1
|
Radomsky T, Anderson RC, Millar RP, Newton CL. Restoring function to inactivating G protein-coupled receptor variants in the hypothalamic-pituitary-gonadal axis 1. J Neuroendocrinol 2024; 36:e13418. [PMID: 38852954 DOI: 10.1111/jne.13418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 03/30/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024]
Abstract
G protein-coupled receptors (GPCRs) are central to the functioning of the hypothalamic-pituitary-gonadal axis (HPG axis) and include the rhodopsin-like GPCR family members, neurokinin 3 receptor, kappa-opioid receptor, kisspeptin 1 receptor, gonadotropin-releasing hormone receptor, and the gonadotropin receptors, luteinizing hormone/choriogonadotropin receptor and follicle-stimulating hormone receptor. Unsurprisingly, inactivating variants of these receptors have been implicated in a spectrum of reproductive phenotypes, including failure to undergo puberty, and infertility. Clinical induction of puberty in patients harbouring such variants is possible, but restoration of fertility is not always a realisable outcome, particularly for those patients suffering from primary hypogonadism. Thus, novel pharmaceuticals and/or a fundamental change in approach to treating these patients are required. The increasing wealth of data describing the effects of coding-region genetic variants on GPCR function has highlighted that the majority appear to be dysfunctional as a result of misfolding of the encoded receptor protein, which, in turn, results in impaired receptor trafficking through the secretory pathway to the cell surface. As such, these intracellularly retained receptors may be amenable to 'rescue' using a pharmacological chaperone (PC)-based approach. PCs are small, cell permeant molecules hypothesised to interact with misfolded intracellularly retained proteins, stabilising their folding and promoting their trafficking through the secretory pathway. In support of the use of this approach as a viable therapeutic option, it has been observed that many rescued variant GPCRs retain at least a degree of functionality when 'rescued' to the cell surface. In this review, we examine the GPCR PC research landscape, focussing on the rescue of inactivating variant GPCRs with important roles in the HPG axis, and describe what is known regarding the mechanisms by which PCs restore trafficking and function. We also discuss some of the merits and obstacles associated with taking this approach forward into a clinical setting.
Collapse
Affiliation(s)
- Tarryn Radomsky
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ross C Anderson
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Robert P Millar
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Claire L Newton
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
2
|
Shpakov AO. Hormonal and Allosteric Regulation of the Luteinizing Hormone/Chorionic Gonadotropin Receptor. FRONT BIOSCI-LANDMRK 2024; 29:313. [PMID: 39344322 DOI: 10.31083/j.fbl2909313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 10/01/2024]
Abstract
Luteinizing hormone (LH) and human chorionic gonadotropin (CG), like follicle-stimulating hormone, are the most important regulators of the reproductive system. They exert their effect on the cell through the LH/CG receptor (LHCGR), which belongs to the family of G protein-coupled receptors. Binding to gonadotropin induces the interaction of LHCGR with various types of heterotrimeric G proteins (Gs, Gq/11, Gi) and β-arrestins, which leads to stimulation (Gs) or inhibition (Gi) of cyclic adenosine monophosphate-dependent cascades, activation of the phospholipase pathway (Gq/11), and also to the formation of signalosomes that mediate the stimulation of mitogen-activated protein kinases (β-arrestins). The efficiency and selectivity of activation of intracellular cascades by different gonadotropins varies, which is due to differences in their interaction with the ligand-binding site of LHCGR. Gonadotropin signaling largely depends on the status of N- and O-glycosylation of LH and CG, on the formation of homo- and heterodimeric receptor complexes, on the cell-specific microenvironment of LHCGR and the presence of autoantibodies to it, and allosteric mechanisms are important in the implementation of these influences, which is due to the multiplicity of allosteric sites in different loci of the LHCGR. The development of low-molecular-weight allosteric regulators of LHCGR with different profiles of pharmacological activity, which can be used in medicine for the correction of reproductive disorders and in assisted reproductive technologies, is promising. These and other issues regarding the hormonal and allosteric regulation of LHCGR are summarized and discussed in this review.
Collapse
Affiliation(s)
- Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
3
|
Duan J, He XH, Li SJ, Xu HE. Cryo-electron microscopy for GPCR research and drug discovery in endocrinology and metabolism. Nat Rev Endocrinol 2024; 20:349-365. [PMID: 38424377 DOI: 10.1038/s41574-024-00957-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/29/2024] [Indexed: 03/02/2024]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors, with many GPCRs having crucial roles in endocrinology and metabolism. Cryogenic electron microscopy (cryo-EM) has revolutionized the field of structural biology, particularly regarding GPCRs, over the past decade. Since the first pair of GPCR structures resolved by cryo-EM were published in 2017, the number of GPCR structures resolved by cryo-EM has surpassed the number resolved by X-ray crystallography by 30%, reaching >650, and the number has doubled every ~0.63 years for the past 6 years. At this pace, it is predicted that the structure of 90% of all human GPCRs will be completed within the next 5-7 years. This Review highlights the general structural features and principles that guide GPCR ligand recognition, receptor activation, G protein coupling, arrestin recruitment and regulation by GPCR kinases. The Review also highlights the diversity of GPCR allosteric binding sites and how allosteric ligands could dictate biased signalling that is selective for a G protein pathway or an arrestin pathway. Finally, the authors use the examples of glycoprotein hormone receptors and glucagon-like peptide 1 receptor to illustrate the effect of cryo-EM on understanding GPCR biology in endocrinology and metabolism, as well as on GPCR-related endocrine diseases and drug discovery.
Collapse
Affiliation(s)
- Jia Duan
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, China.
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Xin-Heng He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shu-Jie Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Department of Traditional Chinese Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - H Eric Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
4
|
Makkonen K, Jännäri M, Crisóstomo L, Kuusi M, Patyra K, Melnyk V, Linnossuo V, Ojala J, Ravi R, Löf C, Mäkelä JA, Miettinen P, Laakso S, Ojaniemi M, Jääskeläinen J, Laakso M, Bossowski F, Sawicka B, Stożek K, Bossowski A, Kleinau G, Scheerer P, FinnGen F, Reeve MP, Kero J. Mechanisms of thyrotropin receptor-mediated phenotype variability deciphered by gene mutations and M453T-knockin model. JCI Insight 2024; 9:e167092. [PMID: 38194289 PMCID: PMC11143923 DOI: 10.1172/jci.insight.167092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
The clinical spectrum of thyrotropin receptor-mediated (TSHR-mediated) diseases varies from loss-of-function mutations causing congenital hypothyroidism to constitutively active mutations (CAMs) leading to nonautoimmune hyperthyroidism (NAH). Variation at the TSHR locus has also been associated with altered lipid and bone metabolism and autoimmune thyroid diseases. However, the extrathyroidal roles of TSHR and the mechanisms underlying phenotypic variability among TSHR-mediated diseases remain unclear. Here we identified and characterized TSHR variants and factors involved in phenotypic variability in different patient cohorts, the FinnGen database, and a mouse model. TSHR CAMs were found in all 16 patients with NAH, with 1 CAM in an unexpected location in the extracellular leucine-rich repeat domain (p.S237N) and another in the transmembrane domain (p.I640V) in 2 families with distinct hyperthyroid phenotypes. In addition, screening of the FinnGen database revealed rare functional variants as well as distinct common noncoding TSHR SNPs significantly associated with thyroid phenotypes, but there was no other significant association between TSHR variants and more than 2,000 nonthyroid disease endpoints. Finally, our TSHR M453T-knockin model revealed that the phenotype was dependent on the mutation's signaling properties and was ameliorated by increased iodine intake. In summary, our data show that TSHR-mediated disease risk can be modified by variants at the TSHR locus both inside and outside the coding region as well as by altered TSHR-signaling and dietary iodine, supporting the need for personalized treatment strategies.
Collapse
Affiliation(s)
- Kristiina Makkonen
- Department of Clinical Sciences, Faculty of Medicine, and
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Meeri Jännäri
- Department of Clinical Sciences, Faculty of Medicine, and
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Luís Crisóstomo
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Matilda Kuusi
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Konrad Patyra
- Department of Clinical Sciences, Faculty of Medicine, and
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Veli Linnossuo
- Department of Clinical Sciences, Faculty of Medicine, and
| | - Johanna Ojala
- Department of Clinical Sciences, Faculty of Medicine, and
| | - Rowmika Ravi
- Department of Clinical Sciences, Faculty of Medicine, and
| | - Christoffer Löf
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Juho-Antti Mäkelä
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Päivi Miettinen
- New Children’s Hospital, Helsinki University Hospital, Helsinki, Finland
| | - Saila Laakso
- New Children’s Hospital, Helsinki University Hospital, Helsinki, Finland
| | - Marja Ojaniemi
- Department of Pediatrics and Adolescence, PEDEGO Research Unit and Medical Research Center, University and University Hospital of Oulu, Oulu, Finland
| | | | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Filip Bossowski
- Department of Pediatrics, Endocrinology, Diabetes with a Cardiology Unit, Medical University in Białystok, Bialystok, Poland
| | - Beata Sawicka
- Department of Pediatrics, Endocrinology, Diabetes with a Cardiology Unit, Medical University in Białystok, Bialystok, Poland
| | - Karolina Stożek
- Department of Pediatrics, Endocrinology, Diabetes with a Cardiology Unit, Medical University in Białystok, Bialystok, Poland
| | - Artur Bossowski
- Department of Pediatrics, Endocrinology, Diabetes with a Cardiology Unit, Medical University in Białystok, Bialystok, Poland
| | - Gunnar Kleinau
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and
- Humboldt - Universität zu Berlin, Institute of Medical Physics, Biophysics, Group Structural Biology of Cellular Signaling, Berlin, Germany
| | - Patrick Scheerer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and
- Humboldt - Universität zu Berlin, Institute of Medical Physics, Biophysics, Group Structural Biology of Cellular Signaling, Berlin, Germany
| | - FinnGen FinnGen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- FinnGen is detailed in Supplemental Acknowledgments
| | - Mary Pat Reeve
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jukka Kero
- Department of Clinical Sciences, Faculty of Medicine, and
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| |
Collapse
|
5
|
Kleinau G, Ali AH, Wiechert F, Szczepek M, Schmidt A, Spahn CMT, Liebscher I, Schöneberg T, Scheerer P. Intramolecular activity regulation of adhesion GPCRs in light of recent structural and evolutionary information. Pharmacol Res 2023; 197:106971. [PMID: 38032292 DOI: 10.1016/j.phrs.2023.106971] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023]
Abstract
The class B2 of GPCRs known as adhesion G protein-coupled receptors (aGPCRs) has come under increasing academic and nonacademic research focus over the past decade due to their physiological importance as mechano-sensors in cell-cell and cell-matrix contexts. A major advance in understanding signal transduction of aGPCRs was achieved by the identification of the so-called Stachel sequence, which acts as an intramolecular agonist at the interface between the N terminus (Nt) and the seven-transmembrane helix domain (7TMD). Distinct extracellular signals received by the Nt are integrated at the Stachel into structural changes of the 7TMD towards an active state conformation. Until recently, little information was available on how the activation process of aGPCRs is realized at the molecular level. In the past three years several structures of the 7TMD plus the Stachel in complex with G proteins have been determined, which provide new insights into the architecture and molecular function of this receptor class. Herein, we review this structural information to extract common and distinct aGPCR features with particular focus on the Stachel binding site within the 7TMD. Our analysis extends the current view of aGPCR activation and exposes similarities and differences not only between diverse aGPCR members, but also compared to other GPCR classes.
Collapse
Affiliation(s)
- Gunnar Kleinau
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Charitéplatz 1, D-10117 Berlin, Germany
| | - Amal Hassan Ali
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Charitéplatz 1, D-10117 Berlin, Germany
| | - Franziska Wiechert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Berlin, Germany
| | - Michal Szczepek
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Charitéplatz 1, D-10117 Berlin, Germany
| | - Andrea Schmidt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Charitéplatz 1, D-10117 Berlin, Germany
| | - Christian M T Spahn
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Berlin, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Johannisallee 30, 04103 Leipzig, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Johannisallee 30, 04103 Leipzig, Germany; School of Medicine, University of Global Health Equity (UGHE), Kigali, Rwanda.
| | - Patrick Scheerer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Structural Biology of Cellular Signaling, Charitéplatz 1, D-10117 Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
6
|
Erlandson SC, Rawson S, Osei-Owusu J, Brock KP, Liu X, Paulo JA, Mintseris J, Gygi SP, Marks DS, Cong X, Kruse AC. The relaxin receptor RXFP1 signals through a mechanism of autoinhibition. Nat Chem Biol 2023; 19:1013-1021. [PMID: 37081311 PMCID: PMC10530065 DOI: 10.1038/s41589-023-01321-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 03/27/2023] [Indexed: 04/22/2023]
Abstract
The relaxin family peptide receptor 1 (RXFP1) is the receptor for relaxin-2, an important regulator of reproductive and cardiovascular physiology. RXFP1 is a multi-domain G protein-coupled receptor (GPCR) with an ectodomain consisting of a low-density lipoprotein receptor class A (LDLa) module and leucine-rich repeats. The mechanism of RXFP1 signal transduction is clearly distinct from that of other GPCRs, but remains very poorly understood. In the present study, we determine the cryo-electron microscopy structure of active-state human RXFP1, bound to a single-chain version of the endogenous agonist relaxin-2 and the heterotrimeric Gs protein. Evolutionary coupling analysis and structure-guided functional experiments reveal that RXFP1 signals through a mechanism of autoinhibition. Our results explain how an unusual GPCR family functions, providing a path to rational drug development targeting the relaxin receptors.
Collapse
Affiliation(s)
- Sarah C Erlandson
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Shaun Rawson
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - James Osei-Owusu
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Kelly P Brock
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Xinyue Liu
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Joao A Paulo
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Julian Mintseris
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Steven P Gygi
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Debora S Marks
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Xiaojing Cong
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Andrew C Kruse
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Seufert F, Chung YK, Hildebrand PW, Langenhan T. 7TM domain structures of adhesion GPCRs: what's new and what's missing? Trends Biochem Sci 2023; 48:726-739. [PMID: 37349240 DOI: 10.1016/j.tibs.2023.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/05/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023]
Abstract
Adhesion-type G protein-coupled receptors (aGPCRs) have long resisted approaches to resolve the structural details of their heptahelical transmembrane (7TM) domains. Single-particle cryogenic electron microscopy (cryo-EM) has recently produced aGPCR 7TM domain structures for ADGRD1, ADGRG1, ADGRG2, ADGRG3, ADGRG4, ADGRG5, ADGRF1, and ADGRL3. We review the unique properties, including the position and conformation of their activating tethered agonist (TA) and signaling motifs within the 7TM bundle, that the novel structures have helped to identify. We also discuss questions that the kaleidoscope of novel aGPCR 7TM domain structures have left unanswered. These concern the relative positions, orientations, and interactions of the 7TM and GPCR autoproteolysis-inducing (GAIN) domains with one another. Clarifying their interplay remains an important goal of future structural studies on aGPCRs.
Collapse
Affiliation(s)
- Florian Seufert
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstrasse 16-18, 04107 Leipzig, Germany
| | - Yin Kwan Chung
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany
| | - Peter W Hildebrand
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, Härtelstrasse 16-18, 04107 Leipzig, Germany; Institute of Medical Physics and Biophysics, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Tobias Langenhan
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany.
| |
Collapse
|
8
|
Shpakov AO. Allosteric Regulation of G-Protein-Coupled Receptors: From Diversity of Molecular Mechanisms to Multiple Allosteric Sites and Their Ligands. Int J Mol Sci 2023; 24:6187. [PMID: 37047169 PMCID: PMC10094638 DOI: 10.3390/ijms24076187] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Allosteric regulation is critical for the functioning of G protein-coupled receptors (GPCRs) and their signaling pathways. Endogenous allosteric regulators of GPCRs are simple ions, various biomolecules, and protein components of GPCR signaling (G proteins and β-arrestins). The stability and functional activity of GPCR complexes is also due to multicenter allosteric interactions between protomers. The complexity of allosteric effects caused by numerous regulators differing in structure, availability, and mechanisms of action predetermines the multiplicity and different topology of allosteric sites in GPCRs. These sites can be localized in extracellular loops; inside the transmembrane tunnel and in its upper and lower vestibules; in cytoplasmic loops; and on the outer, membrane-contacting surface of the transmembrane domain. They are involved in the regulation of basal and orthosteric agonist-stimulated receptor activity, biased agonism, GPCR-complex formation, and endocytosis. They are targets for a large number of synthetic allosteric regulators and modulators, including those constructed using molecular docking. The review is devoted to the principles and mechanisms of GPCRs allosteric regulation, the multiplicity of allosteric sites and their topology, and the endogenous and synthetic allosteric regulators, including autoantibodies and pepducins. The allosteric regulation of chemokine receptors, proteinase-activated receptors, thyroid-stimulating and luteinizing hormone receptors, and beta-adrenergic receptors are described in more detail.
Collapse
Affiliation(s)
- Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
9
|
Duan J, Xu P, Zhang H, Luan X, Yang J, He X, Mao C, Shen DD, Ji Y, Cheng X, Jiang H, Jiang Y, Zhang S, Zhang Y, Xu HE. Mechanism of hormone and allosteric agonist mediated activation of follicle stimulating hormone receptor. Nat Commun 2023; 14:519. [PMID: 36720854 PMCID: PMC9889800 DOI: 10.1038/s41467-023-36170-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/18/2023] [Indexed: 02/02/2023] Open
Abstract
Follicle stimulating hormone (FSH) is an essential glycoprotein hormone for human reproduction, which functions are mediated by a G protein-coupled receptor, FSHR. Aberrant FSH-FSHR signaling causes infertility and ovarian hyperstimulation syndrome. Here we report cryo-EM structures of FSHR in both inactive and active states, with the active structure bound to FSH and an allosteric agonist compound 21 f. The structures of FSHR are similar to other glycoprotein hormone receptors, highlighting a conserved activation mechanism of hormone-induced receptor activation. Compound 21 f formed extensive interactions with the TMD to directly activate FSHR. Importantly, the unique residue H6157.42 in FSHR plays an essential role in determining FSHR selectivity for various allosteric agonists. Together, our structures provide a molecular basis of FSH and small allosteric agonist-mediated FSHR activation, which could inspire the design of FSHR-targeted drugs for the treatment of infertility and controlled ovarian stimulation for in vitro fertilization.
Collapse
Affiliation(s)
- Jia Duan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Peiyu Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Huibing Zhang
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Immunity and Inflammatory diseases, Hangzhou, Zhejiang, China
| | - Xiaodong Luan
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,School of medicine, Tsinghua university, Beijing, China.,Tsinghua-Peking Center for life science, Tsinghua university, Beijing, China
| | - Jiaqi Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Xinheng He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Chunyou Mao
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Immunity and Inflammatory diseases, Hangzhou, Zhejiang, China
| | - Dan-Dan Shen
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang Provincial Key Laboratory of Immunity and Inflammatory diseases, Hangzhou, Zhejiang, China
| | - Yujie Ji
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Xi Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Hualiang Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China.,Lingang Laboratory, 200031, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Yi Jiang
- Lingang Laboratory, 200031, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China. .,Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China. .,School of medicine, Tsinghua university, Beijing, China. .,Tsinghua-Peking Center for life science, Tsinghua university, Beijing, China.
| | - Yan Zhang
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China. .,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China. .,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China. .,Zhejiang Provincial Key Laboratory of Immunity and Inflammatory diseases, Hangzhou, Zhejiang, China.
| | - H Eric Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China. .,University of Chinese Academy of Sciences, 100049, Beijing, China. .,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China.
| |
Collapse
|
10
|
Núñez Miguel R, Sanders P, Allen L, Evans M, Holly M, Johnson W, Sullivan A, Sanders J, Furmaniak J, Rees Smith B. Structure of full-length TSH receptor in complex with antibody K1-70™. J Mol Endocrinol 2023; 70:e220120. [PMID: 36069797 PMCID: PMC9782461 DOI: 10.1530/jme-22-0120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/06/2022] [Indexed: 01/19/2023]
Abstract
Determination of the full-length thyroid-stimulating hormone receptor (TSHR) structure by cryo-electron microscopy (cryo-EM) is described. The TSHR complexed with human monoclonal TSHR autoantibody K1-70™ (a powerful inhibitor of TSH action) was detergent solubilised, purified to homogeneity and analysed by cryo-EM. The structure (global resolution 3.3 Å) is a monomer with all three domains visible: leucine-rich domain (LRD), hinge region (HR) and transmembrane domain (TMD). The TSHR extracellular domain (ECD, composed of the LRD and HR) is positioned on top of the TMD extracellular surface. Extensive interactions between the TMD and ECD are observed in the structure, and their analysis provides an explanation of the effects of various TSHR mutations on TSHR constitutive activity and on ligand-induced activation. K1-70™ is seen to be well clear of the lipid bilayer. However, superimposition of M22™ (a human monoclonal TSHR autoantibody which is a powerful stimulator of the TSHR) on the cryo-EM structure shows that it would clash with the bilayer unless the TSHR HR rotates upwards as part of the M22™ binding process. This rotation could have an important role in TSHR stimulation by M22™ and as such provides an explanation as to why K1-70™ blocks the binding of TSH and M22™ without activating the receptor itself.
Collapse
Affiliation(s)
| | - Paul Sanders
- FIRS Laboratories, RSR Ltd, Parc Ty Glas, Llanishen, Cardiff, UK
| | - Lloyd Allen
- FIRS Laboratories, RSR Ltd, Parc Ty Glas, Llanishen, Cardiff, UK
| | - Michele Evans
- FIRS Laboratories, RSR Ltd, Parc Ty Glas, Llanishen, Cardiff, UK
| | - Matthew Holly
- FIRS Laboratories, RSR Ltd, Parc Ty Glas, Llanishen, Cardiff, UK
| | - William Johnson
- FIRS Laboratories, RSR Ltd, Parc Ty Glas, Llanishen, Cardiff, UK
| | - Andrew Sullivan
- FIRS Laboratories, RSR Ltd, Parc Ty Glas, Llanishen, Cardiff, UK
| | - Jane Sanders
- FIRS Laboratories, RSR Ltd, Parc Ty Glas, Llanishen, Cardiff, UK
| | | | | |
Collapse
|
11
|
Nagayama Y, Nishihara E. Thyrotropin receptor antagonists and inverse agonists, and their potential application to thyroid diseases. Endocr J 2022; 69:1285-1293. [PMID: 36171093 DOI: 10.1507/endocrj.ej22-0391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The thyrotropin receptor (TSHR) plays critical roles in thyroid growth and function and in the pathogenesis of several thyroid diseases including Graves' hyperthyroidism and ophthalmopathy, non-autoimmune hyperthyroidism and thyroid cancer. Several low-molecular weight compounds (LMWCs) and anti-TSHR monoclonal antibodies (mAbs) with receptor antagonistic and inverse agonistic activities have been reported. The former binds to the pocket formed by the receptor transmembrane bundle, and the latter to the extracellular TSH binding site. Both are effective inhibitors of TSH/thyroid stimulating antibody-stimulated cAMP and/or hyaluronic acid production in TSHR-expressing cells. Anti-insulin-like growth factor 1 inhibitors are also found to inhibit TSHR signaling. Each agent has advantages and disadvantages; for example, mAbs have a higher affinity and longer half-life but are more costly than LMWCs. At present, mAbs appear most promising, yet the development of more efficacious LMWCs is desirable. These agents are anticipated to be efficacious not only for the above-mentioned diseases but also for resistance to thyroid hormone and have utility for thyroid cancer radionuclide scintigraphy/therapy as a new theranostic.
Collapse
Affiliation(s)
- Yuji Nagayama
- Department of Molecular Medicine, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki 852-8523, Japan
| | - Eijun Nishihara
- Center for Excellence in Thyroid Care, Kuma Hospital, Kobe 650-0011, Japan
| |
Collapse
|
12
|
Hinge Region Mediates Signal Transmission of Luteinizing Hormone and Chorionic Gonadotropin Receptor. Comput Struct Biotechnol J 2022; 20:6503-6511. [DOI: 10.1016/j.csbj.2022.11.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/19/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022] Open
|
13
|
Faust B, Billesbølle CB, Suomivuori CM, Singh I, Zhang K, Hoppe N, Pinto AFM, Diedrich JK, Muftuoglu Y, Szkudlinski MW, Saghatelian A, Dror RO, Cheng Y, Manglik A. Autoantibody mimicry of hormone action at the thyrotropin receptor. Nature 2022; 609:846-853. [PMID: 35940205 PMCID: PMC9678024 DOI: 10.1038/s41586-022-05159-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 07/28/2022] [Indexed: 11/08/2022]
Abstract
Thyroid hormones are vital in metabolism, growth and development1. Thyroid hormone synthesis is controlled by thyrotropin (TSH), which acts at the thyrotropin receptor (TSHR)2. In patients with Graves' disease, autoantibodies that activate the TSHR pathologically increase thyroid hormone activity3. How autoantibodies mimic thyrotropin function remains unclear. Here we determined cryo-electron microscopy structures of active and inactive TSHR. In inactive TSHR, the extracellular domain lies close to the membrane bilayer. Thyrotropin selects an upright orientation of the extracellular domain owing to steric clashes between a conserved hormone glycan and the membrane bilayer. An activating autoantibody from a patient with Graves' disease selects a similar upright orientation of the extracellular domain. Reorientation of the extracellular domain transduces a conformational change in the seven-transmembrane-segment domain via a conserved hinge domain, a tethered peptide agonist and a phospholipid that binds within the seven-transmembrane-segment domain. Rotation of the TSHR extracellular domain relative to the membrane bilayer is sufficient for receptor activation, revealing a shared mechanism for other glycoprotein hormone receptors that may also extend to other G-protein-coupled receptors with large extracellular domains.
Collapse
MESH Headings
- Cell Membrane/metabolism
- Cryoelectron Microscopy
- Graves Disease/immunology
- Graves Disease/metabolism
- Humans
- Immunoglobulins, Thyroid-Stimulating/chemistry
- Immunoglobulins, Thyroid-Stimulating/immunology
- Immunoglobulins, Thyroid-Stimulating/pharmacology
- Immunoglobulins, Thyroid-Stimulating/ultrastructure
- Phospholipids/metabolism
- Protein Domains
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/chemistry
- Receptors, G-Protein-Coupled/ultrastructure
- Receptors, Thyrotropin/agonists
- Receptors, Thyrotropin/chemistry
- Receptors, Thyrotropin/immunology
- Receptors, Thyrotropin/ultrastructure
- Rotation
- Thyrotropin/chemistry
- Thyrotropin/metabolism
- Thyrotropin/pharmacology
Collapse
Affiliation(s)
- Bryan Faust
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
- Biophysics Graduate Program, University of California, San Francisco, CA, USA
| | | | - Carl-Mikael Suomivuori
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Isha Singh
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Kaihua Zhang
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Nicholas Hoppe
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Biophysics Graduate Program, University of California, San Francisco, CA, USA
| | - Antonio F M Pinto
- Mass Spectrometry Core for Proteomics and Metabolomics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jolene K Diedrich
- Mass Spectrometry Core for Proteomics and Metabolomics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | | | - Alan Saghatelian
- Clayton Foundation Laboratory for Peptide Biology Lab, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ron O Dror
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA.
- Biophysics Graduate Program, University of California, San Francisco, CA, USA.
- Howard Hughes Medical Institute, University of California, San Francisco, CA, USA.
| | - Aashish Manglik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA.
- Biophysics Graduate Program, University of California, San Francisco, CA, USA.
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
14
|
Fokina EF, Shpakov AO. Thyroid-Stimulating Hormone Receptor: the Role in the Development of Thyroid Pathology and Its Correction. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022050143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Abstract
One of the key elements responsible for the thyroid response
to thyroid-stimulating hormone (TSH) is the TSH receptor (TSHR),
which belongs to the G protein-coupled receptor superfamily. Binding
of TSH or stimulatory autoantibodies to the TSHR extracellular domain
triggers multiple signaling pathways in target cells that are mediated
through various types of G proteins and β-arrestins. Inhibitory
autoantibodies, in contrast, suppress TSHR activity, inducing hypothyroid states.
Activating mutations lead to constitutively active TSHR forms and
can trigger cancer. Therefore, the TSHR is one of the key targets
for the regulation of thyroid function and thyroid status, as well
as correction of diseases caused by changes in TSHR activity (autoimmune
hyper- and hypothyroidism, Graves’ ophthalmopathy, thyroid cancer).
TSH preparations are extremely rarely used in medicine due to their
immunogenicity and severe side effects. Most promising is the development
of low-molecular allosteric TSHR regulators with an activity of
full and inverse agonists and neutral antagonists, which are able
to penetrate into the allosteric site located in the TSHR transmembrane
domain and specifically bind to it, thus controlling the ability
of the receptor to interact with G proteins and β-arrestins. Allosteric
regulators do not affect the binding of TSH and autoantibodies to
the receptor, which enables mild and selective regulation of thyroid function,
while avoiding critical changes in TSH and thyroid hormone levels.
The present review addresses the current state of the problem of
regulating TSHR activity, including the possibility of using ligands
of its allosteric sites.
Collapse
|
15
|
Nagel M, Moretti R, Paschke R, von Bergen M, Meiler J, Kalkhof S. Integrative model of the FSH receptor reveals the structural role of the flexible hinge region. Structure 2022; 30:1424-1431.e3. [PMID: 35973423 DOI: 10.1016/j.str.2022.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/16/2021] [Accepted: 07/20/2022] [Indexed: 11/16/2022]
Abstract
The follicle-stimulating hormone receptor (FSHR) belongs to the glycoprotein hormone receptors, a subfamily of G-protein-coupled receptors (GPCRs). FSHR is involved in reproductive processes such as gonadal development and maturation. Structurally, the extensive extracellular domain, which contains the hormone-binding site and is linked to the transmembrane domain by the hinge region (HR), is characteristic for these receptors. How this HR is involved in hormone binding and signal transduction is still an open question. We combined in vitro and in situ chemical crosslinking, disulfide pattern analysis, and mutation data with molecular modeling to generate experimentally driven full-length models. These models provide insights into the interface, important side-chain interactions, and activation mechanism. The interface indicates a strong involvement of the connecting loop. A major rearrangement of the HR seems implausible due to the tight arrangement and fixation by disulfide bonds. The models are expected to allow for testable hypotheses about signal transduction and drug development for GPHRs.
Collapse
Affiliation(s)
- Marcus Nagel
- Department for Molecular Systems Biology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany; Center for Structural Biology, Vanderbilt University, Nashville, TN 37212, USA; Division of Endocrinology, Department of Endocrinology and Nephrology, University Clinic Leipzig, Germany
| | - Rocco Moretti
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37212, USA
| | - Ralf Paschke
- Division of Endocrinology, Department of Endocrinology and Nephrology, University Clinic Leipzig, Germany; Department of Medicine, Division of Endocrinology, Departments of Oncology, Pathology, and Biochemistry and Molecular Biology & Arnie Charbonneau Cancer Institute Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Martin von Bergen
- Department for Molecular Systems Biology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany; Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, Leipzig, Germany
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37212, USA; Leipzig University Medical School, Institute for Drug Discovery, 04103 Leipzig, Germany.
| | - Stefan Kalkhof
- Department for Molecular Systems Biology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany; Institute for Bioanalysis, University of Applied Sciences Coburg, Coburg, Germany; Fraunhofer Institute for Cell Therapy and Immunology, Department of Preclinical Development and Validation, 04103 Leipzig, Germany.
| |
Collapse
|
16
|
Hormone- and antibody-mediated activation of the thyrotropin receptor. Nature 2022; 609:854-859. [PMID: 35940204 DOI: 10.1038/s41586-022-05173-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 08/02/2022] [Indexed: 11/08/2022]
Abstract
Thyroid stimulating hormone (TSH), through activation of its G protein-coupled thyrotropin receptor (TSHR), controls the synthesis of thyroid hormone (TH), an essential metabolic hormone1-3. Aberrant signaling of TSHR by autoantibodies causes Graves' disease and hypothyroidism that affect millions of patients worldwide4. Here we report the active structures of TSHR with TSH and an activating autoantibody M225, both bound to an allosteric agonist ML-1096, as well as an inactivated TSHR structure with inhibitory antibody K1-707. Both TSH and M22 push the extracellular domain (ECD) of TSHR into the upright active conformation. In contrast, K1-70 blocks TSH binding and is incapable of pushing the ECD to the upright conformation. Comparisons of the active and inactivated structures of TSHR with those of the luteinizing hormone-choriogonadotropin receptor (LHCGR) reveal a universal activation mechanism of glycoprotein hormone receptors, in which a conserved 10-residue fragment (P10) from the hinge C-terminal loop mediates ECD interactions with the TSHR transmembrane domain8. One surprisingly feature is that there are over 15 cholesterols surrounding TSHR, supporting its preferential location in lipid rafts9. These structures also highlight a similar ECD-push mechanism for TSH and autoantibody M22 to activate TSHR, thus providing the molecular basis for Graves' disease.
Collapse
|
17
|
Speck D, Kleinau G, Meininghaus M, Erbe A, Einfeldt A, Szczepek M, Scheerer P, Pütter V. Expression and Characterization of Relaxin Family Peptide Receptor 1 Variants. Front Pharmacol 2022; 12:826112. [PMID: 35153771 PMCID: PMC8832513 DOI: 10.3389/fphar.2021.826112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/31/2021] [Indexed: 12/31/2022] Open
Abstract
G-protein coupled receptors (GPCR) transduce extracellular stimuli into the cell interior and are thus centrally involved in almost all physiological-neuronal processes. This essential function and association with many diseases or pathological conditions explain why GPCRs are one of the priority targets in medical and pharmacological research, including structure determination. Despite enormous experimental efforts over the last decade, both the expression and purification of these membrane proteins remain elusive. This is attributable to specificities of each GPCR subtype and the finding of necessary experimental in vitro conditions, such as expression in heterologous cell systems or with accessory proteins. One of these specific GPCRs is the leucine-rich repeat domain (LRRD) containing GPCR 7 (LGR7), also termed relaxin family peptide receptor 1 (RXFP1). This receptor is characterized by a large extracellular region of around 400 amino acids constituted by several domains, a rare feature among rhodopsin-like (class A) GPCRs. In the present study, we describe the expression and purification of RXFP1, including the design of various constructs suitable for functional/biophysical studies and structure determination. Based on available sequence information, homology models, and modern biochemical and genetic tools, several receptor variations with different purification tags and fusion proteins were prepared and expressed in Sf9 cells (small-scale), followed by an analytic fluorescence-detection size-exclusion chromatography (F-SEC) to evaluate the constructs. The most promising candidates were expressed and purified on a large-scale, accompanied by ligand binding studies using surface plasmon resonance spectroscopy (SPR) and by determination of signaling capacities. The results may support extended studies on RXFP1 receptor constructs serving as targets for small molecule ligand screening or structural elucidation by protein X-ray crystallography or cryo-electron microscopy.
Collapse
Affiliation(s)
- David Speck
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Protein X-ray Crystallography & Signal Transduction, Berlin, Germany
| | - Gunnar Kleinau
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Protein X-ray Crystallography & Signal Transduction, Berlin, Germany
| | - Mark Meininghaus
- Bayer AG, Research and Development, Pharmaceuticals, Wuppertal, Germany
| | - Antje Erbe
- Bayer AG, Research and Development, Pharmaceuticals, Berlin, Germany
- NUVISAN ICB GmbH, Berlin, Germany
| | - Alexandra Einfeldt
- Bayer AG, Research and Development, Pharmaceuticals, Berlin, Germany
- NUVISAN ICB GmbH, Berlin, Germany
| | - Michal Szczepek
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Protein X-ray Crystallography & Signal Transduction, Berlin, Germany
| | - Patrick Scheerer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Physics and Biophysics, Group Protein X-ray Crystallography & Signal Transduction, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- *Correspondence: Patrick Scheerer, ; Vera Pütter,
| | - Vera Pütter
- Bayer AG, Research and Development, Pharmaceuticals, Berlin, Germany
- NUVISAN ICB GmbH, Berlin, Germany
- *Correspondence: Patrick Scheerer, ; Vera Pütter,
| |
Collapse
|
18
|
Structural Insights into the Unique Modes of Relaxin-Binding and Tethered-Agonist Mediated Activation of RXFP1 and RXFP2. J Mol Biol 2021; 433:167217. [PMID: 34454945 DOI: 10.1016/j.jmb.2021.167217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/09/2021] [Accepted: 08/19/2021] [Indexed: 01/01/2023]
Abstract
Our poor understanding of the mechanism by which the peptide-hormone H2 relaxin activates its G protein coupled receptor, RXFP1 and the related receptor RXFP2, has hindered progress in its therapeutic development. Both receptors possess large ectodomains, which bind H2 relaxin, and contain an N-terminal LDLa module that is essential for receptor signaling and postulated to be a tethered agonist. Here, we show that a conserved motif (GDxxGWxxxF), C-terminal to the LDLa module, is critical for receptor activity. Importantly, this motif adopts different structures in RXFP1 and RXFP2, suggesting distinct activation mechanisms. For RXFP1, the motif is flexible, weakly associates with the LDLa module, and requires H2 relaxin binding to stabilize an active conformation. Conversely, the GDxxGWxxxF motif in RXFP2 is more closely associated with the LDLa module, forming an essential binding interface for H2 relaxin. These differences in the activation mechanism will aid drug development targeting these receptors.
Collapse
|
19
|
Duan J, Xu P, Cheng X, Mao C, Croll T, He X, Shi J, Luan X, Yin W, You E, Liu Q, Zhang S, Jiang H, Zhang Y, Jiang Y, Xu HE. Structures of full-length glycoprotein hormone receptor signalling complexes. Nature 2021; 598:688-692. [PMID: 34552239 DOI: 10.1038/s41586-021-03924-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 08/18/2021] [Indexed: 11/09/2022]
Abstract
Luteinizing hormone and chorionic gonadotropin are glycoprotein hormones that are related to follicle-stimulating hormone and thyroid-stimulating hormone1,2. Luteinizing hormone and chorionic gonadotropin are essential to human reproduction and are important therapeutic drugs3-6. They activate the same G-protein-coupled receptor, luteinizing hormone-choriogonadotropin receptor (LHCGR), by binding to the large extracellular domain3. Here we report four cryo-electron microscopy structures of LHCGR: two structures of the wild-type receptor in the inactive and active states; and two structures of the constitutively active mutated receptor. The active structures are bound to chorionic gonadotropin and the stimulatory G protein (Gs), and one of the structures also contains Org43553, an allosteric agonist7. The structures reveal a distinct 'push-and-pull' mechanism of receptor activation, in which the extracellular domain is pushed by the bound hormone and pulled by the extended hinge loop next to the transmembrane domain. A highly conserved 10-residue fragment (P10) from the hinge C-terminal loop at the interface between the extracellular domain and the transmembrane domain functions as a tethered agonist to induce conformational changes in the transmembrane domain and G-protein coupling. Org43553 binds to a pocket of the transmembrane domain and interacts directly with P10, which further stabilizes the active conformation. Together, these structures provide a common model for understanding the signalling of glycoprotein hormone receptors and a basis for drug discovery for endocrine diseases.
Collapse
Affiliation(s)
- Jia Duan
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Peiyu Xu
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xi Cheng
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Chunyou Mao
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, China.,Zheijang Provincial Key Laboratory of Immunity and Inflammatory Diseases, Hangzhou, China.,Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tristan Croll
- Cambridge Institute for Medical Research, Department of Haematology, University of Cambridge, Cambridge, UK
| | - Xinheng He
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jingjing Shi
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xiaodong Luan
- School of Medicine, Tsinghua University, Beijing, China.,Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Wanchao Yin
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Erli You
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Qiufeng Liu
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shuyang Zhang
- School of Medicine, Tsinghua University, Beijing, China.,Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Hualiang Jiang
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yan Zhang
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China. .,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, China. .,Zheijang Provincial Key Laboratory of Immunity and Inflammatory Diseases, Hangzhou, China.
| | - Yi Jiang
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China.
| | - H Eric Xu
- The CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
20
|
Ostróżka-Cieślik A, Dolińska B, Ryszka F. Effectiveness Assessment of a Modified Preservation Solution Containing Thyrotropin or Follitropin Based on Biochemical Analysis in Perfundates and Homogenates of Isolated Porcine Kidneys after Static Cold Storage. Int J Mol Sci 2021; 22:ijms22168360. [PMID: 34445068 PMCID: PMC8395071 DOI: 10.3390/ijms22168360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/29/2021] [Accepted: 07/31/2021] [Indexed: 12/15/2022] Open
Abstract
In this paper, we assess the nephroprotective effects of thyrotropin and follitropin during ischaemia. The studies were performed in vitro in a model of isolated porcine kidneys stored in Biolasol (FZNP, Biochefa, Sosnowiec, Poland) and modified Biolasol (TSH: 1 µg/L; FSH 1 µg/L). We used the static cold storage method. The study was carried out based on 30 kidneys. The kidneys were placed in 500 mL of preservation solution chilled to 4 °C. The samples for biochemical tests were collected during the first kidney perfusion (after 2 h of storage) and during the second perfusion (after 48 h of storage). The results of ALT, AST, and LDH activities confirm the effectiveness of Biolasol + p-TSH in maintaining the structural integrity of renal cell membranes. Significantly reduced biochemical parameters of kidney function, i.e., creatinine and protein concentrations were also observed after 48 h storage. The protective effect of Biasol + p-TSH is most pronounced after 2 h of storage, suggesting a mild course of damage thereafter. A mild deterioration of renal function was observed after 48 h. The results of our analyses did not show any protective effect of Biolasol + p-FSH on the kidneys during ischaemia.
Collapse
Affiliation(s)
- Aneta Ostróżka-Cieślik
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Kasztanowa 3, 41-200 Sosnowiec, Poland;
- Correspondence:
| | - Barbara Dolińska
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Kasztanowa 3, 41-200 Sosnowiec, Poland;
- “Biochefa” Pharmaceutical Research and Production Plant, Kasztanowa 3, 41-200 Sosnowiec, Poland;
| | - Florian Ryszka
- “Biochefa” Pharmaceutical Research and Production Plant, Kasztanowa 3, 41-200 Sosnowiec, Poland;
| |
Collapse
|
21
|
Sveidahl Johansen O, Ma T, Hansen JB, Markussen LK, Schreiber R, Reverte-Salisa L, Dong H, Christensen DP, Sun W, Gnad T, Karavaeva I, Nielsen TS, Kooijman S, Cero C, Dmytriyeva O, Shen Y, Razzoli M, O'Brien SL, Kuipers EN, Nielsen CH, Orchard W, Willemsen N, Jespersen NZ, Lundh M, Sustarsic EG, Hallgren CM, Frost M, McGonigle S, Isidor MS, Broholm C, Pedersen O, Hansen JB, Grarup N, Hansen T, Kjær A, Granneman JG, Babu MM, Calebiro D, Nielsen S, Rydén M, Soccio R, Rensen PCN, Treebak JT, Schwartz TW, Emanuelli B, Bartolomucci A, Pfeifer A, Zechner R, Scheele C, Mandrup S, Gerhart-Hines Z. Lipolysis drives expression of the constitutively active receptor GPR3 to induce adipose thermogenesis. Cell 2021; 184:3502-3518.e33. [PMID: 34048700 PMCID: PMC8238500 DOI: 10.1016/j.cell.2021.04.037] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 02/10/2021] [Accepted: 04/23/2021] [Indexed: 12/19/2022]
Abstract
Thermogenic adipocytes possess a therapeutically appealing, energy-expending capacity, which is canonically cold-induced by ligand-dependent activation of β-adrenergic G protein-coupled receptors (GPCRs). Here, we uncover an alternate paradigm of GPCR-mediated adipose thermogenesis through the constitutively active receptor, GPR3. We show that the N terminus of GPR3 confers intrinsic signaling activity, resulting in continuous Gs-coupling and cAMP production without an exogenous ligand. Thus, transcriptional induction of Gpr3 represents the regulatory parallel to ligand-binding of conventional GPCRs. Consequently, increasing Gpr3 expression in thermogenic adipocytes is alone sufficient to drive energy expenditure and counteract metabolic disease in mice. Gpr3 transcription is cold-stimulated by a lipolytic signal, and dietary fat potentiates GPR3-dependent thermogenesis to amplify the response to caloric excess. Moreover, we find GPR3 to be an essential, adrenergic-independent regulator of human brown adipocytes. Taken together, our findings reveal a noncanonical mechanism of GPCR control and thermogenic activation through the lipolysis-induced expression of constitutively active GPR3.
Collapse
Affiliation(s)
- Olivia Sveidahl Johansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Embark Biotech ApS, Copenhagen, Denmark; Center for Adipocyte Signaling, University of Southern Denmark, Odense, Denmark
| | - Tao Ma
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Embark Biotech ApS, Copenhagen, Denmark
| | - Jakob Bondo Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Embark Biotech ApS, Copenhagen, Denmark
| | - Lasse Kruse Markussen
- Center for Adipocyte Signaling, University of Southern Denmark, Odense, Denmark; Functional Genomics and Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Laia Reverte-Salisa
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Hua Dong
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | | | - Wenfei Sun
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Thorsten Gnad
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Iuliia Karavaeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Svava Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Cheryl Cero
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Oksana Dmytriyeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Yachen Shen
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Shannon L O'Brien
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK; Institute of Pharmacology and Toxicology and Bio-Imaging Center, University of Würzburg, Würzburg, Germany
| | - Eline N Kuipers
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Carsten Haagen Nielsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen, Denmark
| | | | - Nienke Willemsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Naja Zenius Jespersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Morten Lundh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Elahu Gosney Sustarsic
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Cecilie Mørch Hallgren
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Frost
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Seth McGonigle
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Marie Sophie Isidor
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Christa Broholm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Bo Hansen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Kjær
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet, Copenhagen, Denmark
| | - James G Granneman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - M Madan Babu
- MRC Laboratory of Molecular Biology, Cambridge, UK; Department of Structural Biology and Center for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Davide Calebiro
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK; Institute of Pharmacology and Toxicology and Bio-Imaging Center, University of Würzburg, Würzburg, Germany
| | - Søren Nielsen
- Centre of Inflammation and Metabolism and Centre for Physical Activity Research, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Raymond Soccio
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jonas Thue Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Thue Walter Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Embark Biotech ApS, Copenhagen, Denmark
| | - Brice Emanuelli
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Mandrup
- Center for Adipocyte Signaling, University of Southern Denmark, Odense, Denmark; Functional Genomics and Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Embark Biotech ApS, Copenhagen, Denmark; Center for Adipocyte Signaling, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
22
|
Functional impact of intramolecular cleavage and dissociation of adhesion G protein-coupled receptor GPR133 (ADGRD1) on canonical signaling. J Biol Chem 2021; 296:100798. [PMID: 34022221 PMCID: PMC8215292 DOI: 10.1016/j.jbc.2021.100798] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/04/2021] [Accepted: 05/16/2021] [Indexed: 12/22/2022] Open
Abstract
GPR133 (ADGRD1), an adhesion G protein–coupled receptor (GPCR) whose canonical signaling activates GαS-mediated generation of cytosolic cAMP, has been shown to be necessary for the growth of glioblastoma (GBM), a brain malignancy. The extracellular N terminus of GPR133 is thought to be autoproteolytically cleaved into N-terminal and C- terminal fragments (NTF and CTF, respectively). However, the role of this cleavage in receptor activation remains unclear. Here, we used subcellular fractionation and immunoprecipitation approaches to show that the WT GPR133 receptor is cleaved shortly after protein synthesis and generates significantly more canonical signaling than an uncleavable point mutant GPR133 (H543R) in patient-derived GBM cultures and HEK293T cells. After cleavage, the resulting NTF and CTF remain noncovalently bound to each other until the receptor is trafficked to the plasma membrane, where we demonstrated NTF–CTF dissociation occurs. Using a fusion of the CTF of GPR133 and the N terminus of thrombin-activated human protease-activated receptor 1 as a controllable proxy system to test the effect of intramolecular cleavage and dissociation, we also showed that thrombin-induced cleavage and shedding of the human protease-activated receptor 1 NTF increased intracellular cAMP levels. These results support a model wherein dissociation of the NTF from the CTF at the plasma membrane promotes GPR133 activation and downstream signaling. These findings add depth to our understanding of the molecular life cycle and mechanism of action of GPR133 and provide critical insights that will inform therapeutic targeting of GPR133 in GBM.
Collapse
|
23
|
Newton CL, Anderson RC, Kreuchwig A, Krause G, Katz AA, Millar RP. Rescue of Function of Mutant Luteinising Hormone Receptors with Deficiencies in Cell Surface Expression, Hormone Binding, and Hormone Signalling. Neuroendocrinology 2021; 111:451-464. [PMID: 32316022 DOI: 10.1159/000508000] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 04/18/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION G protein-coupled receptor (GPCR) mutations are implicated in many diseases. Most inactivating mutations cause receptor misfolding and prevent trafficking to the plasma membrane. Pharmacological chaperones can "rescue" cell surface expression of such mutants, presumably by stabilising correct folding of the nascent protein. OBJECTIVE Here we examine the scope of intracellularly retained luteinising hormone receptor (LHR) mutants that can be "rescued" by the pharmacological chaperone LHR-Chap, and whether this allosteric agonist can also restore the function of mutant LHRs with deficiencies in hormone binding or hormone-induced signalling. METHODS Mutant LHRs were expressed in HEK 293-T cells. Cell surface expression/localisation, hormone binding, and hCG/LHR-Chap signalling were determined by ELISA, radioligand binding, and inositol phosphate accumulation assays, respectively. Molecular modelling predicted LHR-Chap interactions. RESULTS LHR-Chap increased cell surface expression of a subset of retained mutants located in transmembrane helices predicted to be stabilised by LHR-Chap binding. For 3 (T4613.47I, L5024.61P, and S6167.46Y) hCG-responsiveness was increased following treatment. LHRs with mutations in the hormone-binding site (C131ECDR and I152ECDT) or in the hinge region (E354HingeK) had good cell surface expression but poor response to hormone stimulation, yet were responsive to allosteric activation by LHR-Chap. CONCLUSIONS LHR-Chap, in addition to rescuing cell surface expression of intracellularly retained LHR mutants, can rescue function in mutant receptors with binding and signalling deficiencies that have normal cell surface expression. This demonstration of rescue of multiple elements of LHR dysfunction arising from inactivating mutations offers exceptional potential for treating patients with diseases arising from GPCR mutations in general.
Collapse
Affiliation(s)
- Claire Louise Newton
- Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa,
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa,
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom,
| | - Ross Calley Anderson
- Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Annika Kreuchwig
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Gerd Krause
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Arieh Anthony Katz
- UCT Receptor Biology Research Unit and SAMRC Gynaecology Cancer Research Centre, Department of Integrative Biomedical Sciences and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Robert Peter Millar
- Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- School of Medicine, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
24
|
Krause G, Eckstein A, Schülein R. Modulating TSH Receptor Signaling for Therapeutic Benefit. Eur Thyroid J 2020; 9:66-77. [PMID: 33511087 PMCID: PMC7802447 DOI: 10.1159/000511871] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022] Open
Abstract
Autoimmune thyroid-stimulating antibodies are activating the thyrotropin receptor (TSHR) in both the thyroid and the eye, but different molecular mechanisms are induced in both organs, leading to Graves' disease (GD) and Graves' orbitopathy (GO), respectively. Therapy with anti-thyroid drugs to reduce hyperthyroidism (GD) by suppressing the biosynthesis of thyroid hormones has only an indirect effect on GO, since it does not causally address pathogenic TSHR activation itself. GO is thus very difficult to treat. The activated TSHR but also the cross-interacting insulin-like growth factor 1 receptor (IGF-1R) contribute to this issue. The TSHR is a heptahelical G-protein-coupled receptor, whereas the IGF-1R is a receptor tyrosine kinase. Despite these fundamental structural differences, both receptors are phosphorylated by G-protein receptor kinases, which enables β-arrestin binding. Arrestins mediate receptor internalization and also activate the mitogen-activated protein kinase pathway. Moreover, emerging results suggest that arrestin plays a critical role in the cross-interaction of the TSHR and the IGF-1R either in their common signaling pathway and/or during an indirect or potential TSHR/IGF-1R interaction. In this review, novel pharmacological strategies with allosteric small-molecule modulators to treat GO and GD on the level of the TSHR and/or the TSHR/IGF-1R cross-interaction will be discussed. Moreover, monoclonal antibody approaches targeting the TSHR or the IGF-1R and thereby preventing activation of either receptor will be presented. Another chapter addresses the immunomodulation to treat GO using TSHR-derived peptides targeting the human leukocyte antigen DR isotope (HLA-DR), which is a feasible approach to tackle GO, since HLA-DR and TSHR are overexpressed in orbital tissues of GO patients.
Collapse
Affiliation(s)
- Gerd Krause
- Structural Biology, Leibniz-Forschungsinstitut für molekulare Pharmakologie (FMP), Berlin, Germany
| | - Anja Eckstein
- Department of Ophthalmology, University Hospital Essen, Essen, Germany
| | - Ralf Schülein
- Protein Trafficking, Leibniz-Forschungsinstitut für molekulare Pharmakologie (FMP), Berlin, Germany
| |
Collapse
|
25
|
Schöneberg T, Liebscher I. Mutations in G Protein-Coupled Receptors: Mechanisms, Pathophysiology and Potential Therapeutic Approaches. Pharmacol Rev 2020; 73:89-119. [PMID: 33219147 DOI: 10.1124/pharmrev.120.000011] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
There are approximately 800 annotated G protein-coupled receptor (GPCR) genes, making these membrane receptors members of the most abundant gene family in the human genome. Besides being involved in manifold physiologic functions and serving as important pharmacotherapeutic targets, mutations in 55 GPCR genes cause about 66 inherited monogenic diseases in humans. Alterations of nine GPCR genes are causatively involved in inherited digenic diseases. In addition to classic gain- and loss-of-function variants, other aspects, such as biased signaling, trans-signaling, ectopic expression, allele variants of GPCRs, pseudogenes, gene fusion, and gene dosage, contribute to the repertoire of GPCR dysfunctions. However, the spectrum of alterations and GPCR involvement is probably much larger because an additional 91 GPCR genes contain homozygous or hemizygous loss-of-function mutations in human individuals with currently unidentified phenotypes. This review highlights the complexity of genomic alteration of GPCR genes as well as their functional consequences and discusses derived therapeutic approaches. SIGNIFICANCE STATEMENT: With the advent of new transgenic and sequencing technologies, the number of monogenic diseases related to G protein-coupled receptor (GPCR) mutants has significantly increased, and our understanding of the functional impact of certain kinds of mutations has substantially improved. Besides the classical gain- and loss-of-function alterations, additional aspects, such as biased signaling, trans-signaling, ectopic expression, allele variants of GPCRs, uniparental disomy, pseudogenes, gene fusion, and gene dosage, need to be elaborated in light of GPCR dysfunctions and possible therapeutic strategies.
Collapse
Affiliation(s)
- Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig, Germany
| |
Collapse
|
26
|
Atre I, Mizrahi N, Yebra-Pimentel ES, Hausken K, Yom-Din S, Hurvitz A, Dirks R, Degani G, Levavi-Sivan B. Molecular characterization of two Russian sturgeon gonadotropin receptors: Cloning, expression analysis, and functional activity. Gen Comp Endocrinol 2020; 298:113557. [PMID: 32687934 DOI: 10.1016/j.ygcen.2020.113557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 06/24/2020] [Accepted: 07/14/2020] [Indexed: 10/23/2022]
Abstract
Sturgeons are being used in aquaculture because wild populations are now endangered due to overfishing for caviar. A challenge in working with sturgeon as an aquacultured species is its long and slow reproductive development. Reproduction is a hormonally regulated process that involves hierarchical signaling between the brain, pituitary gland, and gonads. In an effort to better understand the hormonal regulation of sturgeon reproduction, we have cloned the Russian sturgeon (st), Acipenser gueldenstaedtii, luteinizing hormone receptor (stLHR) and follicle stimulating hormone receptor (stFSHR) and measured their expression from previtellogenic to mature ovarian follicles. Sturgeon LHR and FSHR expression was elevated in early-vitellogenic and mature follicles compared with pre-vitellogenic and mid-vitellogenic follicles, and only LHR expression increased during late-vitellogenesis. Recombinant sturgeon FSH and LH both activated sturgeon LHR and FSHR in a cAMP reporter assay. Further molecular characterization of these receptors was accomplished by in silico modeling and cAMP reporter assays using heterologous recombinant gonadotropins from human and piscine species. There was no apparent trend in heterologous LH and/or FSH activation of the sturgeon LHR or FSHR. These data suggest that permissive activation of LHR and FSHR are a consequence of some yet undetermined biological characteristic(s) of different piscine species.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Cloning, Molecular
- Female
- Gene Expression Regulation
- Humans
- Models, Molecular
- Phylogeny
- Protein Domains
- Receptors, FSH/chemistry
- Receptors, FSH/genetics
- Receptors, FSH/metabolism
- Receptors, Gonadotropin/chemistry
- Receptors, Gonadotropin/genetics
- Receptors, Gonadotropin/metabolism
- Receptors, LH/chemistry
- Receptors, LH/genetics
- Receptors, LH/metabolism
- Russia
Collapse
Affiliation(s)
- Ishwar Atre
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Naama Mizrahi
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel
| | | | - Krist Hausken
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Svetlana Yom-Din
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel; MIGAL - Galilee Technology Center, P.O. Box 831, Kiryat Shmona 10200, Israel
| | | | - Ron Dirks
- ZF-screens B.V., Leiden, the Netherlands
| | - Gad Degani
- MIGAL - Galilee Technology Center, P.O. Box 831, Kiryat Shmona 10200, Israel
| | - Berta Levavi-Sivan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot 76100, Israel.
| |
Collapse
|
27
|
Althumairy D, Zhang X, Baez N, Barisas G, Roess DA, Bousfield GR, Crans DC. Glycoprotein G-protein Coupled Receptors in Disease: Luteinizing Hormone Receptors and Follicle Stimulating Hormone Receptors. Diseases 2020; 8:E35. [PMID: 32942611 PMCID: PMC7565105 DOI: 10.3390/diseases8030035] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/22/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
Signal transduction by luteinizing hormone receptors (LHRs) and follicle-stimulating hormone receptors (FSHRs) is essential for the successful reproduction of human beings. Both receptors and the thyroid-stimulating hormone receptor are members of a subset of G-protein coupled receptors (GPCRs) described as the glycoprotein hormone receptors. Their ligands, follicle-stimulating hormone (FSH) and luteinizing hormone (LH) and a structurally related hormone produced in pregnancy, human chorionic gonadotropin (hCG), are large protein hormones that are extensively glycosylated. Although the primary physiologic functions of these receptors are in ovarian function and maintenance of pregnancy in human females and spermatogenesis in males, there are reports of LHRs or FSHRs involvement in disease processes both in the reproductive system and elsewhere. In this review, we evaluate the aggregation state of the structure of actively signaling LHRs or FSHRs, their functions in reproduction as well as summarizing disease processes related to receptor mutations affecting receptor function or expression in reproductive and non-reproductive tissues. We will also present novel strategies for either increasing or reducing the activity of LHRs signaling. Such approaches to modify signaling by glycoprotein receptors may prove advantageous in treating diseases relating to LHRs or FSHRs function in addition to furthering the identification of new strategies for modulating GPCR signaling.
Collapse
Affiliation(s)
- Duaa Althumairy
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, USA; (D.A.); (G.B.)
- Department of Biological Sciences, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Xiaoping Zhang
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; (X.Z.); (N.B.)
| | - Nicholas Baez
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; (X.Z.); (N.B.)
| | - George Barisas
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, USA; (D.A.); (G.B.)
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; (X.Z.); (N.B.)
| | - Deborah A. Roess
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA;
| | - George R. Bousfield
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260, USA;
| | - Debbie C. Crans
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, USA; (D.A.); (G.B.)
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA; (X.Z.); (N.B.)
| |
Collapse
|
28
|
Structural Complexity and Plasticity of Signaling Regulation at the Melanocortin-4 Receptor. Int J Mol Sci 2020; 21:ijms21165728. [PMID: 32785054 PMCID: PMC7460885 DOI: 10.3390/ijms21165728] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
The melanocortin-4 receptor (MC4R) is a class A G protein-coupled receptor (GPCR), essential for regulation of appetite and metabolism. Pathogenic inactivating MC4R mutations are the most frequent cause of monogenic obesity, a growing medical and socioeconomic problem worldwide. The MC4R mediates either ligand-independent or ligand-dependent signaling. Agonists such as α-melanocyte-stimulating hormone (α-MSH) induce anorexigenic effects, in contrast to the endogenous inverse agonist agouti-related peptide (AgRP), which causes orexigenic effects by suppressing high basal signaling activity. Agonist action triggers the binding of different subtypes of G proteins and arrestins, leading to concomitant induction of diverse intracellular signaling cascades. An increasing number of experimental studies have unraveled molecular properties and mechanisms of MC4R signal transduction related to physiological and pathophysiological aspects. In addition, the MC4R crystal structure was recently determined at 2.75 Å resolution in an inactive state bound with a peptide antagonist. Underpinned by structural homology models of MC4R complexes simulating a presumably active-state conformation compared to the structure of the inactive state, we here briefly summarize the current understanding and key players involved in the MC4R switching process between different activity states. Finally, these perspectives highlight the complexity and plasticity in MC4R signaling regulation and identify gaps in our current knowledge.
Collapse
|
29
|
Schulze A, Kleinau G, Neumann S, Scheerer P, Schöneberg T, Brüser A. The intramolecular agonist is obligate for activation of glycoprotein hormone receptors. FASEB J 2020; 34:11243-11256. [PMID: 32648604 DOI: 10.1096/fj.202000100r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 01/15/2023]
Abstract
In contrast to most rhodopsin-like G protein-coupled receptors, the glycoprotein hormone receptors (GPHR) have a large extracellular N-terminus for hormone binding. The hormones do not directly activate the transmembrane domain but mediate their action via a, thus, far only partially known Tethered Agonistic LIgand (TALI). The existence of such an intramolecular agonist was initially indicated by site-directed mutation studies and activating peptides derived from the extracellular hinge region. It is still unknown precisely how TALI is involved in intramolecular signal transmission. We combined systematic mutagenesis studies at the luteinizing hormone receptor and the thyroid-stimulating hormone receptor (TSHR), stimulation with a drug-like agonist (E2) of the TSHR, and structural homology modeling to unravel the functional and structural properties defining the TALI region. Here, we report that TALI (a) is predisposed to constitutively activate GPHR, (b) can by itself rearrange GPHR into a fully active conformation, (c) stabilizes active GPHR conformation, and (d) is not involved in activation of the TSHR by E2. In the active state conformation, TALI forms specific interactions between the N-terminus and the transmembrane domain. We show that stabilization of an active state is dependent on TALI, including activation by hormones and constitutively activating mutations.
Collapse
Affiliation(s)
- Annelie Schulze
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Gunnar Kleinau
- Group Protein X-ray Crystallography and Signal Transduction, Institute of Medical Physics and Biophysics, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Susanne Neumann
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patrick Scheerer
- Group Protein X-ray Crystallography and Signal Transduction, Institute of Medical Physics and Biophysics, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Antje Brüser
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| |
Collapse
|
30
|
Simoni M, Brigante G, Rochira V, Santi D, Casarini L. Prospects for FSH Treatment of Male Infertility. J Clin Endocrinol Metab 2020; 105:5831300. [PMID: 32374828 DOI: 10.1210/clinem/dgaa243] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022]
Abstract
CONTEXT Despite the new opportunities provided by assisted reproductive technology (ART), male infertility treatment is far from being optimized. One possibility, based on pathophysiological evidence, is to stimulate spermatogenesis with gonadotropins. EVIDENCE ACQUISITION We conducted a comprehensive systematic PubMed literature review, up to January 2020, of studies evaluating the genetic basis of follicle-stimulating hormone (FSH) action, the role of FSH in spermatogenesis, and the effects of its administration in male infertility. Manuscripts evaluating the role of genetic polymorphisms and FSH administration in women undergoing ART were considered whenever relevant. EVIDENCE SYNTHESIS FSH treatment has been successfully used in hypogonadotropic hypogonadism, but with questionable results in idiopathic male infertility. A limitation of this approach is that treatment plans for male infertility have been borrowed from hypogonadism, without daring to overstimulate, as is done in women undergoing ART. FSH effectiveness depends not only on its serum levels, but also on individual genetic variants able to determine hormonal levels, activity, and receptor response. Single-nucleotide polymorphisms in the follicle-stimulating hormone subunit beta (FSHB) and follicle-stimulating hormone receptor (FSHR) genes have been described, with some of them affecting testicular volume and sperm output. The FSHR p.N680S and the FSHB -211G>T variants could be genetic markers to predict FSH response. CONCLUSIONS FSH may be helpful to increase sperm production in infertile men, even if the evidence to recommend the use of FSH in this setting is weak. Placebo-controlled clinical trials, considering the FSHB-FSHR haplotype, are needed to define the most effective dosage, the best treatment length, and the criteria to select candidate responder patients.
Collapse
Affiliation(s)
- Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, 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, Nouzilly, France
| | - Giulia Brigante
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Vincenzo Rochira
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Daniele Santi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Livio Casarini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| |
Collapse
|
31
|
Saikia S, Bordoloi M, Sarmah R. Established and In-trial GPCR Families in Clinical Trials: A Review for Target Selection. Curr Drug Targets 2020; 20:522-539. [PMID: 30394207 DOI: 10.2174/1389450120666181105152439] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/28/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022]
Abstract
The largest family of drug targets in clinical trials constitute of GPCRs (G-protein coupled receptors) which accounts for about 34% of FDA (Food and Drug Administration) approved drugs acting on 108 unique GPCRs. Factors such as readily identifiable conserved motif in structures, 127 orphan GPCRs despite various de-orphaning techniques, directed functional antibodies for validation as drug targets, etc. has widened their therapeutic windows. The availability of 44 crystal structures of unique receptors, unexplored non-olfactory GPCRs (encoded by 50% of the human genome) and 205 ligand receptor complexes now present a strong foundation for structure-based drug discovery and design. The growing impact of polypharmacology for complex diseases like schizophrenia, cancer etc. warrants the need for novel targets and considering the undiscriminating and selectivity of GPCRs, they can fulfill this purpose. Again, natural genetic variations within the human genome sometimes delude the therapeutic expectations of some drugs, resulting in medication response differences and ADRs (adverse drug reactions). Around ~30 billion US dollars are dumped annually for poor accounting of ADRs in the US alone. To curb such undesirable reactions, the knowledge of established and currently in clinical trials GPCRs families can offer huge understanding towards the drug designing prospects including "off-target" effects reducing economical resource and time. The druggability of GPCR protein families and critical roles played by them in complex diseases are explained. Class A, class B1, class C and class F are generally established family and GPCRs in phase I (19%), phase II(29%), phase III(52%) studies are also reviewed. From the phase I studies, frizzled receptors accounted for the highest in trial targets, neuropeptides in phase II and melanocortin in phase III studies. Also, the bioapplications for nanoparticles along with future prospects for both nanomedicine and GPCR drug industry are discussed. Further, the use of computational techniques and methods employed for different target validations are also reviewed along with their future potential for the GPCR based drug discovery.
Collapse
Affiliation(s)
- Surovi Saikia
- Natural Products Chemistry Group, CSIR North East Institute of Science & Technology, Jorhat-785006, Assam, India
| | - Manobjyoti Bordoloi
- Natural Products Chemistry Group, CSIR North East Institute of Science & Technology, Jorhat-785006, Assam, India
| | - Rajeev Sarmah
- Allied Health Sciences, Assam Down Town University, Panikhaiti, Guwahati 781026, Assam, India
| |
Collapse
|
32
|
Xu W, Cao JF, Zhang YY, Shu Y, Wang JH. Boronic acid modified polyoxometalate-alginate hybrid for the isolation of glycoproteins at neutral environment. Talanta 2020; 210:120620. [DOI: 10.1016/j.talanta.2019.120620] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 12/01/2019] [Accepted: 12/06/2019] [Indexed: 01/18/2023]
|
33
|
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: 8.8] [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
|
34
|
Hollander-Cohen L, Böhm B, Hausken K, Levavi-Sivan B. Ontogeny of the specificity of gonadotropin receptors and gene expression in carp. Endocr Connect 2019; 8:1433-1446. [PMID: 31581128 PMCID: PMC6826172 DOI: 10.1530/ec-19-0389] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 10/02/2019] [Indexed: 02/02/2023]
Abstract
The pituitary gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), are the principle endocrine drivers of reproductive processes in the gonads of jawed vertebrates. Canonically, FSH recruits and maintains selected ovarian follicles for maturation and LH induces the stages of germinal vesicle breakdown and ovulation. In mammals, LH and FSH specifically activate cognate G-protein-coupled receptors that affect the proteins involved in steroidogenesis, protein hormone synthesis, and gametogenesis. This dual-gonadotropin model also exists in some fish species, but not in all. In fact, due to their diverse number of species, extended number of ecological niches, and remarkably flexible reproductive strategies, fish are appropriate as models to understand the co-evolution of gonadotropins and their receptors. In this study, we cloned and characterized the expression profile over the final stages of ovarian maturation of carp (Cyprinus carpio) LHCGR and FSHR. Expression of both gonadotropin receptors increased in the later stage of early vitellogenesis, suggesting that both LH and FSH play a role in the development of mature follicles. We additionally tested the activation of cLHCGR and cFSHR using homologous and heterologous recombinant gonadotropins in order to gain insight into an evolutionary model of permissive gonadotropin receptor function. These data suggest that carp (Cyprinus carpio) gonad development and maturation depends on a specific gonadotropin profile that does not reflect the temporally distinct dual-gonadotropin model observed in salmonids or mammals, and that permissive gonadotropin receptor activation is a specific feature of Ostariophysi, not all teleosts.
Collapse
Affiliation(s)
- Lian Hollander-Cohen
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Benjamin Böhm
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Krist Hausken
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Berta Levavi-Sivan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University of Jerusalem, Rehovot, Israel
- Correspondence should be addressed to B Levavi-Sivan:
| |
Collapse
|
35
|
Hausken K, Levavi-Sivan B. Synteny and phylogenetic analysis of paralogous thyrostimulin beta subunits (GpB5) in vertebrates. PLoS One 2019; 14:e0222808. [PMID: 31536580 PMCID: PMC6752823 DOI: 10.1371/journal.pone.0222808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/06/2019] [Indexed: 01/02/2023] Open
Abstract
At some point early in the vertebrate lineage, two whole genome duplication events (1R, 2R) took place that allowed for the diversification and sub-/neo-functionalization of the glycoprotein hormones (GpHs). All jawed vertebrates possess the GpHs luteinizing hormone (LH), follicle stimulating hormone (FSH), and thyroid stimulating hormone (TSH), each of which are heterodimers with a common alpha subunit and unique beta subunits. In 2002, a novel glycoprotein hormone named thyrostimulin was described to have unique GpA2 and GpB5 subunits that were homologous to the vertebrate alpha and beta subunits. The presence of GpA2 and GpB5 in representative protostomes and deuterostomes indicates their ancestry in the GpH family. There are several reports of GpH subunit evolution, but none have included GpA2 and GpB5 for species in each major vertebrate class. Thus, we addressed the ancestry of two paralogous GpB5 subunits (GpB5a and GpB5b) that were previously only recognized in two teleost species. Our search for orthologous GpB5a and GpB5b sequences in representative vertebrates and phylogenetic analysis, in addition to the currently published evolutionary scenarios of the GpH family, supports that GpB5a and GpB5b are paralogs that arose from the first vertebrate whole genome duplication event (1R). Syntenic analysis supports lineage specific losses of GpB5a in chondrichthyes, basal actinopterygians, and tetrapods, and retention in coelacanth and teleosts. Additionally, we were unable to identify GpA2 transcripts from tilapia mRNA, suggesting that this species does not produce heterodimeric thyrostimulin. While the conserved or even species-specific functional role of thyrostimulin or its individual subunits are still unknown in vertebrates, the analyses presented here provide context for future studies on the functional divergence of the GpH family.
Collapse
Affiliation(s)
- Krist Hausken
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Berta Levavi-Sivan
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
36
|
Marcinkowski P, Kreuchwig A, Mendieta S, Hoyer I, Witte F, Furkert J, Rutz C, Lentz D, Krause G, Schülein R. Thyrotropin Receptor: Allosteric Modulators Illuminate Intramolecular Signaling Mechanisms at the Interface of Ecto- and Transmembrane Domain. Mol Pharmacol 2019; 96:452-462. [DOI: 10.1124/mol.119.116947] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/23/2019] [Indexed: 11/22/2022] Open
|
37
|
Lizneva D, Rahimova A, Kim SM, Atabiekov I, Javaid S, Alamoush B, Taneja C, Khan A, Sun L, Azziz R, Yuen T, Zaidi M. FSH Beyond Fertility. Front Endocrinol (Lausanne) 2019; 10:136. [PMID: 30941099 PMCID: PMC6433784 DOI: 10.3389/fendo.2019.00136] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/13/2019] [Indexed: 12/24/2022] Open
Abstract
The traditional view of follicle-stimulating hormone (FSH) as a reproductive hormone is changing. It has been shown that FSH receptors (FSHRs) are expressed in various extra-gonadal tissues and mediate the biological effects of FSH at those sites. Molecular, animal, epidemiologic, and clinical data suggest that elevated serum FSH may play a significant role in the evolution of bone loss and obesity, as well as contributing to cardiovascular and cancer risk. This review summarizes recent data on FSH action beyond reproduction.
Collapse
Affiliation(s)
- Daria Lizneva
- The Mount Sinai Bone Program, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alina Rahimova
- The Mount Sinai Bone Program, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Se-Min Kim
- The Mount Sinai Bone Program, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ihor Atabiekov
- The Mount Sinai Bone Program, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Seher Javaid
- The Mount Sinai Bone Program, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Bateel Alamoush
- The Mount Sinai Bone Program, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Charit Taneja
- The Mount Sinai Bone Program, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ayesha Khan
- The Mount Sinai Bone Program, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Li Sun
- The Mount Sinai Bone Program, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ricardo Azziz
- Academic Health and Hospital Affairs, State University of New York, Albany, NY, United States
| | - Tony Yuen
- The Mount Sinai Bone Program, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mone Zaidi
- The Mount Sinai Bone Program, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
38
|
Marcinkowski P, Hoyer I, Specker E, Furkert J, Rutz C, Neuenschwander M, Sobottka S, Sun H, Nazare M, Berchner-Pfannschmidt U, von Kries JP, Eckstein A, Schülein R, Krause G. A New Highly Thyrotropin Receptor-Selective Small-Molecule Antagonist with Potential for the Treatment of Graves' Orbitopathy. Thyroid 2019; 29:111-123. [PMID: 30351237 DOI: 10.1089/thy.2018.0349] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND The thyrotropin receptor (TSHR) is the target for autoimmune thyroid stimulating antibodies (TSAb) triggering hyperthyroidism. Whereas elevated thyroid hormone synthesis by the thyroid in Graves' disease can be treated by antithyroid agents, for the pathogenic activation of TSHR in retro-orbital fibroblasts of the eye, leading to Graves' orbitopathy (GO), no causal TSHR directed therapy is available. METHODS Due to the therapeutic gap for severe GO, TSHR inhibitors were identified by high-throughput screening in Chinese hamster ovary cells expressing the TSHR. Stereo-selective synthesis of the screening hits led to the molecule S37, which contains seven chiral centers. Enantiomeric separation of the molecule S37 resulted in the enantiopure molecule S37a-a micro-molar antagonist of thyrotropin-induced cyclic adenosine monophosphate accumulation in HEK 293 cells expressing the TSHR. RESULTS The unique rigid bent shape of molecule S37a may mediate the observed high TSHR selectivity. Most importantly, the closely related follitropin and lutropin receptors were not affected by this compound. S37a not only inhibits the TSHR activation by thyrotropin itself but also activation by monoclonal TSAb M22 (human), KSAb1 (murine), and the allosteric small-molecule agonist C2. Disease-related ex vivo studies in HEK 293 cells expressing the TSHR showed that S37a also inhibits cyclic adenosine monophosphate formation by oligoclonal TSAb, which are highly enriched in GO patients' sera. Initial in vivo pharmacokinetic studies revealed no toxicity of S37a and a remarkable 53% oral bioavailability in mice. CONCLUSION In summary, a novel highly selective inhibitor for the TSHR is presented, which has promising potential for further development for the treatment of GO.
Collapse
Affiliation(s)
| | - Inna Hoyer
- 1 Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Edgar Specker
- 1 Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Jens Furkert
- 1 Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Claudia Rutz
- 1 Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | | | - Sebastian Sobottka
- 1 Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Han Sun
- 1 Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Marc Nazare
- 1 Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | | | | | - Anja Eckstein
- 2 Department of Ophthalmology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ralf Schülein
- 1 Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Gerd Krause
- 1 Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| |
Collapse
|
39
|
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: 5.0] [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
|
40
|
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
|
41
|
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: 2.2] [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
|
42
|
Ulloa-Aguirre A, Zariñán T, Jardón-Valadez E, Gutiérrez-Sagal R, Dias JA. Structure-Function Relationships of the Follicle-Stimulating Hormone Receptor. Front Endocrinol (Lausanne) 2018; 9:707. [PMID: 30555414 PMCID: PMC6281744 DOI: 10.3389/fendo.2018.00707] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/09/2018] [Indexed: 12/16/2022] Open
Abstract
The follicle-stimulating hormone receptor (FSHR) plays a crucial role in reproduction. This structurally complex receptor is a member of the G-protein coupled receptor (GPCR) superfamily of membrane receptors. As with the other structurally similar glycoprotein hormone receptors (the thyroid-stimulating hormone and luteinizing hormone-chorionic gonadotropin hormone receptors), the FSHR is characterized by an extensive extracellular domain, where binding to FSH occurs, linked to the signal specificity subdomain or hinge region. This region is involved in ligand-stimulated receptor activation whereas the seven transmembrane domain is associated with receptor activation and transmission of the activation process to the intracellular loops comprised of amino acid sequences, which predicate coupling to effectors, interaction with adapter proteins, and triggering of downstream intracellular signaling. In this review, we describe the most important structural features of the FSHR intimately involved in regulation of FSHR function, including trafficking, dimerization, and oligomerization, ligand binding, agonist-stimulated activation, and signal transduction.
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
- *Correspondence: Alfredo Ulloa-Aguirre
| | - 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
| | - Eduardo Jardón-Valadez
- Departamento de Ciencias Ambientales, Universidad Autónoma Metropolitana Unidad Lerma, Lerma, Mexico
| | - Rubén Gutiérrez-Sagal
- 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, School of Public Health, University at Albany, Albany, NY, United States
| |
Collapse
|
43
|
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: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Kleinau G, Worth CL, Kreuchwig A, Biebermann H, Marcinkowski P, Scheerer P, Krause G. Structural-Functional Features of the Thyrotropin Receptor: A Class A G-Protein-Coupled Receptor at Work. Front Endocrinol (Lausanne) 2017; 8:86. [PMID: 28484426 PMCID: PMC5401882 DOI: 10.3389/fendo.2017.00086] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/03/2017] [Indexed: 12/21/2022] Open
Abstract
The thyroid-stimulating hormone receptor (TSHR) is a member of the glycoprotein hormone receptors, a sub-group of class A G-protein-coupled receptors (GPCRs). TSHR and its endogenous ligand thyrotropin (TSH) are of essential importance for growth and function of the thyroid gland and proper function of the TSH/TSHR system is pivotal for production and release of thyroid hormones. This receptor is also important with respect to pathophysiology, such as autoimmune (including ophthalmopathy) or non-autoimmune thyroid dysfunctions and cancer development. Pharmacological interventions directly targeting the TSHR should provide benefits to disease treatment compared to currently available therapies of dysfunctions associated with the TSHR or the thyroid gland. Upon TSHR activation, the molecular events conveying conformational changes from the extra- to the intracellular side of the cell across the membrane comprise reception, conversion, and amplification of the signal. These steps are highly dependent on structural features of this receptor and its intermolecular interaction partners, e.g., TSH, antibodies, small molecules, G-proteins, or arrestin. For better understanding of signal transduction, pathogenic mechanisms such as autoantibody action and mutational modifications or for developing new pharmacological strategies, it is essential to combine available structural data with functional information to generate homology models of the entire receptor. Although so far these insights are fragmental, in the past few decades essential contributions have been made to investigate in-depth the involved determinants, such as by structure determination via X-ray crystallography. This review summarizes available knowledge (as of December 2016) concerning the TSHR protein structure, associated functional aspects, and based on these insights we suggest several receptor complex models. Moreover, distinct TSHR properties will be highlighted in comparison to other class A GPCRs to understand the molecular activation mechanisms of this receptor comprehensively. Finally, limitations of current knowledge and lack of information are discussed highlighting the need for intensified efforts toward TSHR structure elucidation.
Collapse
Affiliation(s)
- Gunnar Kleinau
- Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin, Berlin, Germany
- Group Protein X-Ray Crystallography and Signal Transduction, Institute of Medical Physics and Biophysics, Charité-Universitätsmedizin, Berlin, Germany
| | | | - Annika Kreuchwig
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Heike Biebermann
- Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin, Berlin, Germany
| | | | - Patrick Scheerer
- Group Protein X-Ray Crystallography and Signal Transduction, Institute of Medical Physics and Biophysics, Charité-Universitätsmedizin, Berlin, Germany
| | - Gerd Krause
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Berlin, Germany
- *Correspondence: Gerd Krause,
| |
Collapse
|
45
|
|
46
|
Kleinau G, Kalveram L, Köhrle J, Szkudlinski M, Schomburg L, Biebermann H, Grüters-Kieslich A. Minireview: Insights Into the Structural and Molecular Consequences of the TSH-β Mutation C105Vfs114X. Mol Endocrinol 2016; 30:954-64. [PMID: 27387040 DOI: 10.1210/me.2016-1065] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Naturally occurring thyrotropin (TSH) mutations are rare, which is also the case for the homologous heterodimeric glycoprotein hormones (GPHs) follitropin (FSH), lutropin (LH), and choriogonadotropin (CG). Patients with TSH-inactivating mutations present with central congenital hypothyroidism. Here, we summarize insights into the most frequent loss-of-function β-subunit of TSH mutation C105Vfs114X, which is associated with isolated TSH deficiency. This review will address the following question. What is currently known on the molecular background of this TSH variant on a protein level? It has not yet been clarified how C105Vfs114X causes early symptoms in affected patients, which are comparably severe to those observed in newborns lacking any functional thyroid tissue (athyreosis). To better understand the mechanisms of this mutant, we have summarized published reports and complemented this information with a structural perspective on GPHs. By including the ancestral TSH receptor agonist thyrostimulin and pathogenic mutations reported for FSH, LH, and choriogonadotropin in the analysis, insightful structure function and evolutionary restrictions become apparent. However, comparisons of immunogenicity and bioactivity of different GPH variants is hindered by a lack of consensus for functional analysis and the diversity of used GPH assays. Accordingly, relevant gaps of knowledge concerning details of GPH mutation-related effects are identified and highlighted in this review. These issues are of general importance as several previous and recent studies point towards the high impact of GPH variants in differential signaling regulation at GPH receptors (GPHRs), both endogenously and under diseased conditions. Further improvement in this area is of decisive importance for the development of novel targeted therapies.
Collapse
Affiliation(s)
- Gunnar Kleinau
- Institute of Experimental Pediatric Endocrinology (G.K., L.K., H.B.), Charité-Universitätsmedizin, Berlin, 13353 Germany; Institute of Experimental Endocrinology (J.K., L.S.), Charité-Universitätsmedizin Berlin, 13353 Germany; Trophogen, Inc (M.S.), Rockville, Maryland 20850; and Department of Pediatric Endocrinology and Diabetes (A.G.-K.), Charité-Universitätsmedizin, Berlin, 13353 Germany
| | - Laura Kalveram
- Institute of Experimental Pediatric Endocrinology (G.K., L.K., H.B.), Charité-Universitätsmedizin, Berlin, 13353 Germany; Institute of Experimental Endocrinology (J.K., L.S.), Charité-Universitätsmedizin Berlin, 13353 Germany; Trophogen, Inc (M.S.), Rockville, Maryland 20850; and Department of Pediatric Endocrinology and Diabetes (A.G.-K.), Charité-Universitätsmedizin, Berlin, 13353 Germany
| | - Josef Köhrle
- Institute of Experimental Pediatric Endocrinology (G.K., L.K., H.B.), Charité-Universitätsmedizin, Berlin, 13353 Germany; Institute of Experimental Endocrinology (J.K., L.S.), Charité-Universitätsmedizin Berlin, 13353 Germany; Trophogen, Inc (M.S.), Rockville, Maryland 20850; and Department of Pediatric Endocrinology and Diabetes (A.G.-K.), Charité-Universitätsmedizin, Berlin, 13353 Germany
| | - Mariusz Szkudlinski
- Institute of Experimental Pediatric Endocrinology (G.K., L.K., H.B.), Charité-Universitätsmedizin, Berlin, 13353 Germany; Institute of Experimental Endocrinology (J.K., L.S.), Charité-Universitätsmedizin Berlin, 13353 Germany; Trophogen, Inc (M.S.), Rockville, Maryland 20850; and Department of Pediatric Endocrinology and Diabetes (A.G.-K.), Charité-Universitätsmedizin, Berlin, 13353 Germany
| | - Lutz Schomburg
- Institute of Experimental Pediatric Endocrinology (G.K., L.K., H.B.), Charité-Universitätsmedizin, Berlin, 13353 Germany; Institute of Experimental Endocrinology (J.K., L.S.), Charité-Universitätsmedizin Berlin, 13353 Germany; Trophogen, Inc (M.S.), Rockville, Maryland 20850; and Department of Pediatric Endocrinology and Diabetes (A.G.-K.), Charité-Universitätsmedizin, Berlin, 13353 Germany
| | - Heike Biebermann
- Institute of Experimental Pediatric Endocrinology (G.K., L.K., H.B.), Charité-Universitätsmedizin, Berlin, 13353 Germany; Institute of Experimental Endocrinology (J.K., L.S.), Charité-Universitätsmedizin Berlin, 13353 Germany; Trophogen, Inc (M.S.), Rockville, Maryland 20850; and Department of Pediatric Endocrinology and Diabetes (A.G.-K.), Charité-Universitätsmedizin, Berlin, 13353 Germany
| | - Annette Grüters-Kieslich
- Institute of Experimental Pediatric Endocrinology (G.K., L.K., H.B.), Charité-Universitätsmedizin, Berlin, 13353 Germany; Institute of Experimental Endocrinology (J.K., L.S.), Charité-Universitätsmedizin Berlin, 13353 Germany; Trophogen, Inc (M.S.), Rockville, Maryland 20850; and Department of Pediatric Endocrinology and Diabetes (A.G.-K.), Charité-Universitätsmedizin, Berlin, 13353 Germany
| |
Collapse
|
47
|
Ulloa-Aguirre A, Zariñán T. The Follitropin Receptor: Matching Structure and Function. Mol Pharmacol 2016; 90:596-608. [DOI: 10.1124/mol.116.104398] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/28/2016] [Indexed: 12/19/2022] Open
|
48
|
Schöneberg T, Kleinau G, Brüser A. What are they waiting for?—Tethered agonism in G protein-coupled receptors. Pharmacol Res 2016; 108:9-15. [DOI: 10.1016/j.phrs.2016.03.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 03/24/2016] [Indexed: 01/02/2023]
|
49
|
Dhar N, Mohan A, Thakur C, Chandra NR, Dighe RR. Dissecting the structural and functional features of the Luteinizing hormone receptor using receptor specific single chain fragment variables. Mol Cell Endocrinol 2016; 427:1-12. [PMID: 26940038 DOI: 10.1016/j.mce.2016.02.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 02/27/2016] [Accepted: 02/27/2016] [Indexed: 01/16/2023]
Abstract
The Luteinizing hormone receptor (LHR) has a large extracellular domain (amino acid residues, a.a.1-355) and a transmembrane domain (TMD; a.a. 356-699), essential for hormone binding and signaling, respectively. The LHR hinge region (a.a. 256-355) connects the two domains and acts as an activating switch for the receptor by an unknown mechanism. LHR hinge-specific Single chain fragment variables (ScFv) stimulated cAMP production by the stable and transiently transfected cell lines expressing LHR in a hormone-independent manner and the C-terminal region of LHR hinge (a.a. 313-349) was identified as the probable epitope for one agonistic ScFv. This epitope attained a helical conformation upon agonistic ScFv binding and the activity of the ScFv was dependent on Y331 sulfation. ScFv was also able to activate TMD mutants, D578Y and A593P, reemphasizing the role of TM helix VI in LHR activation.
Collapse
Affiliation(s)
- Neha Dhar
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Abhilash Mohan
- Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Chandrani Thakur
- Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Nagasuma R Chandra
- Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Rajan R Dighe
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, Karnataka 560012, India.
| |
Collapse
|
50
|
Inaba H, De Groot LJ, Akamizu T. Thyrotropin Receptor Epitope and Human Leukocyte Antigen in Graves' Disease. Front Endocrinol (Lausanne) 2016; 7:120. [PMID: 27602020 PMCID: PMC4994058 DOI: 10.3389/fendo.2016.00120] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/12/2016] [Indexed: 11/13/2022] Open
Abstract
Graves' disease (GD) is an organ-specific autoimmune disease, and thyrotropin (TSH) receptor (TSHR) is a major autoantigen in this condition. Since the extracellular domain of human TSHR (TSHR-ECD) is shed into the circulation, TSHR-ECD is a preferentially immunogenic portion of TSHR. Both genetic factors and environmental factors contribute to development of GD. Inheritance of human leukocyte antigen (HLA) genes, especially HLA-DR3, is associated with GD. TSHR-ECD protein is endocytosed into antigen-presenting cells (APCs), and processed to TSHR-ECD peptides. These peptide epitopes bind to HLA-class II molecules, and subsequently the complex of HLA-class II and TSHR-ECD epitope is presented to CD4+ T cells. The activated CD4+ T cells secrete cytokines/chemokines that stimulate B-cells to produce TSAb, and in turn hyperthyroidism occurs. Numerous studies have been done to identify T- and B-cell epitopes in TSHR-ECD, including (1) in silico, (2) in vitro, (3) in vivo, and (4) clinical experiments. Murine models of GD and HLA-transgenic mice have played a pivotal role in elucidating the immunological mechanisms. To date, linear or conformational epitopes of TSHR-ECD, as well as the molecular structure of the epitope-binding groove in HLA-DR, were reported to be related to the pathogenesis in GD. Dysfunction of central tolerance in the thymus, or in peripheral tolerance, such as regulatory T cells, could allow development of GD. Novel treatments using TSHR antagonists or mutated TSHR peptides have been reported to be effective. We review and update the role of immunogenic TSHR epitopes and HLA in GD, and offer perspectives on TSHR epitope specific treatments.
Collapse
Affiliation(s)
- Hidefumi Inaba
- The First Department of Medicine, Wakayama Medical University, Wakayama, Japan
- *Correspondence: Hidefumi Inaba,
| | - Leslie J. De Groot
- Department of Cellular and Molecular Biology, University of Rhode Island, Providence, RI, USA
| | - Takashi Akamizu
- The First Department of Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|