Training in Reproductive Sciences (sCORTS), Tours, France; Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom
| |
Collapse
|
29
|
León K, Gallay N, Poupon A, Reiter E, Dalbies-Tran R, Crepieux P. Integrating microRNAs into the complexity of gonadotropin signaling networks. Front Cell Dev Biol 2013; 1:3. [PMID: 25364708 PMCID: PMC4206998 DOI: 10.3389/fcell.2013.00003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 12/12/2013] [Indexed: 12/13/2022] Open
Abstract
Follicle-stimulating hormone (FSH) is a master endocrine regulator of mammalian reproductive functions. Hence, it is used to stimulate folliculogenesis in assisted reproductive technologies (ART), both in women and in breeding animals. However, the side effects that hormone administration induces in some instances jeopardize the success of ART. Similarly, the luteinizing hormone (LH) is also of paramount importance in the reproductive function because it regulates steroidogenesis and the LH surge is a pre-requisite to ovulation. Gaining knowledge as extensive as possible on gonadotropin-induced biological responses could certainly lead to precise selection of their effects in vivo by the use of selective agonists at the hormone receptors. Hence, over the years, numerous groups have contributed to decipher the cellular events induced by FSH and LH in their gonadal target cells. Although little is known on the effect of gonadotropins on microRNA expression so far, recent data have highlighted that a microRNA regulatory network is likely to superimpose on the signaling protein network. No doubt that this will dramatically alter our current understanding of the gonadotropin-induced signaling networks. This is the topic of this review to present this additional level of complexity within the gonadotropin signaling network, in the context of recent findings on the microRNA machinery in the gonad.
Collapse
Affiliation(s)
- Kelly León
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais Tours, France
| | - Nathalie Gallay
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais Tours, France
| | - Anne Poupon
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais Tours, France
| | - Eric Reiter
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais Tours, France
| | - Rozenn Dalbies-Tran
- BINGO Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais Tours, France
| | - Pascale Crepieux
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR7247, Nouzilly, France; Université François Rabelais Tours, France
| |
Collapse
|
30
|
Polymorphisms in gonadotropin and gonadotropin receptor genes as markers of ovarian reserve and response in in vitro fertilization. Fertil Steril 2013; 99:970-8.e1. [PMID: 23380184 DOI: 10.1016/j.fertnstert.2013.01.086] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 01/04/2013] [Accepted: 01/04/2013] [Indexed: 11/22/2022]
Abstract
Since gonadotropins are the fundamental hormones that control ovarian activity, genetic polymorphisms may alter gonadal responsiveness to glycoproteins; hence they are important regulators of hormone activity at the target level. The establishment of the pool of primordial follicles takes place during fetal life and is mainly under genetic control. Consequently, single nucleotide polymorphisms (SNPs) in gonadotropins and their receptors do not seem to be associated with any significant modification in the endowment of nongrowing follicles in the ovary. Indeed, the age at menopause, a biological characteristic strongly related to ovarian reserve, as well as markers of functional ovarian reserve such as anti-Müllerian hormone and antral follicle count, are not different in women with different genetic variants. Conversely, some polymorphisms in FSH receptor (FSHR) seem to be associated with modifications in ovarian activity. In particular, studies suggest that the Ser680 genotype for FSHR is a factor of relative resistance to FSH stimulation resulting in slightly higher FSH serum levels, thus leading to a prolonged duration of the menstrual cycle. Moreover, some FSHR gene polymorphisms show a positive association with ovarian response to exogenous gonadotropin administration, hence exhibiting some potential for a pharmacogenetic estimation of the FSH dosage in controlled ovarian stimulation. The study of SNPs of the FSHR gene is an interesting field of research that could provide us with new information about the way each woman responds to exogenous gonadotropin administration during ovulation induction.
Collapse
|
31
|
Arey BJ, López FJ. Are circulating gonadotropin isoforms naturally occurring biased agonists? Basic and therapeutic implications. Rev Endocr Metab Disord 2011; 12:275-88. [PMID: 21706143 DOI: 10.1007/s11154-011-9188-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The gonadotropins, luteinizing hormone, human chorionic gonadotropin and follicle-stimulating hormone, are key regulators of reproduction. As a result of this function, they have been the focus of research for many years. Isolated or recombinant proteins have been successfully used therapeutically for the treatment of infertility; and, in the case of compounds that block gonadotropin activity, for their potential utility in contraception. Until recently, selective small molecules modulating gonadotropin receptor activity have proven difficult to identify. The gonadotropins are glycoproteins that are released into the plasma as differently glycosylated isoforms and bind to specific G protein-coupled receptors. The degree of glycosylation on the gonadotropins has been shown to be important for the biological activities of these hormones and is differentially regulated depending on the steroidal status. Recent data from the study of glycosylated variants of LH, hCG and FSH have revealed that these isoforms have distinct signaling properties that allow for gonadotropin pleiotropic signals to be transduced effectively at the level of the receptor. Thus, glycosylated variants of the gonadotropins behave as biased agonists. Recently, newly developed, small molecule, synthetic allosteric compounds have been identified that are capable of mimicking this biased signaling. This opens the door to development of orally available, drug-like therapies for reproductive disorders that offer similar pleiotropic richness as that offered by the complex, endogenous hormones.
Collapse
Affiliation(s)
- Brian J Arey
- Metabolic and Cardiovascular Drug Discovery, Research and Development, Bristol-Myers Squibb Co., 311 Pennington-Rocky Hill Rd., Mail Stop- 21-1.08, Hopewell, NJ 08543, USA.
| | | |
Collapse
|
32
|
Gloaguen P, Crépieux P, Heitzler D, Poupon A, Reiter E. Mapping the follicle-stimulating hormone-induced signaling networks. Front Endocrinol (Lausanne) 2011; 2:45. [PMID: 22666216 PMCID: PMC3364461 DOI: 10.3389/fendo.2011.00045] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 09/14/2011] [Indexed: 01/14/2023] Open
Abstract
Follicle-stimulating hormone (FSH) is a central regulator of male and female reproductive function. Over the last decade, there has been a growing perception of the complexity associated with FSH-induced cellular signaling. It is now clear that the canonical Gs/cAMP/PKA pathway is not the sole mechanism that must be considered in FSH biological actions. In parallel, consistent with the emerging concept of biased agonism, several examples of ligand-mediated selective signaling pathway activation by gonadotropin receptors have been reported. In this context, it is important to gain an integrative view of the signaling pathways induced by FSH and how they interconnect to form a network. In this review, we propose a first attempt at building topological maps of various pathways known to be involved in the FSH-induced signaling network. We discuss the multiple facets of FSH-induced signaling and how they converge to the hormone integrated biological response. Despite of their incompleteness, these maps of the FSH-induced signaling network represent a first step toward gaining a system-level comprehension of this hormone's actions, which may ultimately facilitate the discovery of novel regulatory processes and therapeutic strategies for infertility and non-steroidal contraception.
Collapse
Affiliation(s)
- Pauline Gloaguen
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des ComportementsNouzilly, France
- UMR6175, CNRSNouzilly, France
- Université François RabelaisTours, France
- L’Institut Français du Cheval et de l’ÉquitationNouzilly, France
| | - Pascale Crépieux
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des ComportementsNouzilly, France
- UMR6175, CNRSNouzilly, France
- Université François RabelaisTours, France
- L’Institut Français du Cheval et de l’ÉquitationNouzilly, France
| | - Domitille Heitzler
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des ComportementsNouzilly, France
- UMR6175, CNRSNouzilly, France
- Université François RabelaisTours, France
- L’Institut Français du Cheval et de l’ÉquitationNouzilly, France
| | - Anne Poupon
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des ComportementsNouzilly, France
- UMR6175, CNRSNouzilly, France
- Université François RabelaisTours, France
- L’Institut Français du Cheval et de l’ÉquitationNouzilly, France
| | - Eric Reiter
- BIOS Group, INRA, UMR85, Unité Physiologie de la Reproduction et des ComportementsNouzilly, France
- UMR6175, CNRSNouzilly, France
- Université François RabelaisTours, France
- L’Institut Français du Cheval et de l’ÉquitationNouzilly, France
- *Correspondence: Eric Reiter, INRA UMR85, CNRS-Université François Rabelais UMR6175, 37380, Nouzilly, France. e-mail:
| |
Collapse
|
33
|
Wehbi V, Decourtye J, Piketty V, Durand G, Reiter E, Maurel MC. Selective modulation of follicle-stimulating hormone signaling pathways with enhancing equine chorionic gonadotropin/antibody immune complexes. Endocrinology 2010; 151:2788-99. [PMID: 20332198 DOI: 10.1210/en.2009-0892] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The injection of equine chorionic gonadotropin (eCG) in dairy goats induces the production of anti-eCG antibodies (Abs) in some females. We have previously shown that Abs negatively modulate the LH and FSH-like bioactivities of eCG, in most cases, compromising fertility in treated females. Surprisingly, we found out that some anti-eCG Abs improved fertility and prolificity of the treated females, in vivo. These Abs, when complexed with eCG, enhanced LH and FSH ability to induce steroidogenesis on specific target cells, in vitro. In the present study, we analyzed the impact of three eCG/anti-eCG Ab-enhancing complexes on two transduction mechanisms triggered by the FSH receptor: guanine nucleotide-binding protein alphaS-subunit/cAMP/protein kinase A (PKA) and beta-arrestin-dependent pathways, respectively. In all cases, significant enhancing effects were observed on ERK phosphorylation compared with eCG alone. However, cAMP production and PKA activation induced by eCG could be differently modulated by Abs. By using a pharmacological inhibitor of PKA and small interfering RNA-mediated knock-down of endogenous beta-arrestin 1 and 2, we demonstrated that signaling bias was induced and was clearly dependent on the complexed Ab. Together, our data show that eCG/anti-eCG Ab-enhancing complexes can differentially modulate cAMP/PKA and beta-arrestin pathways as a function of the complexed Ab. We hypothesize that enhancing Abs may change the eCG conformation, the immune complex acquiring new "biased" pharmacological properties ultimately leading to the physiological effects observed in vivo. The modulation of ligand pharmacological properties by Abs opens promising research avenues towards the optimization of glycoprotein hormone biological activities and, more generally, the development of new therapeutics.
Collapse
Affiliation(s)
- Vanessa Wehbi
- Unité Mixte de Recherche 6175, 37380 Nouzilly, France
| | | | | | | | | | | |
Collapse
|
34
|
Wehbi V, Tranchant T, Durand G, Musnier A, Decourtye J, Piketty V, Butnev VY, Bousfield GR, Crépieux P, Maurel MC, Reiter E. Partially deglycosylated equine LH preferentially activates beta-arrestin-dependent signaling at the follicle-stimulating hormone receptor. Mol Endocrinol 2010; 24:561-73. [PMID: 20107152 DOI: 10.1210/me.2009-0347] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Deglycosylated FSH is known to trigger poor Galphas coupling while efficiently binding its receptor. In the present study, we tested the possibility that a deglycosylated equine LH (eLHdg) might be able to selectively activate beta-arrestin-dependent signaling. We compared native eLH to an eLH derivative [i.e. truncated eLHbeta (Delta121-149) combined with asparagine56-deglycosylated eLHalpha (eLHdg)] previously reported as an antagonist of cAMP accumulation at the FSH receptor (FSH-R). We confirmed that, when used in conjunction with FSH, eLHdg acted as an antagonist for cAMP accumulation in HEK-293 cells stably expressing the FSH-R. Furthermore, when used alone at concentrations up to 1 nM, eLHdg had no detectable agonistic activity on cAMP accumulation, protein kinase A activity or cAMP-responsive element-dependent transcriptional activity. At higher concentrations, however, a weak agonistic action was observed with eLHdg, whereas eLH led to robust responses whatever the concentration. Both eLH and eLHdg triggered receptor internalization and led to beta-arrestin recruitment. Both eLH and eLHdg triggered ERK and ribosomal protein (rp) S6 phosphorylation at 1 nM. The depletion of endogenous beta-arrestins had only a partial effect on eLH-induced ERK and rpS6 phosphorylation. In contrast, ERK and rpS6 phosphorylation was completely abolished at all time points in beta-arrestin-depleted cells. Together, these results show that eLHdg has the ability to preferentially activate beta-arrestin-dependent signaling at the FSH-R. This finding provides a new conceptual and experimental framework to revisit the physiological meaning of gonadotropin structural heterogeneity. Importantly, it also opens a field of possibilities for the development of selective modulators of gonadotropin receptors.
Collapse
Affiliation(s)
- Vanessa Wehbi
- Unité Mixte de Recherche 6175, 37380 Nouzilly, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Arey BJ. Allosteric modulators of glycoprotein hormone receptors: discovery and therapeutic potential. Endocrine 2008; 34:1-10. [PMID: 18956257 DOI: 10.1007/s12020-008-9098-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 06/02/2008] [Accepted: 06/26/2008] [Indexed: 01/27/2023]
Abstract
The glycoprotein hormones, luteinizing hormone, follicle-stimulating hormone and thyroid stimulating hormone, are important regulators of reproductive and metabolic processes. However, because of the nature of their ligand-receptor interactions that contain multiple contact sites, classical small molecule drug discovery strategies have not been successful. However, recent advances in screening and combinatorial chemistry strategies have identified chemical series that act allosterically as positive, negative or mixed modulators of the glycoprotein hormone receptors. This review will discuss the discovery and highlight the currently known series of allosteric modulators to this therapeutically important family of G-protein coupled receptors. Lastly, we will present potential mechanisms whereby the different series could modulate receptor function in the context of currently held theory and known structure of G protein-coupled receptors.
Collapse
Affiliation(s)
- Brian J Arey
- Department of Metabolic and Cardiovascular Drug Discovery, Research and Development, Bristol-Myers Squibb Co, 311 Pennington Rocky-Hill Rd, Mail Stop 21-1.08, Pennington, NJ 08534, USA.
| |
Collapse
|
36
|
Differing pharmacological activities of thiazolidinone analogs at the FSH receptor. Biochem Biophys Res Commun 2008; 368:723-8. [DOI: 10.1016/j.bbrc.2008.01.119] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Accepted: 01/26/2008] [Indexed: 11/24/2022]
|
37
|
Ulloa-Aguirre A, Zariñán T, Pasapera AM, Casas-González P, Dias JA. Multiple facets of follicle-stimulating hormone receptor function. Endocrine 2007; 32:251-63. [PMID: 18246451 DOI: 10.1007/s12020-008-9041-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 01/04/2008] [Accepted: 01/14/2008] [Indexed: 10/22/2022]
Abstract
Follicle-stimulating hormone (FSH) is a glycoprotein hormone produced by the anterior pituitary gland. This gonadotropin plays an essential role in reproduction. Its receptor (FSHR) belongs to the superfamily of G protein-coupled receptors (GPCR), specifically the family of rhodopsin-like receptors. Agonist binding to the FSHR triggers the rapid activation of multiple signaling cascades, mainly the cAMP-adenylyl cyclase-protein kinase A cascade, that impact diverse biological effects of FSH in the gonads. As in other G protein-coupled receptors, the several cytoplasmic domains of the FSHR are involved in signal transduction and termination of the FSH signal. Here we summarize some recent information on the signaling cascades activated by FSH as well as on the role of the intracytoplasmic domains of the FSHR in coupling to membrane and cytosolic proteins linked to key biological functions regulated by the FSH-FSHR system.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Research Unit in Reproductive Medicine, Hospital de Ginecobstetricia "Luis Castelazo Ayala", Instituto Mexicano del Seguro Social, Apartado Postal 99-065, Unidad Independencia, C.P. 10101 Mexico, D.F., Mexico.
| | | | | | | | | |
Collapse
|
38
|
Mönkkönen KS, Aflatoonian R, Lee KF, Yeung WSB, Tsao SW, Laitinen JT, Tuckerman EM, Li TC, Fazeli A. Localization and variable expression of G alpha(i2) in human endometrium and Fallopian tubes. Hum Reprod 2007; 22:1224-30. [PMID: 17347170 DOI: 10.1093/humrep/dem022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Heterotrimeric G proteins take part in membrane-mediated cell signalling and have a role in hormonal regulation. This study clarifies the expression and localization of the G protein subunit G alpha(i2) in the human endometrium and Fallopian tube and changes in G alpha(i2) expression in human endometrium during the menstrual cycle. METHODS The expression of G alpha(i2) was identified by Polymerase chain reaction (PCR), and localization confirmed by immunostaining. Cyclic changes in G alpha(i2) expression during the menstrual cycle were evaluated by quantitative real-time PCR. RESULTS We found G alpha(i2) to be expressed in human endometrium, Fallopian tube tissue and in primary cultures of Fallopian tube epithelial cells. Our studies revealed enriched localization of G alpha(i2) in Fallopian tube cilia and in endometrial glands. We showed that G alpha(i2) expression in human endometrium changes significantly during the menstrual cycle, with a higher level in the secretory versus proliferative and menstrual phases (P < 0.05). CONCLUSIONS G alpha(i2) is specifically localized in human Fallopian tube epithelial cells, particularly in the cilia, and is likely to have a cilia-specific role in reproduction. Significantly variable expression of G alpha(i2) during the menstrual cycle suggests G alpha(i2) might be under hormonal regulation in the female reproductive tract in vivo.
Collapse
Affiliation(s)
- Kati S Mönkkönen
- Department of Pharmacology and Toxicology, University of Kuopio, Kuopio, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ulloa-Aguirre A, Uribe A, Zariñán T, Bustos-Jaimes I, Pérez-Solis MA, Dias JA. Role of the intracellular domains of the human FSH receptor in G(alphaS) protein coupling and receptor expression. Mol Cell Endocrinol 2007; 260-262:153-62. [PMID: 17045734 PMCID: PMC1782136 DOI: 10.1016/j.mce.2005.11.050] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2005] [Accepted: 11/15/2005] [Indexed: 11/18/2022]
Abstract
The human (h) follicle-stimulating hormone receptor (FSHR) belongs to the superfamily of G protein-coupled receptors (GPCRs). This receptor consists of 695 amino acid residues and is preferentially coupled to the G(s) protein. This receptor is highly conserved among species (overall homology, 85%), with a 25-69% homology drop when compared to the human LH and TSH receptors. Although studies in prototypical rhodopsin/beta-adrenergic receptors suggest that multiple domains in the intracellular loops (iL) and the carboxyl-terminus (Ctail) of these receptors contribute to G protein coupling and receptor expression, there is a paucity of structure/function data on the role of these domains in FSHR function. Employing point mutations we have found that several residues present in the iL2 of the hFSHR are important for both coupling the receptor to the G(s) protein and maintaining the receptor molecule in an inactive conformation. In fact, HEK-293 cells expressing several hFSHR mutants with substitutions at R(450) (central to the highly conserved ERW triplet motif) and T(453) (a potential target for phosphorylation) failed to mediate ligand-provoked G(s) protein activation but not agonist binding, whereas substitutions at the hydrophobic L(460) (a conserved residue present in all glycoprotein hormone receptors) conferred elevated basal cAMP to the transfected cells. Thus, this particular loop apparently acts as a conformational switch for allowing the receptor to adopt an active conformation upon agonist stimulation. Residues in both ends of the iL3 are important for signal transduction in a number of GPCRs, including the FSHR. We have recently explored the importance of the reversed BBXXB motif (BXXBB; where B represents a basic residue and X a non-basic residue) present in the juxtamembrane region of the hFSHR iL3. A hFSHR mutant with all basic amino acids present in the iL3 BXXBB motif replaced by alanine failed to bind agonist and activate effector, and was expressed as an immature < or =62kDa form of the receptor. Individual substitutions of basic residues resulted in mutants that bound agonist normally but failed to activate effector when replaced at R(552) or R(556). Triple mutations in the same motif located in the NH(2)-end of the Ctail resulted in a complete inability of the receptor to bind agonist and activate effector, whereas individual substitutions resulted in decreased or virtually abolished agonist binding and cAMP accumulation, with both functions correlating with the detected levels of mature (80kDa) forms of the receptor. Thus, the BXXBB motif at the iL3 of the FSHR is essential for coupling the activated receptor to the G(s) protein, whereas the same motif in the Ctail is apparently more important for membrane expression. The role of cysteine residues present in the Ctail of the FSHR is an enigma since there are no conserved cysteines amongst LHR, FSHR and TSHR. C(629) and C(655) are conserved in the gonadotropin receptors but not in the TSHR. Alanine replacement of C(627) had no effect on hFSHR expression and function, whereas the same mutation at C(629) altered membrane expression and signal transduction. Serine or threonine substitutions of C(655) did not modify any of the parameters analyzed. In the hFSHR, C(629) may be a target for palmitoylation, and apparently it is the only cysteine residue in the Ctail domain that might play an important role in receptor function.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Research Unit in Reproductive Medicine, Hospital de Ginecobstetricia Luis Castelazo Ayala, Instituto Mexicano del Seguro Social, Apartado Postal 99-065, Unidad Independencia, México 10101 D.F., Mexico.
| | | | | | | | | | | |
Collapse
|
40
|
Sairam MR, Babu PS. The tale of follitropin receptor diversity: a recipe for fine tuning gonadal responses? Mol Cell Endocrinol 2007; 260-262:163-71. [PMID: 17081682 DOI: 10.1016/j.mce.2005.11.052] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Accepted: 11/21/2005] [Indexed: 10/24/2022]
Abstract
The original concept (dogma) of a single FSH receptor entity coupling to G(s) protein to activate adenylate cyclase and producing cAMP as second messenger appears inadequate to explain pleiotropic actions of the hormone. The identification and expression of alternatively spliced gonadotropin receptors, suggest that alternative splicing could serve as a mechanism for creating receptor diversity. Studies focused on sheep and mouse gonadal tissues show that the single large gene of approximately 250kb is a modular structure whose pre-mRNA undergoes alternative splicing creating several subtypes (at least four FSH-R1 to R4 identified to date). With segments of the N-terminus that are identical different topographies are generated by differing carboxyl termini. The same gene thus produces receptor types with different motifs that can display dominant positive, dominant negative, growth factor/cytokine type and potentially soluble binding protein features. Functional relevance is shown by modulation of receptor variants during hormonal stimulation. Presence of equivalent segments of the gene in the human and bovine suggests conservation and predicts similarity in structures and function. Thus, the complex cellular biology of follitropin receptors that may interact differently with polymorphic forms (glycosylation variants) of FSH represents an intricate scheme to regulate hormone signaling.
Collapse
Affiliation(s)
- M Ram Sairam
- Molecular Reproduction Research Laboratory, Clinical Research Institute of Montreal, 110 Pine Avenue West, Montreal, Quebec H2W 1R7, Canada.
| | | |
Collapse
|
41
|
Campo S, Ambao V, Creus S, Gottlieb S, Fernandez Vera G, Benencia H, Bergadá C. Carbohydrate complexity and proportions of serum FSH isoforms in the male: lectin-based studies. Mol Cell Endocrinol 2007; 260-262:197-204. [PMID: 17084021 DOI: 10.1016/j.mce.2006.01.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Accepted: 01/03/2006] [Indexed: 11/25/2022]
Abstract
FSH is synthesized and secreted in multiple molecular forms with different oligosaccharide structures which are needed for full expression of biological activity. GnRH and sex steroids modulate oligosaccharide structure and composition. In the present study we have assessed the carbohydrate complexity and proportion of circulating FSH isoforms during puberty, aging and after androgen administration to pubertal anorchid boys. Preparative isoelectrofocusing and lectin chromatography were used to isolate FSH isoforms on the basis of charge and internal carbohydrate complexity. Differences in sialic acid content and a progressive increase of isoforms bearing highly branched oligosaccharides were found during puberty. Less acidic, more bioactive FSH isoforms, secreted at mid-puberty may modulate important maturational events in the Sertoli cell population. Androgen administration to pubertal anorchid boys favoured the secretion of this type of isoforms. In adult men, the predominance of FSH isoforms bearing complex type oligosaccharides remained unchanged until very advanced age. These results show that the predominance of FSH isoforms bearing fully processed oligosaccharides in circulation may contribute to the development and maintenance of seminiferous epithelium function in men.
Collapse
Affiliation(s)
- S Campo
- Centro de Investigaciones Endocrinológicas, Hospital General de Niños "R. Gutiérrez", Gallo 1330, C1425EFD Buenos Aires, Argentina.
| | | | | | | | | | | | | |
Collapse
|
42
|
Yanofsky SD, Shen ES, Holden F, Whitehorn E, Aguilar B, Tate E, Holmes CP, Scheuerman R, MacLean D, Wu MM, Frail DE, López FJ, Winneker R, Arey BJ, Barrett RW. Allosteric activation of the follicle-stimulating hormone (FSH) receptor by selective, nonpeptide agonists. J Biol Chem 2006; 281:13226-13233. [PMID: 16540466 DOI: 10.1074/jbc.m600601200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The pituitary glycoprotein hormones, luteinizing hormone and follicle-stimulating hormone (FSH), act through their cognate receptors to initiate a series of coordinated physiological events that results in germ cell maturation. Given the importance of FSH in regulating folliculogenesis and fertility, the development of FSH mimetics has been sought to treat infertility. Currently, purified and recombinant human FSH are the only FSH receptor (FSH-R) agonists available for infertility treatment. By screening unbiased combinatorial chemistry libraries, using a cAMP-responsive luciferase reporter assay, we discovered thiazolidinone agonists (EC50's = 20 microm) of the human FSH-R. Subsequent analog library screening and parallel synthesis optimization resulted in the identification of a potent agonist (EC50 = 2 nm) with full efficacy compared with FSH that was FSH-R-selective and -dependent. The compound mediated progesterone production in Y1 cells transfected with the human FSH-R (EC50 = 980 nm) and estradiol production from primary rat ovarian granulosa cells (EC50 = 10.5 nm). This and related compounds did not compete with FSH for binding to the FSH-R. Use of human FSH/thyroid-stimulating hormone (TSH) receptor chimeras suggested a novel mechanism for receptor activation through a binding site independent of the natural hormone binding site. This study is the first report of a high affinity small molecule agonist that activates a glycoprotein hormone receptor through an allosteric mechanism. The small molecule FSH receptor agonists described here could lead to an oral alternative to the current parenteral FSH treatments used clinically to induce ovarian stimulation for both in vivo and in vitro fertilization therapy.
Collapse
Affiliation(s)
| | - Emily S Shen
- Women's Health and Musculoskeletal Biology, Wyeth Pharmaceuticals, Collegeville, Pennsylvania, 19426-3930
| | | | | | | | - Emily Tate
- Affymax, Inc., Palo Alto, California 94304
| | | | | | | | - May M Wu
- Women's Health and Musculoskeletal Biology, Wyeth Pharmaceuticals, Collegeville, Pennsylvania, 19426-3930
| | - Donald E Frail
- Women's Health and Musculoskeletal Biology, Wyeth Pharmaceuticals, Collegeville, Pennsylvania, 19426-3930
| | - Francisco J López
- Women's Health and Musculoskeletal Biology, Wyeth Pharmaceuticals, Collegeville, Pennsylvania, 19426-3930
| | - Richard Winneker
- Women's Health and Musculoskeletal Biology, Wyeth Pharmaceuticals, Collegeville, Pennsylvania, 19426-3930
| | - Brian J Arey
- Women's Health and Musculoskeletal Biology, Wyeth Pharmaceuticals, Collegeville, Pennsylvania, 19426-3930
| | | |
Collapse
|
43
|
Maudsley S, Martin B, Luttrell LM. The origins of diversity and specificity in g protein-coupled receptor signaling. J Pharmacol Exp Ther 2005; 314:485-94. [PMID: 15805429 PMCID: PMC2656918 DOI: 10.1124/jpet.105.083121] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The modulation of transmembrane signaling by G protein-coupled receptors (GPCRs) constitutes the single most important therapeutic target in medicine. Drugs acting on GPCRs have traditionally been classified as agonists, partial agonists, or antagonists based on a two-state model of receptor function embodied in the ternary complex model. Over the past decade, however, many lines of investigation have shown that GPCR signaling exhibits greater diversity and "texture" than previously appreciated. Signal diversity arises from numerous factors, among which are the ability of receptors to adopt multiple "active" states with different effector-coupling profiles; the formation of receptor dimers that exhibit unique pharmacology, signaling, and trafficking; the dissociation of receptor "activation" from desensitization and internalization; and the discovery that non-G protein effectors mediate some aspects of GPCR signaling. At the same time, clustering of GPCRs with their downstream effectors in membrane microdomains and interactions between receptors and a plethora of multidomain scaffolding proteins and accessory/chaperone molecules confer signal preorganization, efficiency, and specificity. In this context, the concept of agonist-selective trafficking of receptor signaling, which recognizes that a bound ligand may select between a menu of active receptor conformations and induce only a subset of the possible response profile, presents the opportunity to develop drugs that change the quality as well as the quantity of efficacy. As a more comprehensive understanding of the complexity of GPCR signaling is developed, the rational design of ligands possessing increased specific efficacy and attenuated side effects may become the standard mode of drug development.
Collapse
Affiliation(s)
- Stuart Maudsley
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Gerontology Research Center, 5600 Nathan Shock Drive, Johns Hopkins Medical Center, Baltimore, MD, USA.
| | | | | |
Collapse
|
44
|
Arey BJ, Seethala R, Ma Z, Fura A, Morin J, Swartz J, Vyas V, Yang W, Dickson JK, Feyen JHM. A novel calcium-sensing receptor antagonist transiently stimulates parathyroid hormone secretion in vivo. Endocrinology 2005; 146:2015-22. [PMID: 15637285 DOI: 10.1210/en.2004-1318] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Circulating calcium (Ca(2+)) is a primary regulator of bone homeostasis through its action on PTH secretion. Extracellular Ca(2+) modulates PTH secretion through a cell surface G protein-coupled receptor, the calcium-sensing receptor (CaR). The expression of the CaR suggests a critical role in cellular regulation by calcium in various organs, including parathyroid gland, bone, and kidney. Despite an obvious pharmacological utility for CaR antagonists in the treatment of disease, only a limited number of such classes of compounds exist. We have identified a novel class of small molecules with specific activity at the CaR. This class of compounds is represented by compound 1. It possesses potent antagonist activity at the human CaR with IC(50) values of 64 nm and 230 nm in inhibiting intracellular Ca(2+) flux and inositol phosphate generation in vitro, respectively. When administered to male rats in vivo, compound 1 robustly increased serum PTH levels. The stimulation of PTH secretion was rapid and transient when administered either iv or orally. The pharmacokinetic profile of compound 1 after oral administration revealed that maximal plasma levels of compound were reached within 1 h and the half-life of the compound to be approximately 2 h in rats. These data describe a representative compound of a novel chemical class than previously described allosteric modulators that offer a new avenue for the development of improved treatments of osteoporosis.
Collapse
Affiliation(s)
- Brian J Arey
- Metabolic and Cardiovascular Drug Discovery, Bristol-Myers Squibb Co., 311 Pennington-Rocky Hill Road, Pennington, New Jersey 08534, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Stevis PE, Arey BJ, Deecher DC. A glutathione-S-transferase-FSHalpha subunit hybrid associates with FSHbeta, retains biological activity, and facilitates purification. Biochem Biophys Res Commun 2004; 319:1026-31. [PMID: 15184084 DOI: 10.1016/j.bbrc.2004.05.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2004] [Indexed: 11/17/2022]
Abstract
The glycoprotein hormones are heterodimeric proteins that share a common alpha subunit and have unique beta subunits that confer receptor selectivity. One member of this family, follicle-stimulating hormone (FSH), is secreted by the pituitary and is involved in the control of male and female reproduction. Herein, we describe the construction of baculoviruses for glutathione-S-transferase (GST) fusions of the human FSH (hFSH) subunits and their expression in insect cells, either alone or with the complementary non-fused FSH subunits (FSHalpha or FSHbeta). Only the GST-BV-hFSHalpha monomer and the GST-BV-hFSHalpha/BV-hFSHbeta (GST-BV-hFSH) heterodimer were efficiently secreted into the culture supernatant. The hybrid molecule, GST-BV-hFSH, was affinity purified in one step, and demonstrated activity in receptor-radioligand binding assays and in a cAMP accumulation assay. The use of GST-BV-hFSHalpha provides a novel and efficient method for purifying and studying members of the glycoprotein hormone family derived from the culture supernatant or subcellular fractions of the cell.
Collapse
Affiliation(s)
- Panayiotis E Stevis
- Women's Health and Bone, Wyeth Research, 500 Arcola Road, Collegeville, PA 19426, USA.
| | | | | |
Collapse
|
46
|
Mateos J, Mañanós E, Martínez-Rodríguez G, Carrillo M, Querat B, Zanuy S. Molecular characterization of sea bass gonadotropin subunits (alpha, FSHbeta, and LHbeta) and their expression during the reproductive cycle. Gen Comp Endocrinol 2003; 133:216-32. [PMID: 12928011 DOI: 10.1016/s0016-6480(03)00164-3] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Reproduction is controlled by two pituitary gonadotropin hormones, follicle-stimulating hormone (FSH) and luteinizing hormone (LH). This study reports the cloning, sequence analysis, and gene expression of gonadotropin (GTH) subunits from the European sea bass (Dicentrarchus labrax). The GTH subunits were cloned from a sea bass brain-pituitary cDNA library. The nucleotide sequences of the common alpha, the FSHbeta, and the LHbeta subunit cDNAs were 625, 521, and 591 base pair (bp) long, respectively, encoding for mature peptides of 94, 105, and 115 amino acids (aa), respectively. Sequence analysis showed that sea bass FSHbeta is more similar to higher vertebrate FSHbeta's (35-37%) than to LHbeta's (26-30%), whereas sea bass LHbeta is more similar to LHbeta's (40-53%) than to FSHbeta's (26-41%). Phylogenetic analysis of fish GTH sequences grouped the beta subunits into two groups, FSH and LH, distributed into four classes, corresponding to the accepted divisions of Elopomorphs, Ostariophysis, Salmonids, and Percomorphs. A dot-blot technique was developed to analyze GTH pituitary mRNA levels during the reproductive cycle of male sea bass. From October (initiation of gametogenesis) to February (spermiation), the expression of all three subunits in the pituitary increased in parallel, concomitantly with the gonadosomatic index (GSI) and the accumulation of LH protein in the pituitary, all values declining sharply at post-spermiation. This study demonstrates that the pituitary of sea bass contains two gonadotropin hormones and that both gonadotropins are probably involved in the control of gametogenesis, gamete maturation, and spermiation.
Collapse
Affiliation(s)
- Jorge Mateos
- Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal (Castellón), Spain
| | | | | | | | | | | |
Collapse
|
47
|
Ulloa-Aguirre A, Timossi C, Barrios-de-Tomasi J, Maldonado A, Nayudu P. Impact of carbohydrate heterogeneity in function of follicle-stimulating hormone: studies derived from in vitro and in vivo models. Biol Reprod 2003; 69:379-89. [PMID: 12700183 DOI: 10.1095/biolreprod.103.016915] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Carbohydrates attached to the protein core of glycoprotein hormones influence a number of intracellular and extracellular processes. As with other members of the glycoprotein hormone family, FSH is produced and released as an array of isoforms that differ from each other in the structure of their oligosaccharide attachments. In this review, we discuss how carbohydrate heterogeneity can impact on FSH action in different in vitro and in vivo systems. We present evidence for diverse effects of distinct charge isoforms at the target cell level, including differential and unique effects on various end responses, and discuss how the use of multiple cell-type assays has allowed identification of some specific effects of FSH isoforms on different cell populations and follicle compartments as well as oocyte maturation. Finally, we discuss recent information on the ability of naturally occurring and laboratory manufactured FSH isoforms to evoke particular effects on granulosa cell function and ovarian follicular maturation in vivo. Such studies have provided evidence that the type(s) of FSH signal delivered may in fact regulate distinct biological outcomes irrespective or in addition to outcomes dictated solely by clearance rate differences.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Research Unit in Reproductive Medicine, Hospital de Ginecobstetricia Luis Castelazo Ayala, IMSS, México DF.
| | | | | | | | | |
Collapse
|
48
|
Kenakin T. Predicting therapeutic value in the lead optimization phase of drug discovery. Nat Rev Drug Discov 2003; 2:429-38. [PMID: 12776218 DOI: 10.1038/nrd1110] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Recombinant and natural cellular assays for human G-protein-coupled receptors are used to optimize initial lead molecules obtained from screening. Although the activity of these molecules can be assessed on human genotype receptors, there is increasing evidence that cells impose a phenotypic selectivity to molecules in various cellular backgrounds. This opens the possibility of dissimulations between activity seen in lead optimization assays and the intended therapeutic value in humans. This review discusses the mechanisms by which cells can impose phenotypic selectivity on molecules and approaches to reduce this practical problem for drug discovery.
Collapse
Affiliation(s)
- Terry Kenakin
- Systems Research, GlaxoSmithKline Research and Development, 5 Moore Drive, Research Triangle Park, North Carolina 27709, USA.
| |
Collapse
|
49
|
Lu J, Swinnen E, Proost P, De Vreese B, Vankelecom H, Denef C. Isolation and structure-bioactivity characterization of glycosylated N-pro-opiomelanocortin isoforms. J Neuroendocrinol 2002; 14:869-79. [PMID: 12421340 DOI: 10.1046/j.1365-2826.2002.00851.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The N-terminal fragment of mouse pro-opiomelanocortin (N-POMC) was isolated from AtT-20 cell-conditioned medium on the basis of immunoreactivity to an anti-POMC1-50 monoclonal antibody by a concentration step, a cation exchange step, reversed phase high-performance liquid chromatography (HPLC) and size exclusion HPLC. Two groups of N-POMC isoforms with a molecular weight (MW) of approximately 11 kDa and 13 kDa, respectively, were identified by mass spectrometry and N-terminal amino acid sequencing. C-terminal sequencing indicated that 11 kDa isoforms correspond to POMC1-74 and 13 kDa isoforms to POMC1-95. Isoforms from both groups enhanced the prolactin mRNA content (measured by means of TaqMan real-time reverse transcription-polymerase chain reaction) in cultured rat pituitary cell aggregates in a dose-dependent manner, but not all of them showed this activity. POMC1-74 compounds were significantly more potent than POMC1-95 isoforms. The observed effects were abolished by coincubation with the monoclonal anti-POMC1-50 antibody, showing the specificity of this biological action. Incorporation of bromodeoxyuridine into DNA of immunostained lactotrophs was enhanced by only a minor part of the isoforms. Some of these had no effect on prolactin mRNA expression. The N-POMC isoforms appeared to be N- and at least in part O-glycosylated. After enzymatic N-deglycosylation of selected N-POMC isoforms, the stimulatory effect on the prolactin mRNA level was depressed (in case of the POMC1-95 isoforms) or totally abolished (in case of the POMC1-74 isoforms). The present findings show that N-POMC is a mixture of differentially glycosylated isoforms, that the isoforms of POMC1-74 are the biologically more effective forms and that different isoforms induce different biological responses in the same cell population. The data also show the essential role of N-glycosylation in the biological response.
Collapse
Affiliation(s)
- J Lu
- Laboratory of Cell Pharmacology, University of Leuven (KU Leuven), Medical School, Gasthuisberg O&N, Belgium
| | | | | | | | | | | |
Collapse
|
50
|
Arey BJ, Deecher DC, Shen ES, Stevis PE, Meade EH, Wrobel J, Frail DE, López FJ. Identification and characterization of a selective, nonpeptide follicle-stimulating hormone receptor antagonist. Endocrinology 2002; 143:3822-9. [PMID: 12239093 DOI: 10.1210/en.2002-220372] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The glycoprotein hormones (LH, FSH, and TSH) are critical to the maintenance of physiological homeostasis and control of reproduction. However, despite an obvious utility for synthetic pharmacological agents, there are few reports of selective, nonpeptide agonists or antagonists to receptors for these hormones. We have identified and characterized a novel synthetic molecule capable of inhibiting the action of FSH. This compound, 7-[4-[Bis-(2-carbamoyl-ethyl)-amino]-6-chloro-(1,3,5)-triazin-2-ylamino)-4-hydroxy-3-(4-methoxy-phenylazo)-naphthalene]-2-sulfonic acid, sodium salt (compound 1), is a selective, noncompetitive inhibitor of the human (h) and rat (r) FSH receptors (FSHRs). Compound 1 selectively inhibited binding of [(125)I]hFSH with an IC(50) value of 5.4 +/- 2.3 micro M. Radioligand-binding assays were performed using the baculovirus expressed extracellular domain of hFSHR (BV-tFSHR) to demonstrate site-specific interaction. Compound 1 competed for [(125)I]hFSH binding to BV-tFSHR with an IC(50) value of 10 +/- 2.8 micro M. Functionally, compound 1 inhibited hFSH-induced cAMP accumulation and steroidogenesis in vitro with an IC(50) value of 3 +/- 0.6 micro M. Competition of compound 1 for binding to other glycoprotein hormone receptors and other G protein-coupled receptors demonstrated select activity for FHSRs. Compound 1 inhibited ovulation in immature and cycling adult rats. These data provide proof of concept that selective, small molecule antagonists can be designed for glycoprotein hormone receptors.
Collapse
Affiliation(s)
- Brian J Arey
- Women's Health Research Institute and Medicinal Chemistry, Department of Chemical Sciences, Wyeth Research, Collegeville, Pennsylvania 19680, USA
| | | | | | | | | | | | | | | |
Collapse
|